1
|
Goyal A, Cabrera JR, Blaha CD, Lee KH, Shin H, Oh Y. Ventral tegmental area deep brain stimulation reverses ethanol-induced dopamine increase in the rat nucleus accumbens. Biomed Eng Lett 2024; 14:1347-1354. [PMID: 39465114 PMCID: PMC11502691 DOI: 10.1007/s13534-024-00408-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 06/09/2024] [Accepted: 06/30/2024] [Indexed: 10/29/2024] Open
Abstract
The neurophysiology of alcohol use disorder (AUD) is complex, but a major contributor to addictive phenotypes is the tendency for drugs of abuse to increase tonic extracellular dopamine (DA) levels in the nucleus accumbens (NAc). Repeated exposure to substances of abuse such as ethanol results in the overstimulation of the mesolimbic pathway, causing an excessive release of DA from the ventral tegmental area (VTA) to target regions such as the NAc. This heightened DA signaling is associated with the reinforcing effects of substances, leading to a strong desire for continued use. Recent work has postulated that high frequency deep brain stimulation (DBS) of the ventral tegmental area may reduce dopamine transmission to the nucleus accumbens following acute drug of abuse exposure, thereby mitigating the drug's addictive potential. We first demonstrate ethanol's ability to decrease phasic DA release over time and to increase tonic extracellular DA concentrations in the nucleus accumbens. Next, we demonstrate the capability for high frequency VTA DBS to reverse this ethanol-associated surge in tonic DA concentrations in the nucleus accumbens to levels not significantly different from baseline. This study suggests a promising new avenue for investigating the mechanisms of alcohol use disorder.
Collapse
Affiliation(s)
- Abhinav Goyal
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic, Rochester, MN 55905 USA
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905 USA
| | - Juan Rojas Cabrera
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic, Rochester, MN 55905 USA
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905 USA
| | - Charles D. Blaha
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905 USA
| | - Kendall H. Lee
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905 USA
- Department of Biomedical Engineering, Mayo Clinic, Rochester, MN 55905 USA
| | - Hojin Shin
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905 USA
- Department of Biomedical Engineering, Mayo Clinic, Rochester, MN 55905 USA
| | - Yoonbae Oh
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905 USA
- Department of Biomedical Engineering, Mayo Clinic, Rochester, MN 55905 USA
| |
Collapse
|
2
|
Wang J, Qu S, Li R, Tang S, Li H. Blunted sensitivity to expected value during risky decision making in individuals with problematic pornography use. J Behav Addict 2024; 13:779-790. [PMID: 39141431 PMCID: PMC11457035 DOI: 10.1556/2006.2024.00043] [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: 05/04/2024] [Revised: 06/20/2024] [Accepted: 07/05/2024] [Indexed: 08/16/2024] Open
Abstract
Background and aims Neurobiological models of addiction posit that addiction manifests through an amplified salience towards addiction-associated stimuli and a diminished responsiveness to non-addiction-related incentives. However, existing research on reward processing in individuals with problematic pornography use (PPU) has primarily been limited to sexual cue reactivity. Methods In this event-related potential (ERP) study, we employed a risky decision-making task involving 30 individuals with PPU and 33 healthy controls (HCs) to examine the effects of PPU on non-pornographic (money) reward valuation. Results Compared to HCs, individuals with PPU exhibited compromised sensitivity to monetary rewards. Specifically, while the HC group demonstrated a differential response in late positive potential (LPP) amplitude to various expected value (EV) levels, this pattern was absent in the PPU group. This impairment was associated with poorer adaptive decision-making, as evidenced by PPU participants' inability to adjust risk choices based on changes in EV, leading to a propensity for riskier decisions in disadvantageous situations. Discussion and conclusions The findings of impaired monetary evaluation in individuals with PPU may potentially explain why they continually pursue pornographic rewards while showing insensitivity to other rewards in daily life. Consequently, treatment development strategies may prioritize improving sensitivity to non-pornographic rewards within this population.
Collapse
Affiliation(s)
- Jianfeng Wang
- Institute of Brain and Psychological Sciences, Sichuan Normal University, Chengdu, 610066, China
- School of Psychology, Chengdu Medical College, Chengdu, 610500, China
| | - Shuangyi Qu
- School of Psychology, Chengdu Medical College, Chengdu, 610500, China
| | - Ruiyu Li
- School of Psychology, Chengdu Medical College, Chengdu, 610500, China
| | - Shaoyue Tang
- School of Psychology, Chengdu Medical College, Chengdu, 610500, China
| | - Hong Li
- Institute of Brain and Psychological Sciences, Sichuan Normal University, Chengdu, 610066, China
| |
Collapse
|
3
|
Goyal A, Karanovic U, Blaha CD, Lee KH, Shin H, Oh Y. Toward Precise Modeling of Dopamine Release Kinetics: Comparison and Validation of Kinetic Models Using Voltammetry. ACS OMEGA 2024; 9:33563-33573. [PMID: 39130585 PMCID: PMC11307285 DOI: 10.1021/acsomega.4c01322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 07/12/2024] [Accepted: 07/18/2024] [Indexed: 08/13/2024]
Abstract
Dopamine (DA) is a neurotransmitter present within the animal brain that is responsible for a wide range of physiologic functions, including motivation, reward, and movement control. Changes or dysfunction in the dynamics of DA release are thought to play a pivotal role in regulating various physiological and behavioral processes, as well as leading to neuropsychiatric diseases. Therefore, it is of fundamental interest to neuroscientists to understand and accurately model the kinetics that govern dopaminergic neurotransmission. In the past several decades, many mathematical models have been proposed to attempt to capture the biologic parameters that govern dopaminergic kinetics, with each model seeking to improve upon a previous model. In this review, each of these models are derived, and the ability of each model to properly fit two fast-scan cyclic voltammetry (FSCV) data sets will be demonstrated and discussed. The dopamine oxidation current in both FSCV data sets exhibits hang-up and overshoot behaviors, which have traditionally been difficult for mathematical models to capture. We show that more recent models are better able to model DA release that exhibits these behaviors but that no single model is clearly the best. Rather, models should be selected based on their mathematical properties to best fit the FSCV data one is trying to model. Developing such differential equation models to describe the kinetics of DA release from the synapse confers significant applications both for advancing scientific understanding of DA neurotransmission and for advancing clinical ability to treat neuropsychiatric diseases.
Collapse
Affiliation(s)
- Abhinav Goyal
- Mayo
Clinic Medical Scientist Training Program, Mayo Clinic, Rochester, Minnesota 55905, United States
- Department
of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Una Karanovic
- Department
of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota 55905, United States
- Mayo
Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Charles D. Blaha
- Department
of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Kendall H. Lee
- Department
of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota 55905, United States
- Department
of Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Hojin Shin
- Department
of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota 55905, United States
- Department
of Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Yoonbae Oh
- Department
of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota 55905, United States
- Department
of Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
| |
Collapse
|
4
|
Schumacher R, Rossetti MF, Canesini G, Gaydou L, Garcia AP, Lazzarino GP, Fernandez PR, Stoker C, Carrió MJ, Andreoli MF, Ramos JG. Neonatal overfeeding alters the functioning of the mesolimbic dopaminergic circuitry involving changes in DNA methylation and effects on feeding behavior. J Nutr Biochem 2023; 122:109451. [PMID: 37748623 DOI: 10.1016/j.jnutbio.2023.109451] [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: 02/06/2023] [Revised: 08/23/2023] [Accepted: 09/21/2023] [Indexed: 09/27/2023]
Abstract
Mesolimbic dopaminergic circuit is essential for food reward and motivational behaviors and can contribute to weight gain and obesity. Litter reduction is a classical model for studying the effects of neonatal overfeeding and overweight. Litters of Wistar rats were reduced to 4 pups/dam for small litter (SL) and 10 pups/dam for normal litter at postnatal day (PND) 4. Immediately after performing the feeding behavior tests, the animals were sacrificed in PND21 and PND90. The ventral tegmental area (VTA), Nucleus Accumbens Core (NAcC) and Shell (NAcSh) were isolated from frozen brain sections using the Palkovits micropunch technique. RNA and DNA were extracted from these areas, gene expression was measured by RT-qPCR and DNA methylation levels were measured by MSRM-qPCR technique. SL-PND21 animals presented increased expression levels of Tyrosine Hydroxylase and Dopamine Receptor D2 in VTA, decreased expression levels of dopamine active transporter (DAT) in VTA, and higher expression levels of DAT in NAcC. On the other hand, SL-PND90 animals showed decreased expression levels of Dopamine Receptor D1 and higher expression of DAT in NAcSh. These animals also evidenced impaired sensory-specific satiety. In addition, altered promoter methylation was observed at weaning, and remained in adulthood. This work demonstrates that neonatal overfeeding induces disruptions in the mesolimbic dopaminergic circuitry and causes alterations in feeding behavior from weaning to adulthood, suggesting that the neonatal period is critical for the normal development of dopaminergic circuit that impact on feeding behavior.
Collapse
Affiliation(s)
- Rocio Schumacher
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa-CONICET, Santa Fe, Argentina
| | - Maria Florencia Rossetti
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa-CONICET, Santa Fe, Argentina
| | - Guillermina Canesini
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa-CONICET, Santa Fe, Argentina
| | - Luisa Gaydou
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa-CONICET, Santa Fe, Argentina; Departamento de Bioquímica Clínica y Cuantitativa, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Ana Paula Garcia
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa-CONICET, Santa Fe, Argentina
| | - Gisela Paola Lazzarino
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa-CONICET, Santa Fe, Argentina
| | - Pamela Rocio Fernandez
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa-CONICET, Santa Fe, Argentina
| | - Cora Stoker
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa-CONICET, Santa Fe, Argentina; Departamento de Bioquímica Clínica y Cuantitativa, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Maria Josefina Carrió
- Departamento de Matemática y Laboratorio de Investigaciones y Servicios en Bioestadística (LISEB), FBCB-UNL, Santa Fe, Argentina
| | - Maria Florencia Andreoli
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa-CONICET, Santa Fe, Argentina; Departamento de Bioquímica Clínica y Cuantitativa, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Jorge Guillermo Ramos
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa-CONICET, Santa Fe, Argentina; Departamento de Bioquímica Clínica y Cuantitativa, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina.
| |
Collapse
|
5
|
Zald DH. The influence of dopamine autoreceptors on temperament and addiction risk. Neurosci Biobehav Rev 2023; 155:105456. [PMID: 37926241 PMCID: PMC11330662 DOI: 10.1016/j.neubiorev.2023.105456] [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: 07/31/2023] [Revised: 10/22/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
As a major regulator of dopamine (DA), DA autoreceptors (DAARs) exert substantial influence over DA-mediated behaviors. This paper reviews the physiological and behavioral impact of DAARs. Individual differences in DAAR functioning influences temperamental traits such as novelty responsivity and impulsivity, both of which are associated with vulnerability to addictive behavior in animal models and a broad array of externalizing behaviors in humans. DAARs additionally impact the response to psychostimulants and other drugs of abuse. Human PET studies of D2-like receptors in the midbrain provide evidence for parallels to the animal literature. These data lead to the proposal that weak DAAR regulation is a risk factor for addiction and externalizing problems. The review highlights the potential to build translational models of the functional role of DAARs in behavior. It also draws attention to key limitations in the current literature that would need to be addressed to further advance a weak DAAR regulation model of addiction and externalizing risk.
Collapse
Affiliation(s)
- David H Zald
- Center for Advanced Human Brain Imaging and Department of Psychiatry, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
6
|
Chong TTJ, Fortunato E, Bellgrove MA. Amphetamines Improve the Motivation to Invest Effort in Attention-Deficit/Hyperactivity Disorder. J Neurosci 2023; 43:6898-6908. [PMID: 37666665 PMCID: PMC10573750 DOI: 10.1523/jneurosci.0982-23.2023] [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] [Received: 05/28/2023] [Revised: 07/26/2023] [Accepted: 07/31/2023] [Indexed: 09/06/2023] Open
Abstract
Prevailing frameworks propose that a key feature of attention-deficit/hyperactivity disorder (ADHD) is lower motivation. An important component of motivation is the willingness to engage in cognitively or physically effortful behavior. However, the degree to which effort sensitivity is impaired in ADHD has rarely been tested, and the efficacy of stimulant medication in ameliorating any such impairments is unclear. Here, we tested 20 individuals with ADHD (11 males, 9 females) who were managed with amphetamine-based medication (dexamfetamine, lisdexamfetamine), and 24 controls (8 males, 16 females). Individuals with ADHD were tested over two counterbalanced sessions, ON and OFF their usual amphetamine-based medication. In each session, participants performed an effort-based decision-making task, in which they were required to choose how much cognitive or physical effort they were willing to engage in return for reward. Our results revealed three main findings. First, individuals with ADHD had lower motivation relative to controls to invest effort in both the cognitive and physical domains. Second, amphetamine increased motivation uniformly across both domains. Finally, the net effect of amphetamine treatment was to mostly restore motivation across both domains of effort relative to healthy controls. These data provide clear evidence for a heightened sensitivity to both cognitive and physical effort in ADHD, and reveal the efficacy of amphetamine-based drugs in restoring effort sensitivity to levels similar to controls. These findings confirm the existence of reduced motivational drive in ADHD, and more broadly provide direct causal evidence for a domain-general role of catecholamines in motivating effortful behavior.SIGNIFICANCE STATEMENT A core feature of attention-deficit/hyperactivity disorder (ADHD) is thought to be a heightened aversion to effort. Surprisingly, however, the degree to which effort sensitivity is impaired in ADHD has rarely been tested. More broadly, the relative efficacy of catecholamines in motivating the investment of cognitive and physical effort is unclear. We tested 20 individuals with ADHD ON and OFF amphetamines, and compared their behavior on an effort-based decision-making task to 24 controls. When tested OFF medication, the ADHD group was less cognitively and physically motivated than controls. However, amphetamines led to a comparable increase in motivation across both domains. This demonstrates the efficacy of catecholamines in facilitating domain-general effort, and highlights the broader potential of such drugs to treat disorders of motivation.
Collapse
Affiliation(s)
- Trevor T-J Chong
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Victoria 3800, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria 3004, Australia
- Department of Clinical Neurosciences, St Vincent's Hospital, Melbourne, Victoria 3065, Australia
| | - Erika Fortunato
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Victoria 3800, Australia
| | - Mark A Bellgrove
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
7
|
Lai CW, Chang CH. Pharmacological activation of the amygdala, but not single prolonged footshock-induced acute stress, interferes with cue-induced motivation toward food rewards in rats. Front Behav Neurosci 2023; 17:1252868. [PMID: 37781505 PMCID: PMC10538645 DOI: 10.3389/fnbeh.2023.1252868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/25/2023] [Indexed: 10/03/2023] Open
Abstract
In the face of threats, animals adapt their behaviors to cope with the situation. Under such circumstances, irrelevant behaviors are usually suppressed. In this study, we examined whether food-seeking motivation would decrease under activation of the amygdala, an important nucleus in the regulation of stress response in the central nervous system, or after a physical acute stress session. In Experiment 1, we pharmacologically activated the basolateral nucleus (BLA) or the central nucleus of the amygdala (CeA) before a cue-induced reinstatement test in rats. Our results showed that activation of the BLA or the CeA abolished cue-induced motivation toward food rewards, while locomotor activity and free food intake were not affected. In Experiments 2 and 3, we further assessed anxiety and despair levels, as well as cue-induced reinstatement, after a single prolonged footshock-induced acute stress in rats. Behaviorally, acute stress did not affect anxiety level, despair level, or cue-induced motivation toward food rewards. Physiologically, there was no difference in cellular activities of the amygdala immediately after acute stress. To conclude, our results suggested that pharmacological activation of the amygdala decreased cue-induced motivation toward food reward. However, physiological acute stress did not immediately interfere with the negative emotions, motivation, or amygdala activities of the animals.
Collapse
Affiliation(s)
- Chien-Wen Lai
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Chun-hui Chang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, Taiwan
- Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
8
|
Edwin Thanarajah S, DiFeliceantonio AG, Albus K, Kuzmanovic B, Rigoux L, Iglesias S, Hanßen R, Schlamann M, Cornely OA, Brüning JC, Tittgemeyer M, Small DM. Habitual daily intake of a sweet and fatty snack modulates reward processing in humans. Cell Metab 2023; 35:571-584.e6. [PMID: 36958330 DOI: 10.1016/j.cmet.2023.02.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 10/21/2022] [Accepted: 02/23/2023] [Indexed: 03/25/2023]
Abstract
Western diets rich in fat and sugar promote excess calorie intake and weight gain; however, the underlying mechanisms are unclear. Despite a well-documented association between obesity and altered brain dopamine function, it remains elusive whether these alterations are (1) pre-existing, increasing the individual susceptibility to weight gain, (2) secondary to obesity, or (3) directly attributable to repeated exposure to western diet. To close this gap, we performed a randomized, controlled study (NCT05574660) with normal-weight participants exposed to a high-fat/high-sugar snack or a low-fat/low-sugar snack for 8 weeks in addition to their regular diet. The high-fat/high-sugar intervention decreased the preference for low-fat food while increasing brain response to food and associative learning independent of food cues or reward. These alterations were independent of changes in body weight and metabolic parameters, indicating a direct effect of high-fat, high-sugar foods on neurobehavioral adaptations that may increase the risk for overeating and weight gain.
Collapse
Affiliation(s)
- Sharmili Edwin Thanarajah
- Max Planck Institute for Metabolism Research, Cologne, Germany; Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Alexandra G DiFeliceantonio
- Fralin Biomedical Research Institute at Virginia Tech Carilion & Department of Human Nutrition, Foods, and Exercise, College of Agriculture and Life Sciences, Roanoke, VA, USA
| | - Kerstin Albus
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) & Excellence Center for Medical Mycology (ECMM), Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | | | - Lionel Rigoux
- Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Sandra Iglesias
- Translational Neuromodeling Unit, Institute for Biomedical Engineering, University of Zurich and Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Ruth Hanßen
- Max Planck Institute for Metabolism Research, Cologne, Germany; Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEPD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Marc Schlamann
- Department of Neuroradiology, University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Oliver A Cornely
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) & Excellence Center for Medical Mycology (ECMM), Faculty of Medicine and University Hospital Cologne, Cologne, Germany; German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany; Clinical Trials Centre Cologne (ZKS Köln), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Jens C Brüning
- Max Planck Institute for Metabolism Research, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEPD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Marc Tittgemeyer
- Max Planck Institute for Metabolism Research, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.
| | - Dana M Small
- Modern Diet and Physiology Research Center, New Haven, CT, USA; Yale University School of Medicine, Department of Psychiatry, New Haven, CT, USA.
| |
Collapse
|
9
|
Yuen J, Goyal A, Rusheen AE, Kouzani AZ, Berk M, Kim JH, Tye SJ, Blaha CD, Bennet KE, Lee KH, Shin H, Oh Y. High frequency deep brain stimulation can mitigate the acute effects of cocaine administration on tonic dopamine levels in the rat nucleus accumbens. Front Neurosci 2023; 17:1061578. [PMID: 36793536 PMCID: PMC9922701 DOI: 10.3389/fnins.2023.1061578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 01/09/2023] [Indexed: 01/31/2023] Open
Abstract
Cocaine's addictive properties stem from its capacity to increase tonic extracellular dopamine levels in the nucleus accumbens (NAc). The ventral tegmental area (VTA) is a principal source of NAc dopamine. To investigate how high frequency stimulation (HFS) of the rodent VTA or nucleus accumbens core (NAcc) modulates the acute effects of cocaine administration on NAcc tonic dopamine levels multiple-cyclic square wave voltammetry (M-CSWV) was used. VTA HFS alone decreased NAcc tonic dopamine levels by 42%. NAcc HFS alone resulted in an initial decrease in tonic dopamine levels followed by a return to baseline. VTA or NAcc HFS following cocaine administration prevented the cocaine-induced increase in NAcc tonic dopamine. The present results suggest a possible underlying mechanism of NAc deep brain stimulation (DBS) in the treatment of substance use disorders (SUDs) and the possibility of treating SUD by abolishing dopamine release elicited by cocaine and other drugs of abuse by DBS in VTA, although further studies with chronic addiction models are required to confirm that. Furthermore, we demonstrated the use of M-CSWV can reliably measure tonic dopamine levels in vivo with both drug administration and DBS with minimal artifacts.
Collapse
Affiliation(s)
- Jason Yuen
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Barwon Health, Deakin University, Geelong, VIC, Australia
| | - Abhinav Goyal
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
- Medical Scientist Training Program, Mayo Clinic, Rochester, MN, United States
| | - Aaron E. Rusheen
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
- Medical Scientist Training Program, Mayo Clinic, Rochester, MN, United States
| | - Abbas Z. Kouzani
- School of Engineering, Deakin University, Geelong, VIC, Australia
| | - Michael Berk
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Barwon Health, Deakin University, Geelong, VIC, Australia
| | - Jee Hyun Kim
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Barwon Health, Deakin University, Geelong, VIC, Australia
| | - Susannah J. Tye
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Charles D. Blaha
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Kevin E. Bennet
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
- Division of Engineering, Mayo Clinic, Rochester, MN, United States
| | - Kendall H. Lee
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
- Department of Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Hojin Shin
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
- Department of Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Yoonbae Oh
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
- Department of Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
10
|
D1 receptor-expressing neurons in ventral tegmental area alleviate mouse anxiety-like behaviors via glutamatergic projection to lateral septum. Mol Psychiatry 2023; 28:625-638. [PMID: 36195641 PMCID: PMC9531220 DOI: 10.1038/s41380-022-01809-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 09/18/2022] [Accepted: 09/20/2022] [Indexed: 11/08/2022]
Abstract
Dopamine (DA) acts as a key regulator in controlling emotion, and dysfunction of DA signal has been implicated in the pathophysiology of some psychiatric disorders, including anxiety. Ventral tegmental area (VTA) is one of main regions with DA-producing neurons. VTA DAergic projections in mesolimbic brain regions play a crucial role in regulating anxiety-like behaviors, however, the function of DA signal within VTA in regulating emotion remains unclear. Here, we observe that pharmacological activation/inhibition of VTA D1 receptors will alleviate/aggravate mouse anxiety-like behaviors, and knockdown of VTA D1 receptor expression also exerts anxiogenic effect. With fluorescence in situ hybridization and electrophysiological recording, we find that D1 receptors are functionally expressed in VTA neurons. Silencing/activating VTA D1 neurons bidirectionally modulate mouse anxiety-like behaviors. Furthermore, knocking down D1 receptors in VTA DA and glutamate neurons elevates anxiety-like state, but in GABA neurons has the opposite effect. In addition, we identify the glutamatergic projection from VTA D1 neurons to lateral septum is mainly responsible for the anxiolytic effect induced by activating VTA D1 neurons. Thus, our study not only characterizes the functional expression of D1 receptors in VTA neurons, but also uncovers the pivotal role of DA signal within VTA in mediating anxiety-like behaviors.
Collapse
|
11
|
Rodríguez-Flores TC, Palomo-Briones GA, Robles F, Ramos F. Proposal for a computational model of incentive memory. COGN SYST RES 2022. [DOI: 10.1016/j.cogsys.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
12
|
The Modification of Offspring Stress-Related Behavior and the Expression of Drd1, Drd2, and Nr3c1 by a Western-Pattern Diet in Mus Musculus. Int J Mol Sci 2022; 23:ijms23169245. [PMID: 36012509 PMCID: PMC9409213 DOI: 10.3390/ijms23169245] [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: 06/30/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
Abstract
The impact of early developmental experience on neurobiological pathways that may contribute to the association between diet and behavior have not yet been elucidated. The focus of the current study was to determine whether the impact of prenatal stress (PS) could be mitigated by a diet that stimulates the same neuroendocrine systems influenced by early stress, using a mouse model. Behavioral and genetic approaches were used to assess how a Western-pattern diet (WPD) interacts with PS and sex to impact the expression of anxiety-like behavior in an open-field arena, as well as the expression of the glucocorticoid receptor in the hippocampus, D1 dopamine receptors in the nucleus accumbens, and D2 dopamine receptors in the ventral tegmental area. Overall, the results demonstrated that a prenatal WPD mitigates the effects of maternal stress in dams and offspring. These results help to elucidate the relationship between pre- and post-natal nutrition, gene expression, and behaviors that lead to long-term health effects.
Collapse
|
13
|
Neurotensin Release from Dopamine Neurons Drives Long-Term Depression of Substantia Nigra Dopamine Signaling. J Neurosci 2022; 42:6186-6194. [PMID: 35794014 PMCID: PMC9374153 DOI: 10.1523/jneurosci.1395-20.2022] [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: 06/02/2020] [Revised: 05/11/2022] [Accepted: 06/12/2022] [Indexed: 11/21/2022] Open
Abstract
Midbrain dopamine neurons play central physiological roles in voluntary movement, reward learning, and motivated behavior. Inhibitory signaling at somatodendritic dopamine D2 receptor (D2R) synapses modulates excitability of dopamine neurons. The neuropeptide neurotensin is expressed by many inputs to the midbrain and induces LTD of D2R synaptic currents (LTDDA); however, the source of neurotensin that is responsible for LTDDA is not known. Here we show, in brain slices from male and female mice, that LTDDA is driven by neurotensin released by dopamine neurons themselves. Optogenetic stimulation of dopamine neurons was sufficient to induce LTDDA in the substantia nigra, but not the VTA, and was dependent on neurotensin receptor signaling, postsynaptic calcium, and vacuolar-type H+-ATPase activity in the postsynaptic cell. These findings reveal a novel form of signaling between dopamine neurons involving release of the peptide neurotensin, which may act as a feedforward mechanism to increase dopamine neuron excitability.SIGNIFICANCE STATEMENT Dopamine neurons in the midbrain play a critical role in reward learning and the initiation of movement. Aberrant dopamine neuron function is implicated in a range of diseases and disorders, including Parkinson's disease, schizophrenia, obesity, and substance use disorders. D2 receptor-mediated PSCs are produced by a rare form of dendrodendritic synaptic transmission between dopamine neurons. These D2 receptor-mediated PSCs undergo LTD following application of the neuropeptide neurotensin. Here we show that release of neurotensin by dopamine neurons themselves is sufficient to induce LTD of dopamine transmission in the substantia nigra. Neurotensin signaling therefore mediates a second form of interdopamine neuron communication and may provide a mechanism by which dopamine neurons maintain excitability when nigral dopamine is elevated.
Collapse
|
14
|
Guleken Z, Uzbay T. Neurobiological and neuropharmacological aspects of food addiction. Neurosci Biobehav Rev 2022; 139:104760. [PMID: 35780976 DOI: 10.1016/j.neubiorev.2022.104760] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 06/04/2022] [Accepted: 06/26/2022] [Indexed: 11/17/2022]
Abstract
This review aims to draw attention to current studies on syndromes related to food eating behavior, including food addiction, and to highlight the neurobiological and neuropharmacological aspects of food addiction toward the development of new therapies. Food addiction and eating disorders are influenced by several neurobiological factors. Changes in feeding behavior, food addiction, and its pharmacological therapy are related to complex neurobiological processes in the brain. Thus, it is not surprising that there is inconsistency among various individual studies. In this review, we assessed literature including both experimental and clinical studies regarding food addiction as a feeding disorder. We selected articles from animal studies, randomized clinical trials, meta-analyses, narrative, and systemic reviews given that, crucial quantitative data with a measure of neurobiological, neuropharmacological aspects and current therapies of food addiction as an outcome. Thus, the main goal to outline here is to investigate and discuss the association between the brain reward system and feeding behavior in the frame of food addiction in the light of current literature.
Collapse
Affiliation(s)
- Zozan Guleken
- Uskudar University Faculty of Medicine, Department of Physiology, İstanbul, Turkey
| | - Tayfun Uzbay
- Uskudar University, Faculty of Medicine, Department of Medical Pharmacology, İstanbul, Turkey; Üsküdar University, Neuropsychopharmacology Application, and Research Center (NPARC), İstanbul, Turkey.
| |
Collapse
|
15
|
Martins JS, Joyner KJ, McCarthy DM, Morris DH, Patrick CJ, Bartholow BD. Differential brain responses to alcohol-related and natural rewards are associated with alcohol use and problems: Evidence for reward dysregulation. Addict Biol 2022; 27:e13118. [PMID: 34877771 PMCID: PMC8891069 DOI: 10.1111/adb.13118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 10/15/2021] [Accepted: 11/04/2021] [Indexed: 12/17/2022]
Abstract
Multiple theoretical perspectives posit that drug use leads to biased valuation of drug-related reward, at the expense of naturally occurring rewarding activities (i.e., reward dysregulation). Recent research suggests that the comparative balance of drug-related and nondrug-related reward valuation is a powerful determinant of substance misuse and addiction. We examined differential neurophysiological responses-indexed with the P3 component of the event-related potential (ERP)-elicited by visual alcohol cues and cues depicting natural reward as a neurobiological indicator of problematic drinking. Nondependent, young adult drinkers (N = 143, aged 18-30 years) completed questionnaire measures assessing alcohol use and problems, and viewed alcohol cues (pictures of alcoholic beverages), high-arousing natural reward cues (erotica, adventure scenes), nonalcoholic beverage cues, and neutral scenes (e.g., household items) while ERPs were recorded. When examined separately, associations of P3-ERP reactivity to alcohol cues and natural reward cues with alcohol use and problems were weak. However, differential P3 response to the two types of cues (i.e., reward dysregulation P3) showed consistent and robust associations with all indices of alcohol use and problems and differentiated high-risk from lower-risk drinkers. The current results support the idea that the differential incentive-motivational value of alcohol, relative to naturally rewarding activities, is associated with increased risk for substance misuse and dependence, and highlight a novel neurophysiological indicator-the reward dysregulation P3-of this differential reward valuation.
Collapse
|
16
|
Increased elasticity of sucrose demand during hyperdopaminergic states in rats. Psychopharmacology (Berl) 2022; 239:773-794. [PMID: 35102422 PMCID: PMC8891210 DOI: 10.1007/s00213-022-06068-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/17/2022] [Indexed: 10/29/2022]
Abstract
RATIONALE Deficits in cost-benefit decision-making are a core feature of several psychiatric disorders, including substance addiction, eating disorders and bipolar disorder. Mesocorticolimbic dopamine signalling has been implicated in various processes related to cognition and reward, but its precise role in reward valuation and cost-benefit trade-off decisions remains incompletely understood. OBJECTIVES We assessed the role of mesocorticolimbic dopamine signalling in the relationship between price and consumption of sucrose, to better understand its role in cost-benefit decisions. METHODS Dopamine neurons in the ventral tegmental area (VTA) were chemogenetically activated in rats, and a behavioural economics approach was used to quantify the relationship between price and consumption of sucrose. Motivation for sucrose was also assessed under a progressive ratio (PR) schedule of reinforcement. To further gauge the role of dopamine in cost-benefit trade-offs for sucrose, the effects of treatment with D-amphetamine and the dopamine receptor antagonist alpha-flupentixol were assessed. RESULTS Chemogenetic activation of VTA dopamine neurons increased demand elasticity, while responding for sucrose under a PR schedule of reinforcement was augmented upon stimulation of VTA dopamine neurons. Treatment with amphetamine partially replicated the effects of chemogenetic dopamine neuron activation, whereas treatment with alpha-flupentixol reduced free consumption of sucrose and had mixed effects on demand elasticity. CONCLUSIONS Stimulation of mesocorticolimbic dopaminergic neurotransmission altered cost-benefit trade-offs in a complex manner. It reduced the essential value of palatable food, increased incentive motivation and left free consumption unaltered. Together, these findings imply that mesocorticolimbic dopamine signalling differentially influences distinct components of cost expenditure processes aimed at obtaining rewards.
Collapse
|
17
|
Simpson EH, Gallo EF, Balsam PD, Javitch JA, Kellendonk C. How changes in dopamine D2 receptor levels alter striatal circuit function and motivation. Mol Psychiatry 2022; 27:436-444. [PMID: 34385603 PMCID: PMC8837728 DOI: 10.1038/s41380-021-01253-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 02/07/2023]
Abstract
It was first posited, more than five decades ago, that the etiology of schizophrenia involves overstimulation of dopamine receptors. Since then, advanced clinical research methods, including brain imaging, have refined our understanding of the relationship between striatal dopamine and clinical phenotypes as well as disease trajectory. These studies point to striatal dopamine D2 receptors, the main target for all current antipsychotic medications, as being involved in both positive and negative symptoms. Simultaneously, animal models have been central to investigating causal relationships between striatal dopamine D2 receptors and behavioral phenotypes relevant to schizophrenia. We begin this article by reviewing the circuit, cell-type and subcellular locations of dopamine D2 receptors and their downstream signaling pathways. We then summarize results from several mouse models in which D2 receptor levels were altered in various brain regions, cell-types and developmental periods. Behavioral, electrophysiological and anatomical consequences of these D2 receptor perturbations are reviewed with a selective focus on striatal circuit function and alterations in motivated behavior, a core negative symptom of schizophrenia. These studies show that D2 receptors serve distinct physiological roles in different cell types and at different developmental time points, regulating motivated behaviors in sometimes opposing ways. We conclude by considering the clinical implications of this complex regulation of striatal circuit function by D2 receptors.
Collapse
Affiliation(s)
- Eleanor H. Simpson
- Division of Developmental Neuroscience, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY 10032, United States,Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY 10032, United States
| | - Eduardo F. Gallo
- Department of Biological Sciences, Fordham University, 441 East Fordham Road, Bronx, NY 10458
| | - Peter D. Balsam
- Division of Developmental Neuroscience, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY 10032, United States,Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY 10032, United States,Department of Psychology, Barnard College, 3009 Broadway, New York, NY 10027,Department of Psychology, Columbia University, 1190 Amsterdam Ave, New York, NY 10027
| | - Jonathan A. Javitch
- Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY 10032, United States,Division of Molecular Therapeutics, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY 10032,Department of Molecular Pharmacology and Therapeutics, Columbia University, 1051 Riverside Drive, New York, NY 10032
| | - Christoph Kellendonk
- Department of Psychiatry, Columbia University, New York, NY, USA. .,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA. .,Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY, USA.
| |
Collapse
|
18
|
Poisson CL, Engel L, Saunders BT. Dopamine Circuit Mechanisms of Addiction-Like Behaviors. Front Neural Circuits 2021; 15:752420. [PMID: 34858143 PMCID: PMC8631198 DOI: 10.3389/fncir.2021.752420] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/08/2021] [Indexed: 12/16/2022] Open
Abstract
Addiction is a complex disease that impacts millions of people around the world. Clinically, addiction is formalized as substance use disorder (SUD), with three primary symptom categories: exaggerated substance use, social or lifestyle impairment, and risky substance use. Considerable efforts have been made to model features of these criteria in non-human animal research subjects, for insight into the underlying neurobiological mechanisms. Here we review evidence from rodent models of SUD-inspired criteria, focusing on the role of the striatal dopamine system. We identify distinct mesostriatal and nigrostriatal dopamine circuit functions in behavioral outcomes that are relevant to addictions and SUDs. This work suggests that striatal dopamine is essential for not only positive symptom features of SUDs, such as elevated intake and craving, but also for impairments in decision making that underlie compulsive behavior, reduced sociality, and risk taking. Understanding the functional heterogeneity of the dopamine system and related networks can offer insight into this complex symptomatology and may lead to more targeted treatments.
Collapse
Affiliation(s)
- Carli L. Poisson
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, MN, United States
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Liv Engel
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, MN, United States
| | - Benjamin T. Saunders
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, MN, United States
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
19
|
Trace amine-associated receptor 1 (TAAR1): Potential application in mood disorders: A systematic review. Neurosci Biobehav Rev 2021; 131:192-210. [PMID: 34537265 DOI: 10.1016/j.neubiorev.2021.09.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/07/2021] [Accepted: 09/12/2021] [Indexed: 12/29/2022]
Abstract
There is a need for innovation with respect to therapeutics in psychiatry. Available evidence indicates that the trace amine-associated receptor 1 (TAAR1) agonist SEP-363856 is promising, as it improves measures of cognitive and reward function in schizophrenia. Hedonic and cognitive impairments are transdiagnostic and constitute major burdens in mood disorders. Herein, we systematically review the behavioural and genetic literature documenting the role of TAAR1 in reward and cognitive function, and propose a mechanistic model of TAAR1's functions in the brain. Notably, TAAR1 activity confers antidepressant-like effects, enhances attention and response inhibition, and reduces compulsive reward seeking without impairing normal function. Further characterization of the responsible mechanisms suggests ion-homeostatic, metabolic, neurotrophic, and anti-inflammatory enhancements in the limbic system. Multiple lines of evidence establish the viability of TAAR1 as a biological target for the treatment of mood disorders. Furthermore, the evidence suggests a role for TAAR1 in reward and cognitive function, which is attributed to a cascade of events that are relevant to the cellular integrity and function of the central nervous system.
Collapse
|
20
|
Individual differences in dopamine uptake in the dorsomedial striatum prior to cocaine exposure predict motivation for cocaine in male rats. Neuropsychopharmacology 2021; 46:1757-1767. [PMID: 33953341 PMCID: PMC8357974 DOI: 10.1038/s41386-021-01009-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/27/2021] [Accepted: 03/31/2021] [Indexed: 11/08/2022]
Abstract
A major theme of addiction research has focused on the neural substrates of individual differences in the risk for addiction; however, little is known about how vulnerable populations differ from those that are relatively protected. Here, we prospectively measured dopamine (DA) neurotransmission prior to cocaine exposure to predict the onset and course of cocaine use. Using in vivo voltammetry, we first generated baseline profiles of DA release and uptake in the dorsomedial striatum (DMS) and nucleus accumbens of drug-naïve male rats prior to exposing them to cocaine using conditioned place preference (CPP) or operant self-administration. We found that the innate rate of DA uptake in the DMS strongly predicted motivation for cocaine and drug-primed reinstatement, but not CPP, responding when "price" was low, or extinction. We then assessed the impact of baseline variations in DA uptake on cocaine potency in the DMS using ex vivo voltammetry in naïve rats and in rats with DA transporter (DAT) knockdown. DA uptake in the DMS of naïve rats predicted the neurochemical response to cocaine, such that rats with innately faster rates of DA uptake demonstrated higher cocaine potency at the DAT and rats with DAT knockdown displayed reduced potency compared to controls. Together, these data demonstrate that inherent variability in DA uptake in the DMS predicts the behavioral response to cocaine, potentially by altering the apparent potency of cocaine.
Collapse
|
21
|
Clare K, Pan C, Kim G, Park K, Zhao J, Volkow ND, Lin Z, Du C. Cocaine Reduces the Neuronal Population While Upregulating Dopamine D2-Receptor-Expressing Neurons in Brain Reward Regions: Sex-Effects. Front Pharmacol 2021; 12:624127. [PMID: 33912043 PMCID: PMC8072657 DOI: 10.3389/fphar.2021.624127] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/08/2021] [Indexed: 02/03/2023] Open
Abstract
Addiction to cocaine is associated with dysfunction of the dopamine mesocortical system including impaired dopamine-2 receptor (D2r) signaling. However, the effects of chronic cocaine on neuronal adaptations in this system have not been systematically examined and data available is mostly from males. Here, we investigated changes in the total neuronal density and relative concentration of D2r-expressing neurons in the medial prefrontal cortex (mPFC), dorsal striatum (Dstr), nucleus accumbens (NAc), and ventral tegmental area (VTA) in both male and female mice passively exposed to cocaine for two weeks. In parallel experiments, we measured mRNA levels for Drd2 and for opioid peptides (mPenk and mPdyn). Through a combination of large field of view fluorescent imaging with BAC transgenic D2r-eGFP mice and immunostaining, we observed that cocaine exposed mice had a higher density of D2r-positive cells that was most prominent in mPFC and VTA and larger for females than for males. This occurred amidst an overall significant decrease in neuronal density (measured with NeuN) in both sexes. However, increases in Drd2 mRNA levels with cocaine were only observed in mPFC and Dstr in females, which might reflect the limited sensitivity of the method. Our findings, which contrast with previous findings of cocaine-induced downregulation of D2r binding availability, could reflect a phenotypic shift in neurons that did not previously express Drd2 and merits further investigation. Additionally, the neuronal loss particularly in mPFC with chronic cocaine might contribute to the cognitive impairments observed with cocaine use disorder.
Collapse
Affiliation(s)
- Kevin Clare
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, United States
| | - Chelsea Pan
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, United States
| | - Gloria Kim
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, United States
| | - Kicheon Park
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, United States
| | - Juan Zhao
- Laboratory of Psychiatric Neurogenomics, Basic Neuroscience Division, McLean Hospital, Belmont, MA, United States
| | - Nora D Volkow
- National Institute on Drug Abuse, Bethesda, MD, United States
| | - Zhicheng Lin
- Laboratory of Psychiatric Neurogenomics, Basic Neuroscience Division, McLean Hospital, Belmont, MA, United States
| | - Congwu Du
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
22
|
Differential Impact of Inhibitory G-Protein Signaling Pathways in Ventral Tegmental Area Dopamine Neurons on Behavioral Sensitivity to Cocaine and Morphine. eNeuro 2021; 8:ENEURO.0081-21.2021. [PMID: 33707203 PMCID: PMC8114902 DOI: 10.1523/eneuro.0081-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/21/2022] Open
Abstract
Drugs of abuse engage overlapping but distinct molecular and cellular mechanisms to enhance dopamine (DA) signaling in the mesocorticolimbic circuitry. DA neurons of the ventral tegmental area (VTA) are key substrates of drugs of abuse and have been implicated in addiction-related behaviors. Enhanced VTA DA neurotransmission evoked by drugs of abuse can engage inhibitory G-protein-dependent feedback pathways, mediated by GABAB receptors (GABABRs) and D2 DA receptors (D2Rs). Chemogenetic inhibition of VTA DA neurons potently suppressed baseline motor activity, as well as the motor-stimulatory effect of cocaine and morphine, confirming the critical influence of VTA DA neurons and inhibitory G-protein signaling in these neurons on this addiction-related behavior. To resolve the relative influence of GABABR-dependent and D2R-dependent signaling pathways in VTA DA neurons on behavioral sensitivity to drugs of abuse, we developed a neuron-specific viral CRISPR/Cas9 approach to ablate D2R and GABABR in VTA DA neurons. Ablation of GABABR or D2R did not impact baseline physiological properties or excitability of VTA DA neurons, but it did preclude the direct somatodendritic inhibitory influence of GABABR or D2R activation. D2R ablation potentiated the motor-stimulatory effect of cocaine in male and female mice, whereas GABABR ablation selectively potentiated cocaine-induced activity in male subjects only. Neither D2R nor GABABR ablation impacted morphine-induced motor activity. Collectively, our data show that cocaine and morphine differ in the extent to which they engage inhibitory G-protein-dependent feedback pathways in VTA DA neurons and highlight key sex differences that may impact susceptibility to various facets of addiction.
Collapse
|
23
|
Corticosterone Attenuates Reward-Seeking Behavior and Increases Anxiety via D2 Receptor Signaling in Ventral Tegmental Area Dopamine Neurons. J Neurosci 2021; 41:1566-1581. [PMID: 33372063 PMCID: PMC7896015 DOI: 10.1523/jneurosci.2533-20.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 01/13/2023] Open
Abstract
Corticosteroids (CORT) have been widely used in anti-inflammatory medication. Chronic CORT treatment can cause mesocorticolimbic system dysfunctions, which are known to play a key role for the development of psychiatric disorders. The VTA is a critical site in the mesocorticolimbic pathway and is responsible for motivation and reward-seeking behaviors. However, the mechanism by which chronic CORT alters VTA dopamine neuronal activity is largely unknown. We treated periadolescent male mice with vehicle, 1 d, or 7 d CORT in the drinking water, examined behavioral impacts with light/dark box, elevated plus maze, operant chamber, and open field tests, measured the effects of CORT on VTA dopamine neuronal activity using patch-clamp electrophysiology and dopamine concentration using fast-scan cyclic voltammetry, and tested the effects of dopamine D2 receptor (D2R) blockade by intra-VTA infusion of a D2R antagonist. CORT treatment induced anxiety-like behavior as well as decreased food-seeking behaviors. We show that chronic CORT treatment decreased excitability and excitatory synaptic transmission onto VTA dopamine neurons. Furthermore, chronic CORT increased somatodendritic dopamine concentration. The D2R antagonist sulpiride restored decreased excitatory transmission and excitability of VTA dopamine neurons. Furthermore, sulpiride decreased anxiety-like behavior and rescued food-seeking behavior in mice with chronic CORT exposure. Together, 7 d CORT treatment induces anxiety-like behavior and impairs food-seeking in a mildly aversive environment. D2R signaling in the VTA might be a potential target to ameliorate chronic CORT-induced anxiety and reward-seeking deficits. SIGNIFICANCE STATEMENT With widespread anti-inflammatory effects throughout the body, corticosteroids (CORT) have been used in a variety of therapeutic conditions. However, long-term CORT treatment causes cognitive impairments and neuropsychiatric disorders. The impact of chronic CORT on the mesolimbic system has not been elucidated. Here, we demonstrate that 7 d CORT treatment increases anxiety-like behavior and attenuates food-seeking behavior in a mildly aversive environment. By elevating local dopamine concentration in the VTA, a region important for driving motivated behavior, CORT treatment suppresses excitability and synaptic transmission onto VTA dopamine neurons. Intriguingly, blockade of D2 receptor signaling in the VTA restores neuronal excitability and food-seeking and alleviates anxiety-like behaviors. Our findings provide a potential therapeutic target for CORT-induced reward deficits.
Collapse
|
24
|
Using pharmacological manipulations to study the role of dopamine in human reward functioning: A review of studies in healthy adults. Neurosci Biobehav Rev 2020; 120:123-158. [PMID: 33202256 DOI: 10.1016/j.neubiorev.2020.11.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 10/30/2020] [Accepted: 11/01/2020] [Indexed: 01/08/2023]
Abstract
Dopamine (DA) plays a key role in reward processing and is implicated in psychological disorders such as depression, substance use, and schizophrenia. The role of DA in reward processing is an area of highly active research. One approach to this question is drug challenge studies with drugs known to alter DA function. These studies provide good experimental control and can be performed in parallel in laboratory animals and humans. This review aimed to summarize results of studies using pharmacological manipulations of DA in healthy adults. 'Reward' is a complex process, so we separated 'phases' of reward, including anticipation, evaluation of cost and benefits of upcoming reward, execution of actions to obtain reward, pleasure in response to receiving a reward, and reward learning. Results indicated that i) DAergic drugs have different effects on different phases of reward; ii) the relationship between DA and reward functioning appears unlikely to be linear; iii) our ability to detect the effects of DAergic drugs varies depending on whether subjective, behavioral, imaging measures are used.
Collapse
|
25
|
Doyle MA, Stark AR, Fejes-Tóth G, Náray-Fejes-Tóth A, Mazei-Robison MS. Behavioral effects of SGK1 knockout in VTA and dopamine neurons. Sci Rep 2020; 10:14751. [PMID: 32901079 PMCID: PMC7478959 DOI: 10.1038/s41598-020-71681-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/28/2020] [Indexed: 12/15/2022] Open
Abstract
Drugs of abuse cause significant neuroadaptations within the ventral tegmental area (VTA), with alterations in gene expression tied to changes in reward behavior. Serum- and glucocorticoid-inducible kinase 1 (SGK1) transcription, catalytic activity, and phosphorylation are upregulated in the VTA by chronic cocaine or morphine treatment, positioning SGK1 as a critical mediator of reward behavior. Using transgenic mouse models, we investigated the effect of SGK1 knockout in the VTA and in dopamine (DA) neurons to evaluate the necessity of protein expression for natural and drug reward behaviors. SGK1 knockdown in the VTA did not impact reward behaviors. Given VTA cellular heterogeneity, we also investigated a DA neuron-specific SGK1 knockout (KO). DA SGK1 KO significantly decreased body weight of adult mice as well as increased general locomotor activity; however, reward behaviors were similarly unaltered. Given that SGK1 mutants virally overexpressed in the VTA are capable of altering drug-associated behavior, our current results suggest that changes in SGK1 protein signaling may be distinct from expression. This work yields novel information on the impact of SGK1 deletion, critical for understanding the role of SGK1 signaling in the central nervous system and evaluating SGK1 as a potential therapeutic target for treatment of substance use disorders.
Collapse
Affiliation(s)
- Marie A Doyle
- Neuroscience Program, Michigan State University, 766 Service Rd, ISTB 5017, East Lansing, MI, 48824, USA
| | - Ali R Stark
- Neuroscience Program, Michigan State University, 766 Service Rd, ISTB 5017, East Lansing, MI, 48824, USA
| | - Geza Fejes-Tóth
- Department of Molecular and Systems Biology, Dartmouth University, Hanover, USA
| | | | - Michelle S Mazei-Robison
- Neuroscience Program, Michigan State University, 766 Service Rd, ISTB 5017, East Lansing, MI, 48824, USA.
- Department of Physiology, Michigan State University, East Lansing, USA.
| |
Collapse
|
26
|
Jacobs AJ, Roskam AL, Hummel FM, Ronan PJ, Gorres-Martens BK. Exercise improves high-fat diet- and ovariectomy-induced insulin resistance in rats with altered hepatic fat regulation. Curr Res Physiol 2020; 3:11-19. [PMID: 34746816 PMCID: PMC8562195 DOI: 10.1016/j.crphys.2020.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/21/2020] [Accepted: 06/09/2020] [Indexed: 12/28/2022] Open
Abstract
A high-fat diet (HFD) and loss of endogenous estrogens increases the risk for type 2 diabetes (T2D) and insulin resistance. Although exercise is known to prevent and manage insulin resistance, the cellular mechanisms remain largely unknown, especially in the context of a combined HFD and endogenous estrogen loss via ovariectomy (OVX). This study uses female Wistar rats to assess the effect of diet, endogenous estrogens, an exercise on insulin resistance, serum hormones, hepatic AMPK, hepatic regulators of fat metabolism, and expression of signaling molecules of the brain reward pathway. The combination of the HFD/OVX increased the homeostatic model assessment of insulin resistance (HOMA-IR), the glucose-insulin (G-I) index, and the serum adiponectin and leptin values, and exercise decreased these factors. The combination of the HFD/OVX decreased hepatic pAMPK, and exercise restored hepatic pAMPK, an important regulator of fat and glucose metabolism. Furthermore, consumption of the HFD by rats with intact ovaries (and endogenous estrogens) did not result in these drastic changes compared to intact rats fed a standard diet, suggesting that the presence of estrogens provides whole body benefits. Additionally, the HFD decreased the hepatic protein expression of acetyl CoA carboxylase (ACC) and fatty acid synthase (FAS), two proteins involved in de novo lipid synthesis and increased the hepatic protein expression of lipoprotein lipase (LPL), a protein involved in fat storage. Finally, exercise increased mRNA expression of the dopamine D2 receptor and tyrosine hydroxylase in the dopaminergic neuron cell body region of the ventral tegmental area, which is a key component of the brain reward pathway. Overall, this study demonstrates that exercise prevents insulin resistance even when a HFD is combined with OVX, despite hepatic changes in ACC, FAS, and LPL.
Collapse
Affiliation(s)
| | - Adam L Roskam
- Chemistry Department, Mount Marty College, Yankton, SD, USA
| | - Faith M Hummel
- Biology Department, Black Hills State University, Spearfish, SD, USA
| | - Patrick J Ronan
- Veterans Affairs Research Service, Sioux Falls VA Health Care System, Sioux Falls, SD, USA.,Department of Psychiatry and Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | | |
Collapse
|
27
|
Suchanecka A, Chmielowiec J, Chmielowiec K, Masiak J, Sipak-Szmigiel O, Sznabowicz M, Czarny W, Michałowska-Sawczyn M, Trybek G, Grzywacz A. Dopamine Receptor DRD2 Gene rs1076560, Personality Traits and Anxiety in the Polysubstance Use Disorder. Brain Sci 2020; 10:brainsci10050262. [PMID: 32365807 PMCID: PMC7287957 DOI: 10.3390/brainsci10050262] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 11/16/2022] Open
Abstract
Development of an addiction is conditioned by many factors. The dopaminergic system has been shown to be the key element in this process. In this paper, we analyzed the influence of dopamine receptor 2 polymorphism rs1076560 in two groups—polysubstance-dependent male patients (n = 299) and the controls matched for age (n = 301). In both groups, we applied the same questionnaires for testing—Mini-international neuropsychiatric interview, the NEO Five-Factor Inventory, and the State–Trait Anxiety Inventory. The real-time PCR method was used for genotyping. When we compared the controls with the case group subjects, we observed significantly higher scores in the second group on both the state and trait scales of anxiety, as well as on the Neuroticism and Openness scales of the NEO-FFI; and lower scores on the scales of Extraversion and Agreeability of the NEO-FFI. The model 2 × 3 factorial ANOVA of the addicted subjects and controls was performed, and the DRD2 rs1076560 variant interaction was found for the anxiety state and trait scales, and for the NEO-FFI Neuroticism scale. The observed associations allow noticing that analysis of psychological factors in combination with genetic data opens new possibilities in addiction research.
Collapse
Affiliation(s)
- Aleksandra Suchanecka
- Independent Laboratory of Health Promotion of the Pomeranian Medical University in Szczecin, 11 Chlapowskiego St., 70-204 Szczecin, Poland;
| | - Jolanta Chmielowiec
- Department of Hygiene and Epidemiology, Collegium Medicum, University of Zielona Góra, Zyty 28 St., 65-046 Zielona Gora, Poland; (J.C.); (K.C.)
| | - Krzysztof Chmielowiec
- Department of Hygiene and Epidemiology, Collegium Medicum, University of Zielona Góra, Zyty 28 St., 65-046 Zielona Gora, Poland; (J.C.); (K.C.)
| | - Jolanta Masiak
- Neurophysiological Independent Unit, Department of Psychiatry, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Olimpia Sipak-Szmigiel
- Department of Obstetrics and Pathology of Pregnancy, Pomeranian Medical University, 48 Żołnierska St., 71-210 Szczecin, Poland;
| | - Mariusz Sznabowicz
- Indywidual Medical Practice MD M Sznabowicz, Lutówko 14, 74-320 Barlinek, Poland;
| | - Wojciech Czarny
- Faculty of Physical Education, University of Rzeszów, Towarnickiego 3 St., 35-959 Rzeszów, Poland;
| | - Monika Michałowska-Sawczyn
- Faculty of Physical Culture, Gdańsk University of Physical Education and Sport, Kazimierza Górskiego 1 St., 80-336 Gdańsk, Poland;
| | - Grzegorz Trybek
- Department of Oral Surgery, Pomeranian Medical University in Szczecin, 72 Powstańców Wlkp. St., 70-111 Szczecin, Poland;
| | - Anna Grzywacz
- Independent Laboratory of Health Promotion of the Pomeranian Medical University in Szczecin, 11 Chlapowskiego St., 70-204 Szczecin, Poland;
- Correspondence:
| |
Collapse
|
28
|
Effort-based decision making varies by smoking status. Psychopharmacology (Berl) 2020; 237:1081-1090. [PMID: 31900525 PMCID: PMC7125005 DOI: 10.1007/s00213-019-05437-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 12/13/2019] [Indexed: 10/25/2022]
Abstract
RATIONALE A reduced willingness to perform effort based on the magnitude and probability of potential rewards has been associated with diminished dopamine function and may be relevant to chronic drug use. OBJECTIVES Here, we investigated the influence of smoking status on effort-based decisions. We hypothesized that smokers would make fewer high-effort selections than ex-smokers and never-smokers. METHODS Current smokers (n = 25), ex-smokers (≥ 1 year quit, n = 23), and never-smokers (n = 19) completed the Effort Expenditure for Rewards Task in which participants select between low-effort and high-effort options to receive monetary rewards at varying levels of reward magnitude, probability and expected value. RESULTS Overall, participants selected more high-effort options as potential reward magnitude and expected value increased. Smokers did not make fewer high-effort selections overall, but smokers were less sensitive to the changes in magnitude, probability, and expected value compared to never-smokers. Smokers were also less sensitive to the changes in probability and expected value, but not magnitude, compared to ex-smokers. Among smokers and ex-smokers, less nicotine dependence was associated with an increased likelihood of high-effort selections. CONCLUSIONS These results demonstrate the relevance of smoking status to effort-based decisions and suggest that smokers have diminished sensitivity to nondrug reward value. Among ex-smokers, greater pre-existing sensitivity to reward value may have been conducive to smoking cessation, or sensitivity was improved by smoking cessation. Future prospective studies can investigate whether effort-related decision making is predictive of smoking initiation or cessation success. IMPLICATIONS Willingness to perform effort to achieve a goal and sensitivity to changes in reward value are important aspects of motivation. These results showed that smokers have decreased sensitivity to changes in effort-related reward probability and expected value compared to ex-smokers and never-smokers. Potentially, improved sensitivity to rewards among ex-smokers may be a cause or consequence of smoking cessation. These findings may help explain why some smokers are able to achieve long-term abstinence.
Collapse
|
29
|
Abiero A, Botanas CJ, Custodio RJ, Sayson LV, Kim M, Lee HJ, Kim HJ, Lee KW, Jeong Y, Seo JW, Ryu IS, Lee YS, Cheong JH. 4-MeO-PCP and 3-MeO-PCMo, new dissociative drugs, produce rewarding and reinforcing effects through activation of mesolimbic dopamine pathway and alteration of accumbal CREB, deltaFosB, and BDNF levels. Psychopharmacology (Berl) 2020; 237:757-772. [PMID: 31828394 DOI: 10.1007/s00213-019-05412-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 11/21/2019] [Indexed: 12/27/2022]
Abstract
RATIONALE A high number of synthetic dissociative drugs continue to be available through online stores, leading to their misuse. Recent inclusions in this category are 4-MeO-PCP and 3-MeO-PCMo, analogs of phencyclidine. Although the dissociative effects of these drugs and their recreational use have been reported, no studies have investigated their abuse potential. OBJECTIVES To examine their rewarding and reinforcing effects and explore the mechanistic correlations. METHODS We used conditioned place preference (CPP), self-administration, and locomotor sensitization tests to assess the rewarding and reinforcing effects of the drugs. We explored their mechanism of action by pretreating dopamine receptor (DR) D1 antagonist SCH23390 and DRD2 antagonist haloperidol during CPP test and investigated the effects of 4-MeO-PCP and 3-MeO-PCMo on dopamine-related proteins in the ventral tegmental area and nucleus accumbens. We also measured the levels of dopamine, phosphorylated cyclic-AMP response element-binding (p-CREB) protein, deltaFosB, and brain-derived neurotrophic factor (BDNF) in the nucleus accumbens. Additionally, we examined the effects of both drugs on brain wave activity using electroencephalography. RESULTS While both 4-MeO-PCP and 3-MeO-PCMo induced CPP and self-administration, only 4-MeO-PCP elicited locomotor sensitization. SCH23390 and haloperidol inhibited the acquisition of drug CPP. 4-MeO-PCP and 3-MeO-PCMo altered the levels of tyrosine hydroxylase, DRD1, DRD2, and dopamine, as well as that of p-CREB, deltaFosB, and BDNF. All drugs increased the delta and gamma wave activity, whereas pretreatment with SCH23390 and haloperidol inhibited it. CONCLUSION Our results indicate that 4-MeO-PCP and 3-MeO-PCMo induce rewarding and reinforcing effects that are probably mediated by the mesolimbic dopamine system, suggesting an abuse liability in humans.
Collapse
Affiliation(s)
- Arvie Abiero
- Uimyung Research Institute for Neuroscience, College of Pharmacy, Sahmyook University, 815 Hwarangro, Nowon-gu, Seoul, 01795, Republic of Korea
| | - Chrislean Jun Botanas
- Uimyung Research Institute for Neuroscience, College of Pharmacy, Sahmyook University, 815 Hwarangro, Nowon-gu, Seoul, 01795, Republic of Korea
| | - Raly James Custodio
- Uimyung Research Institute for Neuroscience, College of Pharmacy, Sahmyook University, 815 Hwarangro, Nowon-gu, Seoul, 01795, Republic of Korea
| | - Leandro Val Sayson
- Uimyung Research Institute for Neuroscience, College of Pharmacy, Sahmyook University, 815 Hwarangro, Nowon-gu, Seoul, 01795, Republic of Korea
| | - Mikyung Kim
- Uimyung Research Institute for Neuroscience, College of Pharmacy, Sahmyook University, 815 Hwarangro, Nowon-gu, Seoul, 01795, Republic of Korea
| | - Hyun Jun Lee
- Uimyung Research Institute for Neuroscience, College of Pharmacy, Sahmyook University, 815 Hwarangro, Nowon-gu, Seoul, 01795, Republic of Korea
| | - Hee Jin Kim
- Uimyung Research Institute for Neuroscience, College of Pharmacy, Sahmyook University, 815 Hwarangro, Nowon-gu, Seoul, 01795, Republic of Korea
| | - Kun Won Lee
- Medicinal Chemistry Laboratory, Department of Pharmacy & Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul, 02447, Republic of Korea
| | - Youngdo Jeong
- Medicinal Chemistry Laboratory, Department of Pharmacy & Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul, 02447, Republic of Korea
| | - Joung-Wook Seo
- Center for Safety Pharmacology, Korea Institute of Toxicology, Daejeon, 305-343, Republic of Korea
| | - In Soo Ryu
- Center for Safety Pharmacology, Korea Institute of Toxicology, Daejeon, 305-343, Republic of Korea
| | - Yong Sup Lee
- Medicinal Chemistry Laboratory, Department of Pharmacy & Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul, 02447, Republic of Korea.
| | - Jae Hoon Cheong
- Uimyung Research Institute for Neuroscience, College of Pharmacy, Sahmyook University, 815 Hwarangro, Nowon-gu, Seoul, 01795, Republic of Korea. .,School of Pharmacy, Chonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do, 54896, Republic of Korea.
| |
Collapse
|
30
|
Martin TA, Smith HR, Luessen DJ, Chen R, Porrino LJ. Functional brain activity is globally elevated by dopamine D2 receptor knockdown in the ventral tegmental area. Brain Res 2020; 1727:146552. [PMID: 31726041 PMCID: PMC6941665 DOI: 10.1016/j.brainres.2019.146552] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/07/2019] [Accepted: 11/10/2019] [Indexed: 12/14/2022]
Abstract
The mesocorticolimbic system is comprised of dopaminergic neurons in the ventral tegmental area (VTA) and their projection targets in the ventral striatum, amygdala, prefrontal cortex, and hippocampus, among others. Regulation of dopamine transmission within this system is achieved in part through a negative feedback mechanism via dopamine D2 autoreceptors located on somatodendrites and terminals of VTA dopaminergic neurons. Dysregulation of this mechanism has been implicated in addiction and other psychiatric disorders, although the biological bases for these associations are unclear. In order to elucidate the functional consequences of VTA D2 receptor dysregulation, this study investigated alterations in local cerebral glucose utilization throughout the brain following Drd2 knockdown in the VTA. Male Sprague-Dawley rats received bilateral injections of lentivirus encoding shRNAs against the rat dopamine D2 receptor, scrambled shRNA or phosphate buffered saline. The autoradiographic 2-[14C]deoxyglucose metabolic mapping procedure was conducted 22 days post-infection. Brains were sectioned for autoradiography and glucose utilization was measured across distinct regions throughout the brain. Local cerebral glucose utilization was found to be elevated in the Drd2 knockdown group as compared to control groups. These greater levels of metabolic activity following Drd2 knockdown in the VTA were observed not only in the mesocorticolimbic system and associated dopamine pathways, but also in a global pattern that included many areas with far less concentrated VTA dopamine inputs. This suggests that even a partial Drd2 deletion in the VTA can have widespread consequences and impact information flow in diverse networks that process sensory, cognitive, motor and emotional information.
Collapse
Affiliation(s)
- Tamriage A Martin
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Hilary R Smith
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Deborah J Luessen
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Rong Chen
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Linda J Porrino
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States.
| |
Collapse
|
31
|
Ahmadian SM, Alaei H, Ghahremani P. An Assessment between D1 Receptor Agonist and D2 receptor Antagonist into the Ventral Tegmental Area on Conditioned Place Preference and Locomotor Activity. Adv Biomed Res 2019; 8:72. [PMID: 32002395 PMCID: PMC6952765 DOI: 10.4103/abr.abr_82_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 05/22/2019] [Accepted: 05/31/2019] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND The release of dopamine (DA) has certain roles in the induction of conditioned place preference (CPP) and motor learning in the ventral tegmental area (VTA). The aim of this study was to investigate the excitatory effects of DA through DA-D1 agonist (SKF38393) and elimination of the inhibitory effects of DA through DA-D2 antagonist (eticlopride) into the VTA and its synergistic effects with an ineffective dose of morphine in the induction of CPP. MATERIALS AND METHODS Morphine (2.5 mg/kg; s. c.) did not induce a significant CPP, without any effect on the locomotor activity during the testing phase. SKF38393 (0.125, 0.5, and 1 μg/side) and eticlopride (0.5, 1, and 2 μg/side) individually or simultaneously were microinjected bilaterally into the VTA. RESULTS The administration of SKF38393 (1 and 2 μg/rat) with ineffective morphine and also without morphine caused CPP on test day, while eticlopride (2 μg/rat) caused CPP with morphine only. Locomotor activity increased in groups receiving D1 agonist and D2 antagonist that presumed to be caused by the reinforcing effect. In addition, the concurrent administration of ineffective doses of D1 agonist and D2 antagonist into the VTA with ineffective morphine caused CPP but not with saline. CONCLUSIONS This study showed that there was a need for morphine to activate the reward circuit through the D2 receptor in the VTA while the administration of the D1 agonist could independently activate the reward circuit. In addition, there was a probable synergistic effect using ineffective doses of D1 and D2 receptors, in the acquisition of morphine-induced CPP.
Collapse
Affiliation(s)
- Seyed Mostafa Ahmadian
- From the Department of Physiology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hojjatallah Alaei
- From the Department of Physiology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parisa Ghahremani
- From the Department of Physiology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
32
|
Folgueira C, Beiroa D, Porteiro B, Duquenne M, Puighermanal E, Fondevila MF, Barja-Fernández S, Gallego R, Hernández-Bautista R, Castelao C, Senra A, Seoane P, Gómez N, Aguiar P, Guallar D, Fidalgo M, Romero-Pico A, Adan R, Blouet C, Labandeira-García JL, Jeanrenaud F, Kallo I, Liposits Z, Salvador J, Prevot V, Dieguez C, Lopez M, Valjent E, Frühbeck G, Seoane LM, Nogueiras R. Hypothalamic dopamine signaling regulates brown fat thermogenesis. Nat Metab 2019; 1:811-829. [PMID: 31579887 PMCID: PMC6774781 DOI: 10.1038/s42255-019-0099-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dopamine signaling is a crucial part of the brain reward system and can affect feeding behavior. Dopamine receptors are also expressed in the hypothalamus, which is known to control energy metabolism in peripheral tissues. Here we show that pharmacological or chemogenetic stimulation of dopamine receptor 2 (D2R) expressing cells in the lateral hypothalamic area (LHA) and the zona incerta (ZI) decreases body weight and stimulates brown fat activity in rodents in a feeding-independent manner. LHA/ZI D2R stimulation requires an intact sympathetic nervous system and orexin system to exert its action and involves inhibition of PI3K in the LHA/ZI. We further demonstrate that, as early as 3 months after onset of treatment, patients treated with the D2R agonist cabergoline experience an increase in energy expenditure that persists for one year, leading to total body weight and fat loss through a prolactin-independent mechanism. Our results may provide a mechanistic explanation for how clinically used D2R agonists act in the CNS to regulate energy balance.
Collapse
Affiliation(s)
- Cintia Folgueira
- Grupo Fisiopatología Endocrina, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo. Hospitalario Universitario de Santiago (CHUS/SERGAS), Instituto de Investigación Sanitaria, Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Daniel Beiroa
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Begoña Porteiro
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Manon Duquenne
- Jean-Pierre Aubert Research Center (JPArc), Laboratory of Development and Plasticity of the Neuroendocrine Brain, Inserm UMR-S 1172, Lille, France
| | | | - Marcos F Fondevila
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Silvia Barja-Fernández
- Grupo Fisiopatología Endocrina, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo. Hospitalario Universitario de Santiago (CHUS/SERGAS), Instituto de Investigación Sanitaria, Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Rosalia Gallego
- Department of Morphological Sciences, School of Medicine, University of Santiago de Compostela, S. Francisco s/n, 15782 Santiago de Compostela (A Coruña), Spain
| | - René Hernández-Bautista
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
| | - Cecilia Castelao
- Grupo Fisiopatología Endocrina, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo. Hospitalario Universitario de Santiago (CHUS/SERGAS), Instituto de Investigación Sanitaria, Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Ana Senra
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
| | - Patricia Seoane
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Noemi Gómez
- Molecular Imaging Group, Department of Psychiatry, Radiology and Public Health, Faculty of Medicine Universidade de Santiago de Compostela (USC), Santiago de Compostela 15782 Spain; Molecular Imaging Group. Health Research Institute of Santiago de Compostela (IDIS). Travesía da Choupana s/n Santiago de Compostela. Zip Code: 15706. Spain; Nuclear Medicine Department University Clinical Hospital Santiago de Compostela (SERGAS) (CHUS), Travesía Choupana s/n. Santiago de Compostela 15706 Spain
| | - Pablo Aguiar
- Molecular Imaging Group, Department of Psychiatry, Radiology and Public Health, Faculty of Medicine Universidade de Santiago de Compostela (USC), Santiago de Compostela 15782 Spain; Molecular Imaging Group. Health Research Institute of Santiago de Compostela (IDIS). Travesía da Choupana s/n Santiago de Compostela. Zip Code: 15706. Spain; Nuclear Medicine Department University Clinical Hospital Santiago de Compostela (SERGAS) (CHUS), Travesía Choupana s/n. Santiago de Compostela 15706 Spain
| | - Diana Guallar
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
| | - Miguel Fidalgo
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
| | - Amparo Romero-Pico
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Roger Adan
- Brain Center Rudolf Magnus, Department of Neuroscience and Pharmacology, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Clemence Blouet
- MRC Metabolic Disease Unit. Institute of Metabolic Science. University of Cambridge, UK
| | - Jose Luís Labandeira-García
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
- Networking Research Center on Neurodegenerative Diseases, CIBERNED, Madrid, Spain
| | - Françoise Jeanrenaud
- Laboratory of Metabolism, Division of Endocrinology, Diabetology and Nutrition, Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Imre Kallo
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, HAS, 1083, Budapest, Hungary
| | - Zsolt Liposits
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, HAS, 1083, Budapest, Hungary
| | - Javier Salvador
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra & IdiSNA, Pamplona, Spain
| | - Vincent Prevot
- Jean-Pierre Aubert Research Center (JPArc), Laboratory of Development and Plasticity of the Neuroendocrine Brain, Inserm UMR-S 1172, Lille, France
| | - Carlos Dieguez
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Miguel Lopez
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Emmanuel Valjent
- IGF, Inserm, CNRS, Univ. Montpellier, F-34094 Montpellier, France
| | - Gema Frühbeck
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra & IdiSNA, Pamplona, Spain
| | - Luisa M Seoane
- Grupo Fisiopatología Endocrina, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo. Hospitalario Universitario de Santiago (CHUS/SERGAS), Instituto de Investigación Sanitaria, Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Ruben Nogueiras
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| |
Collapse
|
33
|
Addicott MA, Schechter JC, Sapyta JJ, Selig JP, Kollins SH, Weiss MD. Methylphenidate increases willingness to perform effort in adults with ADHD. Pharmacol Biochem Behav 2019; 183:14-21. [PMID: 31226260 DOI: 10.1016/j.pbb.2019.06.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/31/2019] [Accepted: 06/16/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND A reduced willingness to perform effort based on the magnitude and probability of potential rewards has been associated with diminished dopamine function and may be relevant to attention-deficit/hyperactivity disorder (ADHD). Here, we investigated the influence of ADHD status and methylphenidate on effort-based decisions. We hypothesized that ADHD participants would make fewer high-effort selections than non-ADHD subjects, and that methylphenidate would increase the number of high-effort selections. Furthermore, we hypothesized there would be associations among ADHD severity and methylphenidate-related changes in effort-based and attentional performance across all participants. METHODS AND PARTICIPANTS ADHD (n = 23) and non-ADHD (n = 23) adults completed the Effort Expenditure for Rewards Task in which participants select between low-effort and high-effort options to receive monetary rewards at varying levels of reward magnitude and probability. A test of attentional performance was also completed. RESULTS Overall, participants made more high-effort selections as potential reward magnitude and probability increased. ADHD participants did not make fewer high-effort selections than non-ADHD participants, but ADHD participants showed greater methylphenidate-related increases in high-effort selections. ADHD participants had worse attentional performance than non-ADHD participants. ADHD severity was associated with methylphenidate-related changes in high-effort selections, but not changes in attentional performance. CONCLUSIONS These results indicate that methylphenidate increases the willingness to perform effort in individuals with ADHD, possibly due to disorder-related motivational deficits. This provides support for theories of insufficient effort allocation among individuals with ADHD. TRIAL REGISTRATION Clinicaltrials.gov Identifier, NCT02630017.
Collapse
Affiliation(s)
| | | | | | - James P Selig
- University of Arkansas for Medical Sciences, United States of America
| | - Scott H Kollins
- Duke University School of Medicine, United States of America
| | - Margaret D Weiss
- University of Arkansas for Medical Sciences, United States of America
| |
Collapse
|
34
|
Reward Processing under Chronic Pain from the Perspective of "Liking" and "Wanting": A Narrative Review. Pain Res Manag 2019; 2019:6760121. [PMID: 31149319 PMCID: PMC6501242 DOI: 10.1155/2019/6760121] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/06/2019] [Accepted: 04/04/2019] [Indexed: 11/29/2022]
Abstract
The therapeutic goals of patients with chronic pain are not only to relieve pain but also to improve the quality of life. Chronic pain negatively affects various aspects of daily life, such as by decreasing the motivation to work and reward sensitivity, which may lead to difficulties in daily life or even unemployment. Human and animal studies have shown that chronic pain damages reward processing; the exploration of associated internal mechanisms may aid the development of treatments to repair this damage. Incentive salience theory, used widely to describe reward processing, divides this processing into “liking” (reward-induced hedonic sensory impact) and “wanting” (reward-induced motivation) components. It has been employed to explain pathological changes in reward processing induced by psychiatric disorders. In this review, we summarize the findings of studies of reward processing under chronic pain and examine the effects of chronic pain on “liking” and “wanting.” Evidence indicates that chronic pain compromises the “wanting” component of reward processing; we also discuss the neural mechanisms that may mediate this effect. We hope that this review aids the development of therapies to improve the quality of life of patients with chronic pain.
Collapse
|
35
|
Yohn SE, Galbraith J, Calipari ES, Conn PJ. Shared Behavioral and Neurocircuitry Disruptions in Drug Addiction, Obesity, and Binge Eating Disorder: Focus on Group I mGluRs in the Mesolimbic Dopamine Pathway. ACS Chem Neurosci 2019; 10:2125-2143. [PMID: 30933466 PMCID: PMC7898461 DOI: 10.1021/acschemneuro.8b00601] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Accumulated data from clinical and preclinical studies suggest that, in drug addiction and states of overeating, such as obesity and binge eating disorder (BED), there is an imbalance in circuits that are critical for motivation, reward saliency, executive function, and self-control. Central to these pathologies and the extensive topic of this Review are the aberrations in dopamine (DA) and glutamate (Glu) within the mesolimbic pathway. Group I metabotropic glutamate receptors (mGlus) are highly expressed in the mesolimbic pathway and are poised in key positions to modulate disruptions in synaptic plasticity and neurotransmitter release observed in drug addiction, obesity, and BED. The use of allosteric modulators of group I mGlus has been studied in drug addiction, as they offer several advantages over traditional orthosteric agents. However, they have yet to be studied in obesity or BED. With the substantial overlap between the neurocircuitry involved in drug addiction and eating disorders, group I mGlus may also provide novel targets for obesity and BED.
Collapse
Affiliation(s)
- Samantha E. Yohn
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, United States
| | - Jordan Galbraith
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, United States
| | - Erin S. Calipari
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, United States
| | - P. Jeffrey Conn
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, United States
| |
Collapse
|
36
|
GIRK Channel Activity in Dopamine Neurons of the Ventral Tegmental Area Bidirectionally Regulates Behavioral Sensitivity to Cocaine. J Neurosci 2019; 39:3600-3610. [PMID: 30837265 DOI: 10.1523/jneurosci.3101-18.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/17/2019] [Accepted: 02/22/2019] [Indexed: 12/19/2022] Open
Abstract
Dopamine (DA) neurons of the VTA have been widely implicated in the cellular and behavioral responses to drugs of abuse. Inhibitory G protein signaling mediated by GABAB receptors (GABABRs) and D2 DA receptors (D2Rs) regulates the excitability of VTA DA neurons, DA neurotransmission, and behaviors modulated by DA. Most of the somatodendritic inhibitory effect of GABABR and D2R activation on DA neurons reflects the activation of G protein-gated inwardly rectifying K+ (GIRK) channels. Furthermore, GIRK-dependent signaling in VTA DA neurons can be weakened by exposure to psychostimulants and strengthened by phasic DA neuron firing. The objective of this study was to determine how the strength of GIRK channel activity in VTA DA neurons influences sensitivity to cocaine. We used a Cre-dependent viral strategy to overexpress the individual GIRK channel subunits in VTA DA neurons of male and female adult mice, leading to enhancement (GIRK2) or suppression (GIRK3) of GIRK channel activity. Overexpression of GIRK3 decreased somatodendritic GABABR- and D2R-dependent signaling and increased cocaine-induced locomotor activity, whereas overexpression of GIRK2 increased GABABR-dependent signaling and decreased cocaine-induced locomotion. Neither manipulation impacted anxiety- or depression-related behavior, despite the link between such behaviors and DA signaling. Together, these data show that behavioral sensitivity to cocaine in mice is inversely proportional to the strength of GIRK channel activity in VTA DA neurons and suggest that direct activators of the unique VTA DA neuron GIRK channel subtype (GIRK2/GIRK3 heteromer) could represent a promising therapeutic target for treatment of addiction.SIGNIFICANCE STATEMENT Inhibitory G protein signaling in dopamine (DA) neurons, including that mediated by G protein-gated inwardly rectifying K+ (GIRK) channels, has been implicated in behavioral sensitivity to cocaine. Here, we used a viral approach to bidirectionally manipulate GIRK channel activity in DA neurons of the VTA. We found that decreasing GIRK channel activity in VTA DA neurons increased behavioral sensitivity to cocaine, whereas increasing GIRK channel activity decreased behavioral sensitivity to cocaine. These manipulations did not alter anxiety- or depression-related behaviors. These data highlight the unique GIRK channel subtype in VTA DA neurons as a possible therapeutic target for addiction.
Collapse
|
37
|
Sebold M, Spitta G, Gleich T, Dembler-Stamm T, Butler O, Zacharias K, Aydin S, Garbusow M, Rapp M, Schubert F, Buchert R, Gallinat J, Heinz A. Stressful life events are associated with striatal dopamine receptor availability in alcohol dependence. J Neural Transm (Vienna) 2019; 126:1127-1134. [DOI: 10.1007/s00702-019-01985-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 02/06/2019] [Indexed: 11/30/2022]
|
38
|
Gallo EF. Disentangling the diverse roles of dopamine D2 receptors in striatal function and behavior. Neurochem Int 2019; 125:35-46. [PMID: 30716356 DOI: 10.1016/j.neuint.2019.01.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/21/2019] [Accepted: 01/27/2019] [Indexed: 02/07/2023]
Abstract
Dopamine D2 receptors (D2Rs) mediate many of the actions of dopamine in the striatum, ranging from movement to the effortful pursuit of reward. Yet despite significant advances in linking D2Rs to striatal functions with pharmacological and genetic strategies in animals, how dopamine orchestrates its myriad actions on different cell populations -each expressing D2Rs- remains unclear. Furthermore, brain imaging and genetic studies in humans have consistently associated striatal D2R alterations with various neurological and neuropsychiatric disorders, but how and which D2Rs are involved in each case is poorly understood. Therefore, a critical first step is to engage in a refined and systematic investigation of the impact of D2R function on specific striatal cells, circuits, and behaviors. Here, I will review recent efforts, primarily in animal models, aimed at unlocking the complex and heterogeneous roles of D2Rs in striatum.
Collapse
Affiliation(s)
- Eduardo F Gallo
- Department of Biological Sciences, Fordham University, Bronx, NY, USA.
| |
Collapse
|
39
|
Modelling Differential Vulnerability to Substance Use Disorder in Rodents: Neurobiological Mechanisms. Handb Exp Pharmacol 2019; 258:203-230. [PMID: 31707470 DOI: 10.1007/164_2019_300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Despite the prevalence of drug use within society, only a subset of individuals actively taking addictive drugs lose control over their intake and develop compulsive drug-seeking and intake that typifies substance use disorder (SUD). Although research in this field continues to be an important and dynamic discipline, the specific neuroadaptations that drive compulsive behaviour in humans addicted to drugs and the neurobiological mechanisms that underlie an individual's innate susceptibility to SUD remain surprisingly poorly understood. Nonetheless, it is clear from research within the clinical domain that some behavioural traits are recurrently co-expressed in individuals with SUD, thereby inviting the hypothesis that certain behavioural endophenotypes may be predictive, or at least act in some way, to modify an individual's probability for developing this disorder. The analysis of such endophenotypes and their catalytic relationship to the expression of addiction-related behaviours has been greatly augmented by experimental approaches in rodents that attempt to capture diagnostically relevant aspects of this progressive brain disorder. This work has evolved from an early focus on aberrant drug reinforcement mechanisms to a now much richer account of the putatively impaired cognitive control processes that ultimately determine individual trajectories to compulsive drug-related behaviours. In this chapter we discuss the utility of experimental approaches in rodents designed to elucidate the neurobiological and genetic underpinnings of so-called risk traits and how these innate vulnerabilities collectively contribute to the pathogenesis of SUD.
Collapse
|
40
|
Alsiö J, Phillips BU, Sala-Bayo J, Nilsson SRO, Calafat-Pla TC, Rizwand A, Plumbridge JM, López-Cruz L, Dalley JW, Cardinal RN, Mar AC, Robbins TW. Dopamine D2-like receptor stimulation blocks negative feedback in visual and spatial reversal learning in the rat: behavioural and computational evidence. Psychopharmacology (Berl) 2019; 236:2307-2323. [PMID: 31218428 PMCID: PMC6695374 DOI: 10.1007/s00213-019-05296-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 06/02/2019] [Indexed: 02/02/2023]
Abstract
RATIONALE Dopamine D2-like receptors (D2R) are important drug targets in schizophrenia and Parkinson's disease, but D2R ligands also cause cognitive inflexibility such as poor reversal learning. The specific role of D2R in reversal learning remains unclear. OBJECTIVES We tested the hypotheses that D2R agonism impairs reversal learning by blocking negative feedback and that antagonism of D1-like receptors (D1R) impairs learning from positive feedback. METHODS Male Lister Hooded rats were trained on a novel visual reversal learning task. Performance on "probe trials", during which the correct or incorrect stimulus was presented with a third, probabilistically rewarded (50% of trials) and therefore intermediate stimulus, revealed individual learning curves for the processes of positive and negative feedback. The effects of D2R and D1R agonists and antagonists were evaluated. A separate cohort was tested on a spatial probabilistic reversal learning (PRL) task after D2R agonism. Computational reinforcement learning modelling was applied to choice data from the PRL task to evaluate the contribution of latent factors. RESULTS D2R agonism with quinpirole dose-dependently impaired both visual reversal and PRL. Analysis of the probe trials on the visual task revealed a complete blockade of learning from negative feedback at the 0.25 mg/kg dose, while learning from positive feedback was intact. Estimated parameters from the model that best described the PRL choice data revealed a steep and selective decrease in learning rate from losses. D1R antagonism had a transient effect on the positive probe trials. CONCLUSIONS D2R stimulation impairs reversal learning by blocking the impact of negative feedback.
Collapse
Affiliation(s)
- Johan Alsiö
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK.
| | - Benjamin U Phillips
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Júlia Sala-Bayo
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Simon R O Nilsson
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
- Neuroscience Institute, New York University Medical Center, New York, NY, USA
- Department of Neuroscience and Physiology, School of Medicine, New York University, New York, NY, USA
| | - Teresa C Calafat-Pla
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Arazo Rizwand
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Jessica M Plumbridge
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Laura López-Cruz
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Jeffrey W Dalley
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Rudolf N Cardinal
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Cambridgeshire & Peterborough NHS Foundation Trust, Box 190 (Liaison Psychiatry), Cambridge Biomedical Campus, Cambridge, UK
| | - Adam C Mar
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
- Neuroscience Institute, New York University Medical Center, New York, NY, USA
- Department of Neuroscience and Physiology, School of Medicine, New York University, New York, NY, USA
| | - Trevor W Robbins
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
41
|
Phillips BU, Lopez-Cruz L, Hailwood J, Heath CJ, Saksida LM, Bussey TJ. Translational approaches to evaluating motivation in laboratory rodents: conventional and touchscreen-based procedures. Curr Opin Behav Sci 2018. [DOI: 10.1016/j.cobeha.2017.12.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
42
|
Karlsson RM, Wang AS, Sonti AN, Cameron HA. Adult neurogenesis affects motivation to obtain weak, but not strong, reward in operant tasks. Hippocampus 2018; 28:512-522. [PMID: 29663595 DOI: 10.1002/hipo.22950] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 03/27/2018] [Accepted: 04/05/2018] [Indexed: 12/17/2022]
Abstract
Decreased motivation to seek rewards is a key feature of mood disorders that correlates with severity and treatment outcome. This anhedonia, or apathy, likely reflects impairment in reward circuitry, but the specific neuronal populations controlling motivation are unclear. Granule neurons generated in the adult hippocampus have been implicated in mood disorders, but are not generally considered as part of reward circuits. We investigated a possible role of these new neurons in motivation to work for food and sucrose rewards in operant conditioning tasks using GFAP-TK pharmacogenetic ablation of adult neurogenesis in both rats and mice. Rats and mice lacking adult neurogenesis showed normal lever press responding during fixed ratio training, reward devaluation, and Pavlovian Instrumental Transfer, suggesting no impairment in learning. However, on an exponentially progressive ratio schedule, or when regular chow was freely available in the testing chamber, TK rats and mice showed less effort to gain sucrose tablets. When working for balanced food tablets, which rats and mice of both genotypes strongly preferred over sucrose, the genotype effects on behavior were lost. This decrease in effort under conditions of low reward suggests that loss of adult neurogenesis decreases motivation to seek reward in a manner that may model behavioral apathy.
Collapse
Affiliation(s)
- Rose-Marie Karlsson
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, 20892
| | - Alice S Wang
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, 20892
| | - Anup N Sonti
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, 20892
| | - Heather A Cameron
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, 20892
| |
Collapse
|
43
|
Abstract
Neuroimaging studies in animal models and human subjects have each revealed that relatively low striatal dopamine D2-like receptor binding potential is associated with poor impulse control and with vulnerability for addiction-related behaviors. These studies cannot, however, disambiguate the roles for various pools of D2 receptors found in the striatum (e.g., those expressed on medium spiny striato-pallidal neurons vs on dopamine-releasing nerve terminals) in these behavioral outcomes. To clarify the role of the latter pool, namely, D2 autoreceptors, we studied mice carrying a conditional DRD2 gene, with or without Cre-recombinase expressed under the transcriptional control of the dopamine transporter gene locus (autoDrd2-KO, n = 19 and controls, n = 21). These mice were tested for locomotor response to cocaine, and spatial reversal learning was assessed in operant conditioning chambers. As predicted, compared to control mice, autoDrd2-KO animals demonstrated heightened sensitivity to the locomotor stimulating effect of cocaine (10 mg/kg, i.p.), confirming previous research using a similar genetic model. In the spatial reversal learning task, autoDrd2-KO mice were slower to reach a learning criterion and had difficulty sustaining a prolonged nose poke response, measurements conceptually related to impaired response inhibition. Rate of learning of the initial discrimination and latencies to collect rewards, to initiate trials and to produce a response were unaffected by genetic deletion of D2 autoreceptors, discarding possible motor and motivational factors. Together, these findings confirm the role of D2 autoreceptors in reversal learning and suggest a broader involvement in behavioral inhibition mechanisms.
Collapse
|
44
|
McLaughlin T, Blum K, Steinberg B, Modestino EJ, Fried L, Baron D, Siwicki D, Braverman ER, Badgaiyan RD. Pro-dopamine regulator, KB220Z, attenuates hoarding and shopping behavior in a female, diagnosed with SUD and ADHD. J Behav Addict 2018; 7:192-203. [PMID: 29316800 PMCID: PMC6035027 DOI: 10.1556/2006.6.2017.081] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Background Addictive-like behaviors (e.g., hoarding and shopping) may be the result of the cumulative effects of dopaminergic and other neurotransmitter genetic variants as well as elevated stress levels. We, therefore, propose that dopamine homeostasis may be the preferred goal in combating such challenging and unwanted behaviors, when simple dopaminergic activation through potent agonists may not provide any resolution. Case presentation C.J. is a 38-year-old, single, female, living with her mother. She has a history of substance use disorder as well as attention deficit hyperactivity disorder, inattentive type. She had been stable on buprenorphine/naloxone combination and amphetamine, dextroamphetamine mixed salts for many years when unexpectedly she lost her job for oversleeping and not calling into work. KB200z (a pro-dopamine compound) was added to her regimen for complaints of low drive and motivation. After taking this nutraceutical for 4 weeks, she noticed a marked improvement in her mental status and many behaviors. She noted that her shopping and hoarding addictions had appreciably decreased. Furthermore, her lifelong history of terrifying lucid dreams was eliminated. Finally, she felt more in control; her locus of control shifted from external to more internal. Discussion The hypothesis is that C.J.'s reported, behavioral, and psychological benefits resulted from the pro-dopamine-regulating effect of KB220Z across the brain reward system. Conclusions This effect, we surmise, could be the result of a new dopamine balance, across C.J.'s brain reward system. Dopamine homeostasis is an effect of KB220Z seen in both animal and human placebo-controlled fMRI experiments.
Collapse
Affiliation(s)
- Thomas McLaughlin
- 1 Department of Psychopharmacology, Center for Psychiatric Medicine , Lawrence, MA, USA
| | - Kenneth Blum
- 2 Department of Psychiatry, Boonshoft School of Medicine, Dayton VA Medical Center, Wright State University , Dayton, OH, USA
- 3 Department of Psychiatry, McKnight Brain Institute, University of Florida College of Medicine , Gainesville, FL, USA
- 4 Department of Psychiatry and Behavioral Sciences, Keck Medicine University of Southern California , Los Angeles, CA, USA
- 5 Division of Applied Clinical Research & Education, Dominion Diagnostics, LLC , North Kingstown, RI, USA
- 6 Department of Precision Medicine, Geneus Health LLC , San Antonio, TX, USA
- 7 Department of Addiction Research & Therapy, Nupathways Inc. , Innsbrook, MO, USA
- 8 Department of Clinical Neurology, Path Foundation , New York, NY, USA
- 9 Division of Neuroscience Based Addiction Therapy, The Shores Treatment & Recovery Center , Port Saint Lucie, FL, USA
- 10 Institute of Psychology, Eötvös Loránd University , Budapest, Hungary
| | - Bruce Steinberg
- 11 Department of Psychology, Curry College , Milton, MA, USA
| | | | - Lyle Fried
- 9 Division of Neuroscience Based Addiction Therapy, The Shores Treatment & Recovery Center , Port Saint Lucie, FL, USA
| | - David Baron
- 4 Department of Psychiatry and Behavioral Sciences, Keck Medicine University of Southern California , Los Angeles, CA, USA
| | - David Siwicki
- 6 Department of Precision Medicine, Geneus Health LLC , San Antonio, TX, USA
| | - Eric R Braverman
- 8 Department of Clinical Neurology, Path Foundation , New York, NY, USA
| | | |
Collapse
|
45
|
Leigh SJ, Lee F, Morris MJ. Hyperpalatability and the Generation of Obesity: Roles of Environment, Stress Exposure and Individual Difference. Curr Obes Rep 2018; 7:6-18. [PMID: 29435959 DOI: 10.1007/s13679-018-0292-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE OF REVIEW This review investigates how exposure to palatable food and its associated cues alters appetite regulation and feeding behaviour to drive overeating and weight gain. RECENT FINDINGS Both supraphysiological and physiological feeding systems are affected by exposure to palatable foods and its associated cues. Preclinical research, largely using rodents, has demonstrated that palatable food modulates feeding-related neural systems and food-seeking behaviour by recruiting the mesolimbic reward pathway. This is supported by studies in adolescents which have shown that mesolimbic activity in response to palatable food cues and consumption predicts future weight gain. Additionally, stress exposure, environmental factors and individual susceptibility have been shown to modulate the effects of highly palatable foods on behaviour. Further preclinical research using free-choice diets modelling the modern obesogenic environment is needed to identify how palatable foods drive overeating. Moreover, future clinical research would benefit from more appropriate quantification of palatability, making use of rating systems and surveys.
Collapse
Affiliation(s)
- Sarah-Jane Leigh
- Department of Pharmacology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Frances Lee
- Department of Pharmacology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Margaret J Morris
- Department of Pharmacology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia.
| |
Collapse
|
46
|
Chen R, McIntosh S, Hemby SE, Sun H, Sexton T, Martin TJ, Childers SR. High and low doses of cocaine intake are differentially regulated by dopamine D2 receptors in the ventral tegmental area and the nucleus accumbens. Neurosci Lett 2018; 671:133-139. [PMID: 29454035 DOI: 10.1016/j.neulet.2018.02.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/01/2018] [Accepted: 02/12/2018] [Indexed: 01/11/2023]
Abstract
Dopamine D2 receptors (D2Rs) in the ventral tegmental area (VTA) and the nucleus accumbens (NAc) are associated with vulnerability to addiction; however, whether D2Rs in these two brain regions play differential roles in regulation of drug intake is unknown. Here, we compared the effect of decreased mRNA level of Drd2 in each region on cocaine self-administration in a dose-response function. Drd2 mRNA levels in rat VTA or NAc were knocked down by bilateral microinjection of lentivirus coding shRNAs against rat Drd2 or scrambled shRNA. Drd2 knockdown was persistent and stable between 20 and 90 days after lentiviral infection. Animals were trained to self-administer cocaine 20 days after Drd2 shRNA treatment. Compared to scrambled shRNA treated rats, Drd2 knockdown in the VTA increased cocaine self-administration at all tested doses (0.02-0.56 mg/kg/infusion) producing an upward shift (both the ascending and descending limb) in the dose-response curve of cocaine self-administration. In contrast, intra-NAc knockdown increased cocaine self-administration only on the ascending limb of the dose-response curve (0.02-0.07 mg/kg/infusion). These data suggest that D2Rs in the VTA, not in the NAc, regulate high-dose cocaine intake. The present study not only demonstrates that low levels of D2Rs in either region increase low doses of cocaine intake, but also reveals for the first time their dissociable roles in limiting high doses of cocaine self-administration.
Collapse
Affiliation(s)
- R Chen
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, United States; Center for the Neurobiology of Addiction Treatment, Wake Forest School of Medicine, Winston Salem, NC 27157, United States; Center for Molecular Signaling, Wake Forest University, Winston Salem, NC 27109, United States.
| | - S McIntosh
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, United States
| | - S E Hemby
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, United States
| | - H Sun
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, United States; Center for the Neurobiology of Addiction Treatment, Wake Forest School of Medicine, Winston Salem, NC 27157, United States
| | - T Sexton
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, United States; Center for the Neurobiology of Addiction Treatment, Wake Forest School of Medicine, Winston Salem, NC 27157, United States
| | - T J Martin
- Center for the Neurobiology of Addiction Treatment, Wake Forest School of Medicine, Winston Salem, NC 27157, United States; Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, NC 27157, United States
| | - S R Childers
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, United States; Center for the Neurobiology of Addiction Treatment, Wake Forest School of Medicine, Winston Salem, NC 27157, United States
| |
Collapse
|
47
|
Boekhoudt L, Wijbrans EC, Man JHK, Luijendijk MCM, de Jong JW, van der Plasse G, Vanderschuren LJMJ, Adan RAH. Enhancing excitability of dopamine neurons promotes motivational behaviour through increased action initiation. Eur Neuropsychopharmacol 2018; 28:171-184. [PMID: 29153928 DOI: 10.1016/j.euroneuro.2017.11.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 08/28/2017] [Accepted: 11/03/2017] [Indexed: 01/05/2023]
Abstract
Motivational deficits are a key symptom in multiple psychiatric disorders, including major depressive disorder, schizophrenia and addiction. A likely neural substrate for these motivational deficits is the brain dopamine (DA) system. In particular, DA signalling in the nucleus accumbens, which originates from DA neurons in the ventral tegmental area (VTA), has been identified as a crucial substrate for effort-related and activational aspects of motivation. Unravelling how VTA DA neuronal activity relates to motivational behaviours is required to understand how motivational deficits in psychiatry can be specifically targeted. In this study, we therefore used designer receptors exclusively activated by designer drugs (DREADD) in TH:Cre rats, in order to determine the effects of chemogenetic DA neuron activation on different aspects of motivational behaviour. We found that chemogenetic activation of DA neurons in the VTA, but not substantia nigra, significantly increased responding for sucrose under a progressive ratio schedule of reinforcement. More specifically, high effort exertion was characterized by increased initiations of reward-seeking actions. This effect was dependent on effort requirements and instrumental contingencies, but was not affected by sucrose pre-feeding. Together, these findings indicate that VTA DA neuronal activation drives motivational behaviour by facilitating action initiation. With this study, we show that enhancing excitability of VTA DA neurons is a viable strategy to improve motivational behaviour.
Collapse
Affiliation(s)
- Linde Boekhoudt
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Centre Utrecht, Universiteitsweg 100, 3585 CG Utrecht, The Netherlands
| | - Ellen C Wijbrans
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Centre Utrecht, Universiteitsweg 100, 3585 CG Utrecht, The Netherlands
| | - Jodie H K Man
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Centre Utrecht, Universiteitsweg 100, 3585 CG Utrecht, The Netherlands
| | - Mieneke C M Luijendijk
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Centre Utrecht, Universiteitsweg 100, 3585 CG Utrecht, The Netherlands
| | - Johannes W de Jong
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Centre Utrecht, Universiteitsweg 100, 3585 CG Utrecht, The Netherlands; Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Geoffrey van der Plasse
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Centre Utrecht, Universiteitsweg 100, 3585 CG Utrecht, The Netherlands
| | - Louk J M J Vanderschuren
- Department of Animals in Science and Society, Division of Behavioural Neuroscience, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Roger A H Adan
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Centre Utrecht, Universiteitsweg 100, 3585 CG Utrecht, The Netherlands.
| |
Collapse
|
48
|
Diamantopoulou A, Sun Z, Mukai J, Xu B, Fenelon K, Karayiorgou M, Gogos JA. Loss-of-function mutation in Mirta22/Emc10 rescues specific schizophrenia-related phenotypes in a mouse model of the 22q11.2 deletion. Proc Natl Acad Sci U S A 2017; 114:E6127-E6136. [PMID: 28696314 PMCID: PMC5544257 DOI: 10.1073/pnas.1615719114] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Identification of protective loss-of-function (LoF) mutations holds great promise for devising novel therapeutic interventions, although it faces challenges due to the scarcity of protective LoF alleles in the human genome. Exploiting the detailed mechanistic characterization of animal models of validated disease mutations offers an alternative. Here, we provide insights into protective-variant biology based on our characterization of a model of the 22q11.2 deletion, a strong genetic risk factor for schizophrenia (SCZ). Postnatal brain up-regulation of Mirta22/Emc10, an inhibitor of neuronal maturation, represents the major transcriptional effect of the 22q11.2-associated microRNA dysregulation. Here, we demonstrate that mice in which the Df(16)A deficiency is combined with a LoF Mirta22 allele show rescue of key SCZ-related deficits, namely prepulse inhibition decrease, working memory impairment, and social memory deficits, as well as synaptic and structural plasticity abnormalities in the prefrontal cortex. Additional analysis of homozygous Mirta22 knockout mice, in which no alteration is observed in the above-mentioned SCZ-related phenotypes, highlights the deleterious effects of Mirta22 up-regulation. Our results support a causal link between dysregulation of a miRNA target and SCZ-related deficits and provide key insights into beneficial LoF mutations and potential new treatments.
Collapse
Affiliation(s)
- Anastasia Diamantopoulou
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY 10032
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Ziyi Sun
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Jun Mukai
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Bin Xu
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Karine Fenelon
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Maria Karayiorgou
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY 10032;
| | - Joseph A Gogos
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032;
- Department of Neuroscience, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| |
Collapse
|
49
|
Karim TJ, Reyes-Vazquez C, Dafny N. Comparison of the VTA and LC response to methylphenidate: a concomitant behavioral and neuronal study of adolescent male rats. J Neurophysiol 2017; 118:1501-1514. [PMID: 28615331 DOI: 10.1152/jn.00145.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 06/12/2017] [Accepted: 06/12/2017] [Indexed: 12/21/2022] Open
Abstract
Methylphenidate (MPD), also known as Ritalin, is a psychostimulant used to treat attention deficit hyperactivity disorder. However, it is increasingly being misused by normal adolescents for recreation and academic advantage. Therefore, it is important to elucidate the behavioral and neurophysiological effects of MPD in normal subjects. MPD inhibits the reuptake of catecholamines, mainly found in the ventral tegmental area (VTA) and locus coeruleus (LC). The VTA and LC normally mediate attention, motivation, and drug reward behaviors. Selective neuronal connections between the VTA and LC have been identified implicating regular interaction between the structures. The objective of this study was to compare the neuronal responses of the VTA and LC to MPD in normal adolescent rats. Animals were implanted with permanent electrodes in the VTA and LC, and neuronal units were recorded following acute and repetitive (chronic) saline or 0.6, 2.5, or 10.0 mg/kg MPD exposure. Animals displayed either behavioral sensitization or tolerance to all three doses of MPD. Acute MPD exposure elicited excitation in the majority of all VTA and LC units. Chronic MPD exposure elicited a further increase in VTA and LC neuronal activity in animals exhibiting behavioral sensitization and an attenuation in VTA and LC neuronal activity in animals exhibiting behavioral tolerance, demonstrating neurophysiological sensitization and tolerance, respectively. The similar pattern in VTA and LC unit activity suggests that the two structures are linked in their response to MPD. These results may help determine the exact mechanism of action of MPD, resulting in optimized treatment of patients.NEW & NOTEWORTHY The same dose of 0.6, 2.5, and 10 mg/kg methylphenidate (MPD) elicits either behavioral sensitization or tolerance in adolescent rats. There is a direct correlation between the ventral tegmental area (VTA) and locus coeruleus (LC) neuronal response to chronic MPD exposure. Both the VTA and LC are involved in the behavioral and neurophysiological effects of chronic MPD.
Collapse
Affiliation(s)
- Tahseen J Karim
- Department of Neurobiology and Anatomy, McGovern Medical School, Houston, Texas
| | - Cruz Reyes-Vazquez
- Department of Neurobiology and Anatomy, McGovern Medical School, Houston, Texas
| | - Nachum Dafny
- Department of Neurobiology and Anatomy, McGovern Medical School, Houston, Texas
| |
Collapse
|
50
|
Dobbs LK, Lemos JC, Alvarez VA. Restructuring of basal ganglia circuitry and associated behaviors triggered by low striatal D2 receptor expression: implications for substance use disorders. GENES BRAIN AND BEHAVIOR 2017; 16:56-70. [PMID: 27860248 PMCID: PMC5243158 DOI: 10.1111/gbb.12361] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 11/14/2016] [Accepted: 11/14/2016] [Indexed: 01/11/2023]
Abstract
Dopamine D2 receptors (D2Rs) consistently emerge as a critical substrate for the etiology of some major psychiatric disorders. Indeed, a central theory of substance use disorders (SUDs) postulates that a reduction in D2R levels in the striatum is a determining factor that confers vulnerability to abuse substances. A large number of clinical and preclinical studies strongly support this link between SUDs and D2Rs; however, identifying the mechanism by which low D2Rs facilitate SUDs has been hindered by the complexity of circuit connectivity, the heterogeneity of D2R expression and the multifaceted constellation of phenotypes observed in SUD patient. Animal models are well‐suited for understanding the mechanisms because they allow access to the circuitry and the genetic tools that enable a dissection of the D2R heterogeneity. This review discusses recent findings on the functional role of D2Rs and highlights the distinctive contributions of D2Rs expressed on specific neuronal subpopulations to the behavioral responses to stimulant drugs. A circuit‐wide restructuring of local and long‐range inhibitory connectivity within the basal ganglia is observed in response to manipulation of striatal D2R levels and is accompanied by multiple alterations in dopamine‐dependent behaviors. Collectively, these new findings provide compelling evidence for a critical role of striatal D2Rs in shaping basal ganglia connectivity; even among neurons that do not express D2Rs. These findings from animal models have deep clinical implications for SUD patients with low levels D2R availability where a similar restructuring of basal ganglia circuitry is expected to take place.
Collapse
Affiliation(s)
- L K Dobbs
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - J C Lemos
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - V A Alvarez
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|