1
|
Yan S, Huang N, Tong Y, Shu Y, Le Q, Ta D, Xu K. Functional Ultrasound Imaging of Cocaine Induced Brain-Wide Neurovascular Response. Neuroimage 2025; 309:121085. [PMID: 39952487 DOI: 10.1016/j.neuroimage.2025.121085] [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: 07/18/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025] Open
Abstract
Extensive studies have reported that cocaine can lead to potent reduction in cerebral blood flow. However, the mechanisms of the cocaine's impact on the neural and vascular system of brain in temporal and spatial aspects remain elusive. Functional ultrasound (fUS) is a novel neurovascular imaging modality acclaimed for its deep penetration, superior spatiotemporal resolution, and high sensitivity to small blood flow dynamics. This study aims to use fUS technique to characterize the regional differences in hemodynamic responses to acute cocaine administration. The CBV responses revealed that the cortex and ventral tegmental area (VTA) were the regions most significantly affected by cocaine. In addition, electroencephalography (EEG) was also utilized to assess the neural activities in the cortex and VTA. In the cortex, the observed CBV changes responded more rapidly to cocaine than local field potential (LFP) activities, indicating that prior to acting on the central nervous system, cocaine may first affect the peripheral nervous system, accelerating heart rate and increasing cardiac output. Both hemodynamic and neural responses showed opposing patterns between cortical and VTA brain regions. Based on these observations, we proposed a two-stage hypothesis to explain acute cocaine's multifaceted impact on the brain. This study underscores the efficacy of fUS as a powerful and sensitive tool for tracking cocaine-induced hemodynamic changes and enhances our understanding of cocaine's effects on the neurovascular system.
Collapse
Affiliation(s)
- Shaoyuan Yan
- Department of Biomedical Engineering, Fudan University, Shanghai 200438, China
| | - Nan Huang
- School of Basic Medical Sciences, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Yusheng Tong
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yousheng Shu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China; Department of Neurology, Huashan Hospital, Institute for Translational Brain Research, Fudan University, Shanghai 200032, China
| | - Qiumin Le
- School of Basic Medical Sciences, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Dean Ta
- Department of Biomedical Engineering, Fudan University, Shanghai 200438, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China.
| | - Kailiang Xu
- Department of Biomedical Engineering, Fudan University, Shanghai 200438, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China; Poda Medical Technology Co., Ltd., Shanghai 200433, China.
| |
Collapse
|
2
|
Zhu Z, Gong R, Rodriguez V, Quach KT, Chen X, Sternson SM. Hedonic eating is controlled by dopamine neurons that oppose GLP-1R satiety. Science 2025; 387:eadt0773. [PMID: 40146831 PMCID: PMC12009138 DOI: 10.1126/science.adt0773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 01/27/2025] [Indexed: 03/29/2025]
Abstract
Hedonic eating is defined as food consumption driven by palatability without physiological need. However, neural control of palatable food intake is poorly understood. We discovered that hedonic eating is controlled by a neural pathway from the peri-locus ceruleus to the ventral tegmental area (VTA). Using photometry-calibrated optogenetics, we found that VTA dopamine (VTADA) neurons encode palatability to bidirectionally regulate hedonic food consumption. VTADA neuron responsiveness was suppressed during food consumption by semaglutide, a glucagon-like peptide receptor 1 (GLP-1R) agonist used as an antiobesity drug. Mice recovered palatable food appetite and VTADA neuron activity during repeated semaglutide treatment, which was reversed by consumption-triggered VTADA neuron inhibition. Thus, hedonic food intake activates VTADA neurons, which sustain further consumption, a mechanism that opposes appetite reduction by semaglutide.
Collapse
Affiliation(s)
- Zhenggang Zhu
- Department of Neurosciences, University of California, San Diego; La Jolla, CA 92093, USA
| | - Rong Gong
- Janelia Research Campus, Howard Hughes Medical Institute; Ashburn, VA 20147, USA
| | - Vicente Rodriguez
- Howard Hughes Medical Institute; University of California, San Diego; La Jolla, CA 92093, USA
| | - Kathleen T. Quach
- Howard Hughes Medical Institute; University of California, San Diego; La Jolla, CA 92093, USA
| | - Xinyu Chen
- Howard Hughes Medical Institute; University of California, San Diego; La Jolla, CA 92093, USA
| | - Scott M. Sternson
- Department of Neurosciences, University of California, San Diego; La Jolla, CA 92093, USA
- Janelia Research Campus, Howard Hughes Medical Institute; Ashburn, VA 20147, USA
- Howard Hughes Medical Institute; University of California, San Diego; La Jolla, CA 92093, USA
| |
Collapse
|
3
|
Wu CT, Gonzalez Magaña D, Roshgadol J, Tian L, Ryan KK. Dietary protein restriction diminishes sucrose reward and reduces sucrose-evoked mesolimbic dopamine signaling in mice. Appetite 2024; 203:107673. [PMID: 39260700 DOI: 10.1016/j.appet.2024.107673] [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: 06/28/2024] [Revised: 09/08/2024] [Accepted: 09/08/2024] [Indexed: 09/13/2024]
Abstract
A growing literature suggests manipulating dietary protein status decreases sweet consumption in rodents and in humans. Underlying neurocircuit mechanisms have not yet been determined, but previous work points towards hedonic rather than homeostatic pathways. Here we hypothesized that a history of protein restriction reduces sucrose seeking by altering mesolimbic dopamine signaling in mice. We tested this hypothesis using established behavioral tests of palatability and conditioned reward, including the palatability contrast and conditioned place preference (CPP) tests. We used modern optical sensors for measuring real-time nucleus accumbens (NAc) dopamine dynamics during voluntary sucrose consumption, via fiber photometry, in male C57/Bl6J mice maintained on low-protein high-carbohydrate (LPHC) or control (CON) diet for ∼5 weeks. Our results showed that a history of protein restriction decreased the consumption of a sucrose 'dessert' in sated mice by ∼50% compared to controls [T-test, p < 0.05]. The dopamine release in NAc during sucrose consumption was reduced, also by ∼50%, in LPHC-fed mice compared to CON [T-test, p < 0.01]. Furthermore, LPHC-feeding blocked the sucrose-conditioned place preference we observed in CON-fed mice [paired T-test, p < 0.05], indicating reduced sucrose reward. This was accompanied by a 33% decrease in neuronal activation of the NAc core, as measured by c-Fos immunolabeling from brains collected directly after the CPP test [T-test, p < 0.05]. Together, these findings advance our mechanistic understanding of how dietary protein restriction decreases the consumption of sweets-by inhibiting the incentive salience of a sucrose reward, together with reduced sucrose-evoked dopamine release in NAc.
Collapse
Affiliation(s)
- Chih-Ting Wu
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, 95616, USA
| | - Diego Gonzalez Magaña
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, 95616, USA
| | - Jacob Roshgadol
- Biomedical Engineering Graduate Group, College of Engineering, University of California, Davis, CA, 95616, USA
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA; Max Planck Florida Institute for Neuroscience, One Max Planck Way, Jupiter, FL, 33458, USA
| | - Karen K Ryan
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
4
|
Xiao T, Roland A, Chen Y, Guffey S, Kash T, Kimbrough A. A role for circuitry of the cortical amygdala in excessive alcohol drinking, withdrawal, and alcohol use disorder. Alcohol 2024; 121:151-159. [PMID: 38447789 PMCID: PMC11371945 DOI: 10.1016/j.alcohol.2024.02.008] [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: 10/13/2023] [Revised: 01/30/2024] [Accepted: 02/26/2024] [Indexed: 03/08/2024]
Abstract
Alcohol use disorder (AUD) poses a significant public health challenge. Individuals with AUD engage in chronic and excessive alcohol consumption, leading to cycles of intoxication, withdrawal, and craving behaviors. This review explores the involvement of the cortical amygdala (CoA), a cortical brain region that has primarily been examined in relation to olfactory behavior, in the expression of alcohol dependence and excessive alcohol drinking. While extensive research has identified the involvement of numerous brain regions in AUD, the CoA has emerged as a relatively understudied yet promising candidate for future study. The CoA plays a vital role in rewarding and aversive signaling and olfactory-related behaviors and has recently been shown to be involved in alcohol-dependent drinking in mice. The CoA projects directly to brain regions that are critically important for AUD, such as the central amygdala, bed nucleus of the stria terminalis, and basolateral amygdala. These projections may convey key modulatory signaling that drives excessive alcohol drinking in alcohol-dependent subjects. This review summarizes existing knowledge on the structure and connectivity of the CoA and its potential involvement in AUD. Understanding the contribution of this region to excessive drinking behavior could offer novel insights into the etiology of AUD and potential therapeutic targets.
Collapse
Affiliation(s)
- Tiange Xiao
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - Alison Roland
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, United States; Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Yueyi Chen
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - Skylar Guffey
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - Thomas Kash
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, United States; Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Adam Kimbrough
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, United States; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States; Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN, United States.
| |
Collapse
|
5
|
Medina J, De Guzman RM, Workman JL. Prolactin mitigates chronic stress-induced maladaptive behaviors and physiology in ovariectomized female rats. Neuropharmacology 2024; 258:110095. [PMID: 39084597 DOI: 10.1016/j.neuropharm.2024.110095] [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: 03/22/2024] [Revised: 06/05/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
Stress is a major risk factor for several neuropsychiatric disorders in women, including postpartum depression. During the postpartum period, diminished ovarian hormone secretion increases susceptibility to developing depressive symptoms. Pleiotropic peptide hormones, like prolactin, are markedly released during lactation and suppress hypothalamic-pituitary-adrenal axis responses in women and acute stress-induced behavioral responses in female rodents. However, the effects of prolactin on chronic stress-induced maladaptive behaviors remain unclear. Here, we used chronic variable stress to induce maladaptive physiology in ovariectomized female rats and concurrently administered prolactin to assess its effects on several depression-relevant behavioral, endocrine, and neural characteristics. We found that chronic stress increased sucrose anhedonia and passive coping in saline-treated, but not prolactin-treated rats. Prolactin treatment did not alter stress-induced thigmotaxis, corticosterone (CORT) concentrations, hippocampal cell activation or survival. However, prolactin treatment reduced basal CORT concentrations and increased dopaminergic cells in the ventral tegmental area. Further, prolactin-treated rats had reduced microglial activation in the ventral hippocampus following chronic stress exposure. Together, these data suggest prolactin mitigates chronic stress-induced maladaptive behaviors and physiology in hypogonadal females. Moreover, these findings imply neuroendocrine-immune mechanisms by which peptide hormones confer stress resilience during periods of low ovarian hormone secretion.
Collapse
Affiliation(s)
- Joanna Medina
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave, Albany, NY, 12222, USA.
| | - Rose M De Guzman
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave, Albany, NY, 12222, USA
| | - Joanna L Workman
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave, Albany, NY, 12222, USA; Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Ave, Albany, NY, 12222, USA
| |
Collapse
|
6
|
Ezzati MR, Ezzati MJ, Fattahi M, Mozafari R, Azizbeigi R, Haghparast A. The role of D1-like dopamine receptors within the ventral tegmental area in the cannabidiol's inhibitory effects on the methamphetamine-induced conditioned place preference in rats. Brain Res Bull 2024; 216:111038. [PMID: 39097033 DOI: 10.1016/j.brainresbull.2024.111038] [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: 04/14/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
Cannabidiol (CBD) is a non-psychoactive drug extracted from marijuana. It is well established that CBD attenuates the reinforcing effects of drugs of abuse, although its mechanism of action is not fully understood. The current study tries to clarify the role of D1-like dopamine receptors (D1R) in the ventral tegmental area (VTA) in the inhibitory effects of the CBD on the acquisition and expression of methamphetamine (METH)-conditioned place preference (CPP). In the CPP training, adult male Wistar rats were conditioned with subcutaneous administration of METH (1 mg/kg) for five days. Three groups of animals were treated with multiple doses of SCH23390 (as a D1R antagonist; 0.25, 1, and 4 μg/0.3 μl saline) in the VTA, respectively, before intracerebroventricular (ICV) injection of CBD (10 μg/5 μl DMSO) in the acquisition phase. In the second experiment of the study, rats received SCH23390 in the VTA before ICV administration of CBD (50 μg/5 μl DMSO) in the expression of METH CPP. Here, the current study demonstrated that CBD inhibits the acquisition and expression of METH CPP, while microinjection of D1R antagonists (1 and 4 μg) into the VTA significantly reduced CBD's suppressive effect on the acquisition and expression of METH place preference. Furthermore, this research demonstrated that either SCH23390 or CBD alone does not lead to place preference in the CPP paradigm. Based on these data, this study suggests that pharmacological manipulations of D1R may alter the CBD's effect on METH-conditioned preference.
Collapse
Affiliation(s)
- Mohammad Reza Ezzati
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Ezzati
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mojdeh Fattahi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roghayeh Mozafari
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ronak Azizbeigi
- Department of Basic Sciences, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; School of Cognitive Sciences, Institute for Research in Fundamental Sciences, Tehran, Iran; Department of Basic Sciences, Iranian Academy of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Wu CT, Magaña DG, Roshgadol J, Tian L, Ryan KK. Dietary protein restriction diminishes sucrose reward and reduces sucrose-evoked mesolimbic dopamine signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.21.600074. [PMID: 38979357 PMCID: PMC11230173 DOI: 10.1101/2024.06.21.600074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Objective A growing literature suggests manipulating dietary protein status decreases sweet consumption in rodents and in humans. Underlying neurocircuit mechanisms have not yet been determined, but previous work points towards hedonic rather than homeostatic pathways. Here we hypothesized that a history of protein restriction reduces sucrose seeking by altering mesolimbic dopamine signaling. Methods We tested this hypothesis using established behavioral tests of palatability and motivation, including the 'palatability contrast' and conditioned place preference (CPP) tests. We used modern optical sensors for measuring real-time nucleus accumbens (NAc) dopamine dynamics during sucrose consumption, via fiber photometry, in male C57/Bl6J mice maintained on low-protein high-carbohydrate (LPHC) or control (CON) diet for ∼5 weeks. Results A history of protein restriction decreased the consumption of a sucrose 'dessert' in sated mice by ∼50% compared to controls [T-test, p< 0.05]. The dopamine release in NAc during sucrose consumption was reduced, also by ∼50%, in LPHC-fed mice compared to CON [T-test, p< 0.01]. Furthermore, LPHC-feeding blocked the sucrose-conditioned place preference we observed in CON-fed mice [paired T-test, p< 0.05], indicating reduced motivation. This was accompanied by a 33% decrease in neuronal activation of the NAc core, as measured by c-Fos immunolabeling from brains collected directly after the CPP test. Conclusions Despite ongoing efforts to promote healthier dietary habits, adherence to recommendations aimed at reducing the intake of added sugars and processed sweets remains challenging. This study highlights chronic dietary protein restriction as a nutritional intervention that suppresses the motivation for sucrose intake, via blunted sucrose-evoke dopamine release in NAc.
Collapse
|
8
|
Fujioka Y, Kawai K, Endo K, Ishibashi M, Iwade N, Tuerde D, Kaibuchi K, Yamashita T, Yamanaka A, Katsuno M, Watanabe H, Sobue G, Ishigaki S. Stress-impaired reward pathway promotes distinct feeding behavior patterns. Front Neurosci 2024; 18:1349366. [PMID: 38784098 PMCID: PMC11111882 DOI: 10.3389/fnins.2024.1349366] [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: 12/04/2023] [Accepted: 04/18/2024] [Indexed: 05/25/2024] Open
Abstract
Although dietary behaviors are affected by neuropsychiatric disorders, various environmental conditions can have strong effects as well. We found that mice under multiple stresses, including social isolation, intermittent high-fat diet, and physical restraint, developed feeding behavior patterns characterized by a deviated bait approach (fixated feeding). All the tested stressors affected dopamine release at the nucleus accumbens (NAcc) shell and dopamine normalization reversed the feeding defects. Moreover, inhibition of dopaminergic activity in the ventral tegmental area that projects into the NAcc shell caused similar feeding pattern aberrations. Given that the deviations were not consistently accompanied by changes in the amount consumed or metabolic factors, the alterations in feeding behaviors likely reflect perturbations to a critical stress-associated pathway in the mesolimbic dopamine system. Thus, deviations in feeding behavior patterns that reflect reward system abnormalities can be sensitive biomarkers of psychosocial and physical stress.
Collapse
Affiliation(s)
- Yusuke Fujioka
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Japan
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Research Division of Dementia and Neurodegenerative Disease, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kaori Kawai
- Research Division of Dementia and Neurodegenerative Disease, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan
| | - Kuniyuki Endo
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Japan
- Research Division of Dementia and Neurodegenerative Disease, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Minaka Ishibashi
- Research Division of Dementia and Neurodegenerative Disease, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nobuyuki Iwade
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Dilina Tuerde
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Research Division of Dementia and Neurodegenerative Disease, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kozo Kaibuchi
- Research Project for Neural and Tumor Signaling, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Takayuki Yamashita
- Department of Physiology, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Akihiro Yamanaka
- Chinese Institute for Brain Research, Beijing (CIBR), Beijing, China
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan
| | - Hirohisa Watanabe
- Department of Neurology, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Gen Sobue
- Aichi Medical University, Nagakute, Japan
| | - Shinsuke Ishigaki
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Japan
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Research Division of Dementia and Neurodegenerative Disease, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
9
|
Toth BA, Chang KS, Fechtali S, Burgess CR. Dopamine release in the nucleus accumbens promotes REM sleep and cataplexy. iScience 2023; 26:107613. [PMID: 37664637 PMCID: PMC10470413 DOI: 10.1016/j.isci.2023.107613] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/21/2023] [Accepted: 08/09/2023] [Indexed: 09/05/2023] Open
Abstract
Patients with the sleep disorder narcolepsy suffer from excessive daytime sleepiness, disrupted nighttime sleep, and cataplexy-the abrupt loss of postural muscle tone during wakefulness, often triggered by strong emotion. The dopamine (DA) system is implicated in both sleep-wake states and cataplexy, but little is known about the function of DA release in the striatum and sleep disorders. Recording DA release in the ventral striatum revealed orexin-independent changes across sleep-wake states as well as striking increases in DA release in the ventral, but not dorsal, striatum prior to cataplexy onset. Tonic low-frequency stimulation of ventral tegmental efferents in the ventral striatum suppressed both cataplexy and rapid eye movement (REM) sleep, while phasic high-frequency stimulation increased cataplexy propensity and decreased the latency to REM sleep. Together, our findings demonstrate a functional role of DA release in the striatum in regulating cataplexy and REM sleep.
Collapse
Affiliation(s)
- Brandon A. Toth
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Katie S. Chang
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Sarah Fechtali
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Christian R. Burgess
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
10
|
Toth BA, Chang KS, Burgess CR. Striatal dopamine regulates sleep states and narcolepsy-cataplexy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.30.542872. [PMID: 37397994 PMCID: PMC10312558 DOI: 10.1101/2023.05.30.542872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Disruptions to sleep can be debilitating and have a severe effect on daily life. Patients with the sleep disorder narcolepsy suffer from excessive daytime sleepiness, disrupted nighttime sleep, and cataplexy - the abrupt loss of postural muscle tone (atonia) during wakefulness, often triggered by strong emotion. The dopamine (DA) system is implicated in both sleep-wake states and cataplexy, but little is known about the function of DA release in the striatum - a major output region of midbrain DA neurons - and sleep disorders. To better characterize the function and pattern of DA release in sleepiness and cataplexy, we combined optogenetics, fiber photometry, and sleep recordings in a murine model of narcolepsy (orexin-/-; OX KO) and in wildtype mice. Recording DA release in the ventral striatum revealed OX-independent changes across sleep-wake states as well as striking increases in DA release in the ventral, but not dorsal, striatum prior to cataplexy onset. Tonic low frequency stimulation of ventral tegmental efferents in the ventral striatum suppressed both cataplexy and REM sleep, while phasic high frequency stimulation increased cataplexy propensity and decreased the latency to rapid eye movement (REM) sleep. Together, our findings demonstrate a functional role of DA release in the striatum in regulating cataplexy and REM sleep.
Collapse
Affiliation(s)
- Brandon A. Toth
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI USA
| | - Katie S. Chang
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI USA
| | - Christian R. Burgess
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI USA
| |
Collapse
|
11
|
Martinez Damonte V, Pomrenze MB, Manning CE, Casper C, Wolfden AL, Malenka RC, Kauer JA. Somatodendritic Release of Cholecystokinin Potentiates GABAergic Synapses Onto Ventral Tegmental Area Dopamine Cells. Biol Psychiatry 2023; 93:197-208. [PMID: 35961792 PMCID: PMC9976994 DOI: 10.1016/j.biopsych.2022.06.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/01/2022] [Accepted: 06/10/2022] [Indexed: 11/02/2022]
Abstract
BACKGROUND Neuropeptides are contained in nearly every neuron in the central nervous system and can be released not only from nerve terminals but also from somatodendritic sites. Cholecystokinin (CCK), among the most abundant neuropeptides in the brain, is expressed in the majority of midbrain dopamine neurons. Despite this high expression, CCK function within the ventral tegmental area (VTA) is not well understood. METHODS We confirmed CCK expression in VTA dopamine neurons through immunohistochemistry and in situ hybridization and detected optogenetically induced CCK release using an enzyme-linked immunosorbent assay. To investigate whether CCK modulates VTA circuit activity, we used whole-cell patch clamp recordings in mouse brain slices. We infused CCK locally in vivo and tested food intake and locomotion in fasted mice. We also used in vivo fiber photometry to measure Ca2+ transients in dopamine neurons during feeding. RESULTS Here we report that VTA dopamine neurons release CCK from somatodendritic regions, where it triggers long-term potentiation of GABAergic (gamma-aminobutyric acidergic) synapses. The somatodendritic release occurs during trains of optogenetic stimuli or prolonged but modest depolarization and is dependent on synaptotagmin-7 and T-type Ca2+ channels. Depolarization-induced long-term potentiation is blocked by a CCK2 receptor antagonist and mimicked by exogenous CCK. Local infusion of CCK in vivo inhibits food consumption and decreases distance traveled in an open field test. Furthermore, intra-VTA-infused CCK reduced dopamine cell Ca2+ signals during food consumption after an overnight fast and was correlated with reduced food intake. CONCLUSIONS Our experiments introduce somatodendritic neuropeptide release as a previously unknown feedback regulator of VTA dopamine cell excitability and dopamine-related behaviors.
Collapse
|
12
|
Park K, Clare K, Volkow ND, Pan Y, Du C. Cocaine's effects on the reactivity of the medial prefrontal cortex to ventral tegmental area stimulation: optical imaging study in mice. Addiction 2022; 117:2242-2253. [PMID: 35293056 PMCID: PMC9801493 DOI: 10.1111/add.15869] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 02/18/2022] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND AIMS The prefrontal cortex (PFC) is modulated by dopaminergic and glutamatergic neurons that project from the ventral tegmental area (VTA) and disruption of this modulation might facilitate impulsive behaviors during cocaine intoxication. Here, we assessed the effects of acute cocaine (30 mg/kg, i.p.) on the reactivity of the PFC to VTA stimulation. METHODS Using a genetically encoded calcium indicator (GCaMP6f), we optically imaged the neuronal Ca2+ reactance in medial PFC (mPFC) in response to 'tonic-like' (5 Hz) and 'phasic-like' (50 Hz) electrical VTA stimulation. The high temporal and spatial resolutions of our optical system allowed us to capture single Ca2+ neuronal transients from individual stimuli with 'tonic-like' stimulation and to visualize single neuronal activation evoked by 'phasic-like' VTA stimulation. RESULTS 'Tonic-like' VTA stimulation induced a rapid increase in mean neuronal Ca2+ in mPFC followed by a plateau and recovery upon termination of stimulation. After cocaine, the mPFC sensitivity to 'tonic-like' VTA stimulation was attenuated, with a 50.4% reduction (P = 0.03) in the number of Ca2+ transients corresponding to single electrical stimuli but the recovery time was lengthened (4.30 ± 0.25 sec to 5.41 ± 0.24 sec, P = 0.03). 'Phasic-like' stimulation evoked a rapid Ca2+ fluorescence increase in mPFC with an immediate decay process, and while cocaine did not affect the peak response (7.17 ± 1.07% versus 7.13 ± 0.96%, P = 0.98) it shortened the recovery time to baseline (3.27 ± 0.11 sec versus 2.38 ± 0.23 sec, P = 0.005). CONCLUSIONS Acute cocaine impairs reactivity of medial prefrontal cortex (mPFC) to ventral tegmental area stimulation, decreasing its sensitivity to 'tonic-like' stimulation and lengthening the recovery time to return to baseline while shortening it for phasic stimulation. These changes in mPFC might contribute to cocaine binging during intoxication.
Collapse
Affiliation(s)
- Kicheon Park
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Kevin Clare
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | | | - Yingtian Pan
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Congwu Du
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
13
|
Grinevich VP, Zakirov AN, Berseneva UV, Gerasimova EV, Gainetdinov RR, Budygin EA. Applying a Fast-Scan Cyclic Voltammetry to Explore Dopamine Dynamics in Animal Models of Neuropsychiatric Disorders. Cells 2022; 11:cells11091533. [PMID: 35563838 PMCID: PMC9100021 DOI: 10.3390/cells11091533] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 02/07/2023] Open
Abstract
Progress in the development of technologies for the real-time monitoring of neurotransmitter dynamics has provided researchers with effective tools for the exploration of etiology and molecular mechanisms of neuropsychiatric disorders. One of these powerful tools is fast-scan cyclic voltammetry (FSCV), a technique which has progressively been used in animal models of diverse pathological conditions associated with alterations in dopamine transmission. Indeed, for several decades FSCV studies have provided substantial insights into our understanding of the role of abnormal dopaminergic transmission in pathogenetic mechanisms of drug and alcohol addiction, Parkinson’s disease, schizophrenia, etc. Here we review the applications of FSCV to research neuropsychiatric disorders with particular attention to recent technological advances.
Collapse
Affiliation(s)
- Vladimir P. Grinevich
- Department of Neurobiology, Sirius University, 1 Olympic Ave., Sirius, Sochi 353340, Russia; (V.P.G.); (A.N.Z.); (U.V.B.); (E.V.G.); (R.R.G.)
| | - Amir N. Zakirov
- Department of Neurobiology, Sirius University, 1 Olympic Ave., Sirius, Sochi 353340, Russia; (V.P.G.); (A.N.Z.); (U.V.B.); (E.V.G.); (R.R.G.)
| | - Uliana V. Berseneva
- Department of Neurobiology, Sirius University, 1 Olympic Ave., Sirius, Sochi 353340, Russia; (V.P.G.); (A.N.Z.); (U.V.B.); (E.V.G.); (R.R.G.)
| | - Elena V. Gerasimova
- Department of Neurobiology, Sirius University, 1 Olympic Ave., Sirius, Sochi 353340, Russia; (V.P.G.); (A.N.Z.); (U.V.B.); (E.V.G.); (R.R.G.)
| | - Raul R. Gainetdinov
- Department of Neurobiology, Sirius University, 1 Olympic Ave., Sirius, Sochi 353340, Russia; (V.P.G.); (A.N.Z.); (U.V.B.); (E.V.G.); (R.R.G.)
- Institute of Translational Biomedicine and St. Petersburg State University Hospital, St. Petersburg State University, Universitetskaya Emb. 7-9, St. Petersburg 199034, Russia
| | - Evgeny A. Budygin
- Department of Neurobiology, Sirius University, 1 Olympic Ave., Sirius, Sochi 353340, Russia; (V.P.G.); (A.N.Z.); (U.V.B.); (E.V.G.); (R.R.G.)
- Correspondence:
| |
Collapse
|
14
|
Logan CN, Rojas G, Wilkinson CS, Polo Escorcia AK, Reichel CM, Peris J, Knackstedt LA. Systemic oxytocin increases glutamate efflux in the nucleus accumbens core of cocaine-experienced male and female rats but only increases dopamine efflux in males. Behav Brain Res 2022; 417:113590. [PMID: 34551348 DOI: 10.1016/j.bbr.2021.113590] [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: 06/11/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 01/10/2023]
Abstract
Oxytocin attenuates cocaine-seeking when administered both systemically and directly into the nucleus accumbens core. This effect is blocked by intra-accumbens antagonism of mGlu2/3 and, together with our finding that intra-accumbens oxytocin increases glutamate concentrations in this brain region, indicates that pre-synaptic regulation of glutamate release by oxytocin influences cocaine relapse. However, mGlu2/3 receptors also regulate dopamine release in the nucleus accumbens. Here we aimed to determine whether systemic oxytocin increases glutamate and dopamine concentrations in the nucleus accumbens core of cocaine-experienced and cocaine-naïve male and female rats. A subset of rats self-administered cocaine (0.5 mg/kg/infusion) and then underwent extinction training for 2-3 weeks. Rats were implanted with microdialysis probes in the accumbens core and samples were collected for a baseline period, and following saline (1 mL/kg), and oxytocin (1 mg/kg, IP) injections. Locomotion was assessed during microdialysis. In cocaine-experienced rats, oxytocin increased glutamate concentrations in the accumbens core to the same extent in males and females but only increased dopamine concentrations in male rats. Oxytocin did not alter glutamate levels in cocaine-naïve rats. Oxytocin did not produce sedation. These results extend previous findings that systemic oxytocin increases nucleus accumbens dopamine in a sex-specific manner in cocaine-experienced rats. These data are the first to find that systemic oxytocin increases nucleus accumbens glutamate after cocaine experience, providing a mechanism of action by which oxytocin attenuates the reinstatement of cocaine seeking in both male and female rats.
Collapse
Affiliation(s)
- C N Logan
- Psychology Department, University of Florida, Gainesville, FL, USA; Center for Addiction Research and Education, University of Florida, Gainesville, FL, USA.
| | - G Rojas
- Psychology Department, University of Florida, Gainesville, FL, USA
| | - C S Wilkinson
- Psychology Department, University of Florida, Gainesville, FL, USA; Center for Addiction Research and Education, University of Florida, Gainesville, FL, USA
| | | | - C M Reichel
- Neuroscience Dept., Medical University of South Carolina, Charleston SC, USA
| | - J Peris
- Center for Addiction Research and Education, University of Florida, Gainesville, FL, USA; Pharmacodynamics Department, University of Florida, Gainesville, FL, USA
| | - L A Knackstedt
- Psychology Department, University of Florida, Gainesville, FL, USA; Center for Addiction Research and Education, University of Florida, Gainesville, FL, USA
| |
Collapse
|
15
|
Bosque-Cordero KY, Vazquez-Torres R, Calo-Guadalupe C, Consuegra-Garcia D, Fois GR, Georges F, Jimenez-Rivera CA. I h blockade reduces cocaine-induced firing patterns of putative dopaminergic neurons of the ventral tegmental area in the anesthetized rat. Prog Neuropsychopharmacol Biol Psychiatry 2022; 112:110431. [PMID: 34454991 PMCID: PMC8489561 DOI: 10.1016/j.pnpbp.2021.110431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/23/2021] [Accepted: 08/23/2021] [Indexed: 01/12/2023]
Abstract
The hyperpolarization-activated cation current (Ih) is a determinant of intrinsic excitability in various cells, including dopaminergic neurons (DA) of the ventral tegmental area (VTA). In contrast to other cellular conductances, Ih is activated by hyperpolarization negative to -55 mV and activating Ih produces a time-dependent depolarizing current. Our laboratory demonstrated that cocaine sensitization, a chronic cocaine behavioral model, significantly reduces Ih amplitude in VTA DA neurons. Despite this reduction in Ih, the spontaneous firing of VTA DA cells after cocaine sensitization remained similar to control groups. Although the role of Ih in controlling VTA DA excitability is still poorly understood, our hypothesis is that Ih reduction could play a role of a homeostatic controller compensating for cocaine-induced change in excitability. Using in vivo single-unit extracellular electrophysiology in isoflurane anesthetized rats, we explored the contribution of Ih on spontaneous firing patterns of VTA DA neurons. A key feature of spontaneous excitability is bursting activity; bursting is defined as trains of two or more spikes occurring within a short interval and followed by a prolonged period of inactivity. Burst activity increases the reliability of information transfer. To elucidate the contribution of Ih to spontaneous firing patterns of VTA DA neurons, we locally infused an Ih blocker (ZD 7288, 8.3 μM) and evaluated its effect. Ih blockade significantly reduced firing rate, bursting frequency, and percent of spikes within a burst. In addition, Ih blockade significantly reduced acute cocaine-induced spontaneous firing rate, bursting frequency, and percent of spikes within a burst. Using whole-cell patch-clamp, we determine the progressive reduction of Ih after acute and chronic cocaine administration (15 mg/k.g intraperitoneally). Our data show a significant reduction (~25%) in Ih amplitude after 24 but not 2 h of acute cocaine administration. These results suggest that a progressive reduction of Ih could serve as a homeostatic regulator of cocaine-induced spontaneous firing patterns related to VTA DA excitability.
Collapse
Affiliation(s)
| | | | | | | | - Giulia R Fois
- University of Bordeaux, Neurodegeneratives Diseases Institute, IMN-UMR-CNRS 5293, 146 rue Léo Saignat, 33076 Bordeaux, France; CNRS, Neurodegeneratives Diseases Institute, IMN-UMR-CNRS 5293, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - François Georges
- University of Bordeaux, Neurodegeneratives Diseases Institute, IMN-UMR-CNRS 5293, 146 rue Léo Saignat, 33076 Bordeaux, France; CNRS, Neurodegeneratives Diseases Institute, IMN-UMR-CNRS 5293, 146 rue Léo Saignat, 33076 Bordeaux, France
| | | |
Collapse
|
16
|
Grinevich VP, Krupitsky EM, Gainetdinov RR, Budygin EA. Linking Ethanol-Addictive Behaviors With Brain Catecholamines: Release Pattern Matters. Front Behav Neurosci 2022; 15:795030. [PMID: 34975429 PMCID: PMC8716449 DOI: 10.3389/fnbeh.2021.795030] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/26/2021] [Indexed: 12/30/2022] Open
Abstract
Using a variety of animal models that simulate key features of the alcohol use disorder (AUD), remarkable progress has been made in identifying neurochemical targets that may contribute to the development of alcohol addiction. In this search, the dopamine (DA) and norepinephrine (NE) systems have been long thought to play a leading role in comparison with other brain systems. However, just recent development and application of optogenetic approaches into the alcohol research field provided opportunity to identify neuronal circuits and specific patterns of neurotransmission that govern the key components of ethanol-addictive behaviors. This critical review summarizes earlier findings, which initially disclosed catecholamine substrates of ethanol actions in the brain and shows how the latest methodologies help us to reveal the significance of DA and NE release changes. Specifically, we focused on recent optogenetic investigations aimed to reveal cause-effect relationships between ethanol-drinking (seeking and taking) behaviors and catecholamine dynamics in distinct brain pathways. These studies gain the knowledge that is needed for the better understanding addiction mechanisms and, therefore, for development of more effective AUD treatments. Based on the reviewed findings, new messages for researches were indicated, which may have broad applications beyond the field of alcohol addiction.
Collapse
Affiliation(s)
- Vladimir P Grinevich
- Department of Neurobiology, Sirius University of Science and Technology, Sochi, Russia
| | - Evgeny M Krupitsky
- V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology, St. Petersburg, Russia.,Laboratory of Clinical Psychopharmacology of Addictions, St.-Petersburg First Pavlov State Medical University, St. Petersburg, Russia
| | - Raul R Gainetdinov
- Department of Neurobiology, Sirius University of Science and Technology, Sochi, Russia.,Institute of Translational Biomedicine and St. Petersburg State University Hospital, St. Petersburg State University, St. Petersburg, Russia
| | - Evgeny A Budygin
- Department of Neurobiology, Sirius University of Science and Technology, Sochi, Russia
| |
Collapse
|
17
|
Medina J, De Guzman RM, Workman JL. Lactation is not required for maintaining maternal care and active coping responses in chronically stressed postpartum rats: Interactions between nursing demand and chronic variable stress. Horm Behav 2021; 136:105035. [PMID: 34488064 DOI: 10.1016/j.yhbeh.2021.105035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 06/11/2021] [Accepted: 07/04/2021] [Indexed: 10/20/2022]
Abstract
Women who do not breastfeed or discontinue breastfeeding early are more likely to develop postpartum depression (PPD) and stress is a significant risk factor for depression, including PPD. Using a rat model, we investigated whether the absence of nursing would increase the susceptibility to chronic stress-related behavioral and neural changes during the postpartum period. Adult female rats underwent thelectomy (thel; removal of teats), sham surgery, or no surgery (control) and were paired with males for breeding. All litters were rotated twice daily until postpartum day (PD) 26. Sham rats served as surrogates for thel litters, yielding a higher nursing demand for sham rats. Concurrently, rats received either no stress or chronic variable stress until PD 25. Rats were observed for maternal behaviors and tested in a series of tasks including open field, sucrose preference, and forced swim. We used immunohistochemistry (IHC) for doublecortin (DCX; to label immature neurons) or for mineralocorticoid receptor (MR). Contrary to our expectations, non-nursing thel rats were resistant to the effects of stress in all dependent measures. Our data indicate that even in chronic adverse conditions, nursing is not required for maintaining stable care to offspring or active coping responses in an acutely stressful task. We discuss the possible role of offspring contact and consider future directions for biomedical and clinical research. In rats with high nursing demand, however, chronic stress increased immobility, hippocampal neurogenesis, and MR expression (largely in opposition to the effects of stress in rats with typical nursing demand). We discuss these patterns in the context of energetics and allostatic load. This research highlights the complexity in relationships between stress, nursing, and neurobehavioral outcomes in the postpartum period and underscores the need for additional biomedical and clinical research geared toward optimizing treatments and interventions for women with PPD, regardless of breastfeeding status. SIGNIFICANCE STATEMENT: The goal of this research was to determine how the absence of nursing and higher nursing demand impact stress-coping behaviors and neural changes associated with chronic stress in order to disentangle the complex interplay of factors that contribute to psychological illness during the postpartum period.
Collapse
Affiliation(s)
- Joanna Medina
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave., Albany, NY 12222, United States of America
| | - Rose M De Guzman
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave., Albany, NY 12222, United States of America
| | - Joanna L Workman
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave., Albany, NY 12222, United States of America; Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Ave., Albany, NY 12222, United States of America.
| |
Collapse
|
18
|
VTA MC3R neurons control feeding in an activity- and sex-dependent manner in mice. Neuropharmacology 2021; 197:108746. [PMID: 34371079 DOI: 10.1016/j.neuropharm.2021.108746] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 01/25/2023]
Abstract
Increasing evidence indicates that the melanocortin and mesolimbic dopamine (DA) systems interact to regulate feeding and body weight. Because melanocortin-3 receptors (MC3R) are highly expressed in the ventral tegmental area (VTA), we tested whether VTA neurons expressing these receptors (VTA MC3R neurons) control feeding and body weight in vivo. We also tested whether there were sex differences in the ability of VTA MC3R neurons to control feeding, as MC3R -/- mice show sex-dependent alterations in reward feeding and DA levels, and there are clear sex differences in multiple DA-dependent behaviors and disorders. Designer receptors exclusively activated by designer drugs (DREADD) were used to acutely activate and inhibit VTA MC3R neurons and changes in food intake and body weight were measured. Acutely altering the activity of VTA MC3R neurons decreased feeding in an activity- and sex-dependent manner, with acute activation decreasing feeding, but only in females, and acute inhibition decreasing feeding, but only in males. These differences did not appear to be due to sex differences in the number of VTA MC3R neurons, the ability of hM3Dq to activate VTA MC3R neurons, or the proportion of VTA MC3R neurons expressing tyrosine hydroxylase (TH). These studies demonstrate an important role for VTA MC3R neurons in the control of feeding and reveal important sex differences in behavior, whereby opposing changes in neuronal activity in male and female mice cause similar changes in behavior.
Collapse
|
19
|
Selective Overexpression of Collybistin in Mouse Hippocampal Pyramidal Cells Enhances GABAergic Neurotransmission and Protects against PTZ-Induced Seizures. eNeuro 2021; 8:ENEURO.0561-20.2021. [PMID: 34083383 PMCID: PMC8281261 DOI: 10.1523/eneuro.0561-20.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 04/02/2021] [Accepted: 05/23/2021] [Indexed: 11/21/2022] Open
Abstract
Collybistin (CB) is a rho guanine exchange factor found at GABAergic and glycinergic postsynapses that interacts with the inhibitory scaffold protein, gephyrin, and induces accumulation of gephyrin and GABA type-A receptors (GABAARs) to the postsynapse. We have previously reported that the isoform without the src homology 3 (SH3) domain, CBSH3-, is particularly active in enhancing the GABAergic postsynapse in both cultured hippocampal neurons as well as in cortical pyramidal neurons after chronic in vivo expression in in utero electroporated (IUE) rats. Deficiency of CB in knock-out (KO) mice results in absence of gephyrin and gephyrin-dependent GABAARs at postsynaptic sites in several brain regions, including hippocampus. In the present study, we have generated an adeno-associated virus (AAV) that expresses CBSH3- in a cre-dependent manner. Using male and female VGLUT1-IRES-cre or VGAT-IRES-cre mice, we explore the effect of overexpression of CBSH3- in hippocampal pyramidal cells or hippocampal interneurons. The results show that: (1) the accumulation of gephyrin and GABAARs at inhibitory postsynapses in hippocampal pyramidal neurons or interneurons can be enhanced by CBSH3- overexpression; (2) overexpression of CBSH3- in hippocampal pyramidal cells can enhance the strength of inhibitory neurotransmission; and (3) these enhanced inhibitory synapses provide protection against pentylenetetrazole (PTZ)-induced seizures. The results indicate that this AAV vector carrying CBSH3- can be used for in vivo enhancement of GABAergic synaptic transmission in selected target neurons in the brain.
Collapse
|
20
|
Obesity and dietary fat influence dopamine neurotransmission: exploring the convergence of metabolic state, physiological stress, and inflammation on dopaminergic control of food intake. Nutr Res Rev 2021; 35:236-251. [PMID: 34184629 DOI: 10.1017/s0954422421000196] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The aim of this review is to explore how metabolic changes induced by diets high in saturated fat (HFD) affect nucleus accumbens (NAc) dopamine neurotransmission and food intake, and to explore how stress and inflammation influence this process. Recent evidence linked diet-induced obesity and HFD with reduced dopamine release and reuptake. Altered dopamine neurotransmission could disrupt satiety circuits between NAc dopamine terminals and projections to the hypothalamus. The NAc directs learning and motivated behaviours based on homeostatic needs and psychological states. Therefore, impaired dopaminergic responses to palatable food could contribute to weight gain by disrupting responses to food cues or stress, which impacts type and quantity of food consumed. Specifically, saturated fat promotes neuronal resistance to anorectic hormones and activation of immune cells that release proinflammatory cytokines. Insulin has been shown to regulate dopamine neurotransmission by enhancing satiety, but less is known about effects of diet-induced stress. Therefore, changes to dopamine signalling due to HFD warrant further examination to characterise crosstalk of cytokines with endocrine and neurotransmitter signals. A HFD promotes a proinflammatory environment that may disrupt neuronal endocrine function and dopamine signalling that could be exacerbated by the hypothalamic-pituitary-adrenal and κ-opioid receptor stress systems. Together, these adaptive changes may dysregulate eating by changing NAc dopamine during hedonic versus homeostatic food intake. This could drive palatable food cravings during energy restriction and hinder weight loss. Understanding links between HFD and dopamine neurotransmission will inform treatment strategies for diet-induced obesity and identify molecular candidates for targeted therapeutics.
Collapse
|
21
|
Gui H, Liu C, He H, Zhang J, Chen H, Zhang Y. Dopaminergic Projections From the Ventral Tegmental Area to the Nucleus Accumbens Modulate Sevoflurane Anesthesia in Mice. Front Cell Neurosci 2021; 15:671473. [PMID: 33994950 PMCID: PMC8119636 DOI: 10.3389/fncel.2021.671473] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/06/2021] [Indexed: 01/03/2023] Open
Abstract
The role of the dopaminergic pathway in general anesthesia and its potential mechanisms are still unknown. In this study, we usedc-Fos staining combined with calcium fiber photometry recording to explore the activity of ventral tegmental area (VTA) dopaminergic neurons(VTA-DA) and nucleus accumbens (NAc) neurons during sevoflurane anesthesia. A genetically encoded dopamine (DA) sensor was used to investigate thefunction of the NAc in sevoflurane anesthesia. Chemogenetics and optogenetics were used to explore the role of the VTA-DA in sevofluraneanesthesia. Electroencephalogram (EEG) spectra, time of loss of righting reflex (LORR) and recovery of righting reflex (RORR) were recorded asassessment indicators. We found that VTA-DA and NAc neurons were inhibited during the induction period and were activated during the recoveryperiod of sevoflurane anesthesia. The fluorescence signals of dopamine decreased in the induction of and increased in the emergence from sevoflurane anesthesia.Activation of VTA-DA and the VTADA-NAc pathway delayed the induction and facilitated the emergence accompanying with thereduction of delta band and the augmentation of the gamma band. These data demonstrate that VTA-DA neurons play a critical role in modulating sevofluraneanesthesia via the VTADA-NAc pathway.
Collapse
Affiliation(s)
- Huan Gui
- Department of Anesthesiology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China.,Guizhou Key Laboratory of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,School of Anesthesiology, Zunyi Medical University, Zunyi, China
| | - Chengxi Liu
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,School of Anesthesiology, Zunyi Medical University, Zunyi, China
| | - Haifeng He
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,School of Anesthesiology, Zunyi Medical University, Zunyi, China
| | - Jie Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China.,Guizhou Key Laboratory of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,School of Anesthesiology, Zunyi Medical University, Zunyi, China
| | - Hong Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China.,School of Anesthesiology, Zunyi Medical University, Zunyi, China
| | - Yi Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China.,Guizhou Key Laboratory of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,School of Anesthesiology, Zunyi Medical University, Zunyi, China
| |
Collapse
|
22
|
Wakabayashi KT, Feja M, Leigh MPK, Baindur AN, Suarez M, Meyer PJ, Bass CE. Chemogenetic Activation of Mesoaccumbal Gamma-Aminobutyric Acid Projections Selectively Tunes Responses to Predictive Cues When Reward Value Is Abruptly Decreased. Biol Psychiatry 2021; 89:366-375. [PMID: 33168181 PMCID: PMC8570639 DOI: 10.1016/j.biopsych.2020.08.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 08/03/2020] [Accepted: 08/21/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Mesolimbic circuits regulate the attribution of motivational significance to incentive cues that predict reward, yet this network also plays a key role in adapting reward-seeking behavior when the contingencies linked to a cue unexpectedly change. Here, we asked whether mesoaccumbal GABA (gamma-aminobutyric acid) projections enhance adaptive responding to incentive cues of abruptly altered reward value, and whether these effects were distinct from global activation of all ventral tegmental area GABA circuits. METHODS We used a viral targeting system to chemogenetically activate mesoaccumbal GABA projections in male rats during a novel cue-dependent operant value-shifting task, in which the volume of a sucrose reward associated with a predictive cue is suddenly altered, from the beginning and throughout the session. We compared the results with global activation of ventral tegmental area GABA neurons, which will activate local inhibitory circuits and long loop projections. RESULTS We found that activation of mesoaccumbal GABA projections decreases responding to incentive cues associated with smaller-than-expected rewards. This tuning of behavioral responses was specific to cues associated with smaller-than-expected rewards but did not impact measures related to consuming the reward. In marked contrast, activating all ventral tegmental area GABA neurons resulted in a uniform decrease in responding to incentive cues irrespective of changes in the size of the reward. CONCLUSIONS Targeted activation of mesoaccumbal GABA neurons facilitates adaptation in reward-seeking behaviors. This suggests that these projections may play a very specific role in associative learning processes.
Collapse
Affiliation(s)
- Ken T Wakabayashi
- Department of Psychology, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Malte Feja
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York
| | - Martin P K Leigh
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York
| | - Ajay N Baindur
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York
| | - Mauricio Suarez
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York; Clinical Research Institute on Addictions, State University of New York at Buffalo, Buffalo, New York
| | - Paul J Meyer
- Department of Psychology, State University of New York at Buffalo, Buffalo, New York
| | - Caroline E Bass
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York; Clinical Research Institute on Addictions, State University of New York at Buffalo, Buffalo, New York.
| |
Collapse
|
23
|
McKendrick G, Sharma S, Sun D, Randall PA, Graziane NM. Acute and chronic bupropion treatment does not prevent morphine-induced conditioned place preference in mice. Eur J Pharmacol 2020; 889:173638. [PMID: 33039460 DOI: 10.1016/j.ejphar.2020.173638] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/28/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022]
Abstract
A substantial barrier to the treatment of Opioid Use Disorder (OUD) is the elevated relapse rates in affected patients, and a significant contributor to these events of relapse is exposure to cues and contexts that are intensely associated with prior drug abuse. The neurotransmitter dopamine plays a key role in reward-related behaviors, and previous studies have illustrated that dopamine hypofunction in periods of abstinence serves to prompt drug craving and seeking. We hypothesized that restoration of dopaminergic signaling could attenuate drug-seeking behaviors. Therefore, we investigated whether use of an FDA-approved drug, bupropion, an inhibitor of the dopamine transporter (DAT), or a dopamine uptake inhibitor with high affinity for DAT, JHW 007, was able to decrease preference for a drug-paired context. In these experiments, mice underwent 5 days of non-contingent morphine (10 mg/kg) exposure in a conditioned place preference (CPP) paradigm. We found that systemic injection of bupropion (20 mg/kg, i. p.) or intracranial injection of JHW 007 into the nucleus accumbens shell did not prevent the expression of morphine CPP. We then investigated whether chronic bupropion treatment (via implanted osmotic pumps) would influence morphine CPP. We observed that chronic bupropion treatment for 21 days following morphine conditioning did not attenuate the prolonged preference for morphine-paired contexts. Overall, with our dose and paradigm, neither acute nor chronic bupropion diminishes morphine CPP. Continued studies should address FDA-approved medications and their potential for recovery in OUD patients.
Collapse
Affiliation(s)
- Greer McKendrick
- Neuroscience Graduate Program, Penn State College of Medicine, Hershey, PA 17033, USA; Department of Anesthesiology and Perioperative Medicine, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Sonakshi Sharma
- Department of Anesthesiology and Perioperative Medicine, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Dongxiao Sun
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Patrick A Randall
- Departments of Anesthesiology and Perioperative Medicine and Pharmacology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Nicholas M Graziane
- Departments of Anesthesiology and Perioperative Medicine and Pharmacology, Penn State College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
24
|
Deal AL, Bass CE, Grinevich VP, Delbono O, Bonin KD, Weiner JL, Budygin EA. Bidirectional Control of Alcohol-drinking Behaviors Through Locus Coeruleus Optoactivation. Neuroscience 2020; 443:84-92. [PMID: 32707291 DOI: 10.1016/j.neuroscience.2020.07.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/10/2020] [Accepted: 07/14/2020] [Indexed: 12/17/2022]
Abstract
The relationship between stress and alcohol-drinking behaviors has been intensively explored; however, neuronal substrates and neurotransmitter dynamics responsible for a causal link between these conditions are still unclear. Here, we optogenetically manipulated locus coeruleus (LC) norepinephrine (NE) activity by applying distinct stimulation protocols in order to explore how phasic and tonic NE release dynamics control alcohol-drinking behaviors. Our results clearly demonstrate contrasting behavioral consequences of LC-NE circuitry activation during low and high frequency stimulation. Specifically, applying tonic stimulation during a standard operant drinking session resulted in increased intake, while phasic stimulation decreased this measure. Furthermore, stimulation during extinction probe trials, when the lever press response was not reinforced, did not significantly alter alcohol-seeking behavior if a tonic pattern was applied. However, phasic stimulation substantially suppressed the number of lever presses, indicating decreased alcohol seeking under the same experimental condition. Given the well-established correlative link between stress and increased alcohol consumption, here we provide the first evidence that tonic LC-NE activity plays a causal role in stress-associated increases in drinking.
Collapse
Affiliation(s)
- Alex L Deal
- Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Caroline E Bass
- Department of Pharmacology and Toxicology, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Valentina P Grinevich
- Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Osvaldo Delbono
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Keith D Bonin
- Department of Physics, Wake Forest University, Winston-Salem, NC, USA
| | - Jeff L Weiner
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Evgeny A Budygin
- Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
25
|
Galinowski A, Miranda R, Lemaitre H, Artiges E, Paillère Martinot M, Filippi I, Penttilä J, Grimmer Y, Noort BM, Stringaris A, Becker A, Isensee C, Struve M, Fadai T, Kappel V, Goodman R, Banaschewski T, Bokde AL, Bromberg U, Brühl R, Büchel C, Cattrell A, Conrod P, Desrivières S, Flor H, Fröhner JH, Frouin V, Gallinat J, Garavan H, Gowland P, Heinz A, Hohmann S, Jurk S, Millenet S, Nees F, Papadopoulos‐Orfanos D, Poustka L, Quinlan EB, Smolka MN, Walter H, Whelan R, Schumann G, Martinot J. Heavy drinking in adolescents is associated with change in brainstem microstructure and reward sensitivity. Addict Biol 2020; 25:e12781. [PMID: 31328396 DOI: 10.1111/adb.12781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 03/31/2019] [Accepted: 05/02/2019] [Indexed: 11/30/2022]
Abstract
Heavy drinker adolescents: altered brainstem microstructure.
Collapse
Affiliation(s)
- André Galinowski
- INSERM, UMR 1000, Research Unit “Neuroimaging and Psychiatry”, Paris Saclay University, Paris Descartes University, DIGITEO Labs, Gif sur Yvette France
| | - Ruben Miranda
- INSERM, UMR 1000, Research Unit “Neuroimaging and Psychiatry”, Paris Saclay University, Paris Descartes University, DIGITEO Labs, Gif sur Yvette France
- Department of Psychiatry and AddictologyPaul Brousse Hospital Villejuif France
| | - Hervé Lemaitre
- INSERM, UMR 1000, Research Unit “Neuroimaging and Psychiatry”, Paris Saclay University, Paris Descartes University, DIGITEO Labs, Gif sur Yvette France
| | - Eric Artiges
- INSERM, UMR 1000, Research Unit “Neuroimaging and Psychiatry”, Paris Saclay University, Paris Descartes University, DIGITEO Labs, Gif sur Yvette France
- Center for Neuroimaging Research (CENIR)Brain & Spine Institute Paris France
- Psychiatry Department 91G16Orsay Hospital Orsay France
| | - Marie‐Laure Paillère Martinot
- INSERM, UMR 1000, Research Unit “Neuroimaging and Psychiatry”, Paris Saclay University, Paris Descartes University, DIGITEO Labs, Gif sur Yvette France
- Department of Child and Adolescent PsychiatryPitié‐Salpêtrière Hospital Paris France
| | - Irina Filippi
- INSERM, UMR 1000, Research Unit “Neuroimaging and Psychiatry”, Paris Saclay University, Paris Descartes University, DIGITEO Labs, Gif sur Yvette France
| | - Jani Penttilä
- Department of Social and Health CarePsychosocial Services Adolescent Outpatient Clinic Lahti Finland
| | - Yvonne Grimmer
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty MannheimHeidelberg University Mannheim Germany
| | - Betteke M. Noort
- Department of Child and Adolescent Psychiatry Psychosomatics and Psychotherapy, Campus Charité MitteCharité‐Universitätsmedizin Berlin Berlin Germany
| | - Argyris Stringaris
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology & NeuroscienceKing's College London London UK
| | - Andreas Becker
- Department of Child and Adolescent Psychiatry and PsychotherapyUniversity Medical Centre Göttingen Göttingen Germany
| | - Corinna Isensee
- Department of Child and Adolescent Psychiatry and PsychotherapyUniversity Medical Centre Göttingen Göttingen Germany
| | - Maren Struve
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty MannheimHeidelberg University Mannheim Germany
| | - Tahmine Fadai
- Department of Systems NeuroscienceUniversity Medical Centre Hamburg‐Eppendorf Hamburg Germany
| | - Viola Kappel
- Department of Child and Adolescent Psychiatry Psychosomatics and Psychotherapy, Campus Charité MitteCharité‐Universitätsmedizin Berlin Berlin Germany
| | - Robert Goodman
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology & NeuroscienceKing's College London London UK
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty MannheimHeidelberg University Mannheim Germany
| | - Arun L.W. Bokde
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of NeurosciencesTrinity College Dublin Ireland
| | - Uli Bromberg
- Department of Systems NeuroscienceUniversity Medical Centre Hamburg‐Eppendorf Hamburg Germany
| | - Rüdiger Brühl
- Physikalisch‐Technische Bundesanstalt, Abbestr. 2 ‐ 12 Berlin Germany
| | - Christian Büchel
- Department of Systems NeuroscienceUniversity Medical Centre Hamburg‐Eppendorf Hamburg Germany
| | - Anna Cattrell
- Medical Research Council—Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & NeuroscienceKing's College London London UK
| | - Patricia Conrod
- Department of PsychiatryMontreal University, CHU Ste Justine Hospital 3175 Côte-Ste-Catherine Montréal, Québec, H3T 1C5 Canada
| | - Sylvane Desrivières
- Medical Research Council—Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & NeuroscienceKing's College London London UK
| | - Herta Flor
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty MannheimHeidelberg University Mannheim Germany
| | - Juliane H. Fröhner
- Department of Psychiatry and PsychotherapyUniversity Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Vincent Frouin
- Neurospin, Commissariat à l'Energie Atomique, CEA‐Saclay Center Paris France
| | - Juergen Gallinat
- Department of Psychiatry and PsychotherapyUniversity Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Hugh Garavan
- Department of PsychiatryUniversity of Vermont Burlington Vermont
- Department of PsychologyUniversity of Vermont Burlington Vermont
| | - Penny Gowland
- Sir Peter Mansfield Imaging Centre School of Physics and AstronomyUniversity of Nottingham, University Park Nottingham UK
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy, Campus Charité MitteCharité‐Universitätsmedizin Berlin Berlin Germany
| | - Sarah Hohmann
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty MannheimHeidelberg University Mannheim Germany
| | - Sarah Jurk
- Department of Psychiatry and Neuroimaging CenterTechnische Universität Dresden Dresden Germany
| | - Sabina Millenet
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty MannheimHeidelberg University Mannheim Germany
| | - Frauke Nees
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty MannheimHeidelberg University Mannheim Germany
| | | | - Luise Poustka
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty MannheimHeidelberg University Mannheim Germany
- Department of Child and Adolescent Psychiatry and PsychotherapyMedical University of Vienna Vienna Austria
| | - Erin Burke Quinlan
- Department of Psychological Medicine and Psychiatry, Institute of Psychiatry, Psychology & NeuroscienceKing's College London London UK
| | - Michael N. Smolka
- Department of Psychiatry and Neuroimaging CenterTechnische Universität Dresden Dresden Germany
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy, Campus Charité MitteCharité‐Universitätsmedizin Berlin Berlin Germany
| | - Robert Whelan
- Department of PsychologyUniversity College Dublin Ireland
| | - Gunter Schumann
- Medical Research Council—Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & NeuroscienceKing's College London London UK
| | - Jean‐Luc Martinot
- INSERM, UMR 1000, Research Unit “Neuroimaging and Psychiatry”, Paris Saclay University, Paris Descartes University, DIGITEO Labs, Gif sur Yvette France
- Center for Neuroimaging Research (CENIR)Brain & Spine Institute Paris France
| | | |
Collapse
|
26
|
Budygin EA, Bass CE, Grinevich VP, Deal AL, Bonin KD, Weiner JL. Opposite Consequences of Tonic and Phasic Increases in Accumbal Dopamine on Alcohol-Seeking Behavior. iScience 2020; 23:100877. [PMID: 32062422 PMCID: PMC7031354 DOI: 10.1016/j.isci.2020.100877] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 08/27/2019] [Accepted: 01/28/2020] [Indexed: 12/21/2022] Open
Abstract
Despite many years of work on dopaminergic mechanisms of alcohol addiction, much of the evidence remains mostly correlative in nature. Fortunately, recent technological advances have provided the opportunity to explore the causal role of alterations in neurotransmission within circuits involved in addictive behaviors. Here, we address this critical gap in our knowledge by integrating an optogenetic approach and an operant alcohol self-administration paradigm to assess directly how accumbal dopamine (DA) release dynamics influences the appetitive (seeking) component of alcohol-drinking behavior. We show that appetitive reward-seeking behavior in rats trained to self-administer alcohol can be shaped causally by ventral tegmental area-nucleus accumbens (VTA-NAc) DA neurotransmission. Our findings reveal that phasic patterns of DA release within this circuit enhance a discrete measure of alcohol seeking, whereas tonic patterns of stimulation inhibit this behavior. Moreover, we provide mechanistic evidence that tonic-phasic interplay within the VTA-NAc DA circuit underlies these seemingly paradoxical effects. VTA-NAc DA transmission can bidirectionally modulate motivated behavior Optogenetic increases in phasic DA release in the NAc enhance alcohol seeking Optogenetic increases in tonic DA release in the NAc inhibit alcohol seeking Phasic DA release can be decreased by the concurrent tonic activation
Collapse
Affiliation(s)
- Evgeny A Budygin
- Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA.
| | - Caroline E Bass
- Department of Pharmacology and Toxicology, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Valentina P Grinevich
- Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Alex L Deal
- Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Keith D Bonin
- Department of Physics, Wake Forest University, Winston-Salem, NC, USA
| | - Jeff L Weiner
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
27
|
Dunn DP, Bastacky JM, Gray CC, Abtahi S, Currie PJ. Role of mesolimbic ghrelin in the acquisition of cocaine reward. Neurosci Lett 2019; 709:134367. [DOI: 10.1016/j.neulet.2019.134367] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 12/27/2022]
|
28
|
Lozano-Montes L, Astori S, Abad S, Guillot de Suduiraut I, Sandi C, Zalachoras I. Latency to Reward Predicts Social Dominance in Rats: A Causal Role for the Dopaminergic Mesolimbic System. Front Behav Neurosci 2019; 13:69. [PMID: 31024272 PMCID: PMC6460316 DOI: 10.3389/fnbeh.2019.00069] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 03/18/2019] [Indexed: 12/22/2022] Open
Abstract
Reward signals encoded in the mesolimbic dopaminergic system guide approach/seeking behaviors to all varieties of life-supporting stimuli (rewards). Differences in dopamine (DA) levels have been found between dominant and submissive animals. However, it is still unclear whether these differences arise as a consequence of the rewarding nature of the acquisition of a dominant rank, or whether they preexist and favor dominance by promoting reward-seeking behavior. Given that acquisition of a social rank determines animals' priority access to resources, we hypothesized that differences in reward-seeking behavior might affect hierarchy establishment and that modulation of the dopaminergic system could affect the outcome of a social competition. We characterized reward-seeking behaviors based on rats' latency to get a palatable-reward when given temporary access to it. Subsequently, rats exhibiting short (SL) and long (LL) latency to get the rewards cohabitated for more than 2 weeks, in order to establish a stable hierarchy. We found that SL animals exhibited dominant behavior consistently in social competition tests [for palatable-rewards and two water competition tests (WCTs)] after hierarchy was established, indicating that individual latency to rewards predicted dominance. Moreover, because SL animals showed higher mesolimbic levels of DA than LL rats, we tested whether stimulation of mesolimbic DA neurons could affect the outcome of a social competition. Indeed, a combination of optical stimulation of mesolimbic DA neurons during individual training and during a social competition test for palatable rewards resulted in improved performance on this test.
Collapse
Affiliation(s)
- Laura Lozano-Montes
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Simone Astori
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Sonia Abad
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Isabelle Guillot de Suduiraut
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Ioannis Zalachoras
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
29
|
Juarez B, Liu Y, Zhang L, Han MH. Optogenetic investigation of neural mechanisms for alcohol-use disorder. Alcohol 2019; 74:29-38. [PMID: 30621856 DOI: 10.1016/j.alcohol.2018.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/17/2018] [Accepted: 05/07/2018] [Indexed: 11/16/2022]
Abstract
Optogenetic techniques have been widely used in the study of neuropsychiatric diseases such as anxiety, depression, and drug addiction. Cell-type specific targeting of optogenetic tools to neurons has contributed to a tremendous understanding of the function of neural circuits for future treatment of neuropsychiatric disorders. Though optogenetics has been widely used in many research areas, the use of optogenetic tools to uncover and elucidate neural circuit mechanisms of alcohol's actions in the brain are still developing. Here in this review article, we will provide a basic introduction to optogenetics and discuss how these optogenetic experimental approaches can be used in alcohol studies to reveal neural circuit mechanisms of alcohol's actions in regions implicated in the development of alcohol addiction.
Collapse
Affiliation(s)
- Barbara Juarez
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Yutong Liu
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Key Laboratory of Functional Proteomics of Guangdong Province, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Lu Zhang
- Key Laboratory of Functional Proteomics of Guangdong Province, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Ming-Hu Han
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
30
|
Chemogenetic activation of ventral tegmental area GABA neurons, but not mesoaccumbal GABA terminals, disrupts responding to reward-predictive cues. Neuropsychopharmacology 2019; 44:372-380. [PMID: 29875446 PMCID: PMC6300533 DOI: 10.1038/s41386-018-0097-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 05/03/2018] [Accepted: 05/05/2018] [Indexed: 12/19/2022]
Abstract
Cues predicting rewards can gain motivational properties and initiate reward-seeking behaviors. Dopamine projections from the ventral tegmental area (VTA) to the nucleus accumbens (NAc) are critical in regulating cue-motivated responding. Although, approximately one third of mesoaccumbal projection neurons are GABAergic, it is unclear how this population influences motivational processes and cue processing. This is largely due to our inability to pharmacologically probe circuit level contributions of VTA-GABA, which arises from diverse sources, including multiple GABA afferents, interneurons, and projection neurons. Here we used a combinatorial viral vector approach to restrict activating Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) to GABA neurons in the VTA of wild-type rats trained to respond during a distinct audiovisual cue for sucrose. We measured different aspects of motivation for the cue or primary reinforcer, while chemogenetically activating either the VTA-GABA neurons or their projections to the NAc. Activation of VTA-GABA neurons decreased cue-induced responding and accuracy, while increasing latencies to respond to the cue and obtain the reward. Perseverative and spontaneous responses decreased, yet the rats persisted in entering the reward cup when the cue and reward were absent. However, activation of the VTA-GABA terminals in the accumbens had no effect on any of these behaviors. Together, we demonstrate that VTA-GABA neuron activity preferentially attenuates the ability of cues to trigger reward-seeking, while some aspects of the motivation for the reward itself are preserved. Additionally, the dense VTA-GABA projections to the NAc do not influence the motivational salience of the cue.
Collapse
|
31
|
Borland JM, Rilling JK, Frantz KJ, Albers HE. Sex-dependent regulation of social reward by oxytocin: an inverted U hypothesis. Neuropsychopharmacology 2019; 44:97-110. [PMID: 29968846 PMCID: PMC6235847 DOI: 10.1038/s41386-018-0129-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/10/2018] [Accepted: 06/15/2018] [Indexed: 12/17/2022]
Abstract
The rewarding properties of social interactions are essential for the expression of social behavior and the development of adaptive social relationships. Here, we review sex differences in social reward, and more specifically, how oxytocin (OT) acts in the mesolimbic dopamine system (MDS) to mediate the rewarding properties of social interactions in a sex-dependent manner. Evidence from rodents and humans suggests that same-sex social interactions may be more rewarding in females than in males. We propose that there is an inverted U relationship between OT dose, social reward, and neural activity within structures of the MDS in both males and females, and that this dose-response relationship is initiated at lower doses in females than males. As a result, depending on the dose of OT administered, OT could reduce social reward in females, while enhancing it in males. Sex differences in the neural mechanisms regulating social reward may contribute to sex differences in the incidence of a large number of psychiatric and neurodevelopmental disorders. This review addresses the potential significance of a sex-dependent inverted U dose-response function for OT's effects on social reward and in the development of gender-specific therapies for these disorders.
Collapse
Affiliation(s)
- Johnathan M Borland
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - James K Rilling
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, USA
- Anthropology, Emory University, Atlanta, GA, USA
- Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
- Center for Translational and Social Neuroscience, Emory University, Atlanta, GA, USA
| | - Kyle J Frantz
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - H Elliott Albers
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, USA.
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
32
|
Wiss DA, Avena N, Rada P. Sugar Addiction: From Evolution to Revolution. Front Psychiatry 2018; 9:545. [PMID: 30464748 PMCID: PMC6234835 DOI: 10.3389/fpsyt.2018.00545] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 10/12/2018] [Indexed: 12/12/2022] Open
Abstract
The obesity epidemic has been widely publicized in the media worldwide. Investigators at all levels have been looking for factors that have contributed to the development of this epidemic. Two major theories have been proposed: (1) sedentary lifestyle and (2) variety and ease of inexpensive palatable foods. In the present review, we analyze how nutrients like sugar that are often used to make foods more appealing could also lead to habituation and even in some cases addiction thereby uniquely contributing to the obesity epidemic. We review the evolutionary aspects of feeding and how they have shaped the human brain to function in "survival mode" signaling to "eat as much as you can while you can." This leads to our present understanding of how the dopaminergic system is involved in reward and its functions in hedonistic rewards, like eating of highly palatable foods, and drug addiction. We also review how other neurotransmitters, like acetylcholine, interact in the satiation processes to counteract the dopamine system. Lastly, we analyze the important question of whether there is sufficient empirical evidence of sugar addiction, discussed within the broader context of food addiction.
Collapse
Affiliation(s)
- David A. Wiss
- Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, United States
| | - Nicole Avena
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Pedro Rada
- School of Medicine, University of Los Andes, Mérida, Venezuela
| |
Collapse
|
33
|
Issa HA, Staes N, Diggs-Galligan S, Stimpson CD, Gendron-Fitzpatrick A, Taglialatela JP, Hof PR, Hopkins WD, Sherwood CC. Comparison of bonobo and chimpanzee brain microstructure reveals differences in socio-emotional circuits. Brain Struct Funct 2018; 224:239-251. [DOI: 10.1007/s00429-018-1751-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/09/2018] [Indexed: 12/24/2022]
|
34
|
Fortin SM, Roitman MF. Challenges to Body Fluid Homeostasis Differentially Recruit Phasic Dopamine Signaling in a Taste-Selective Manner. J Neurosci 2018; 38:6841-6853. [PMID: 29934352 PMCID: PMC6070668 DOI: 10.1523/jneurosci.0399-18.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 06/01/2018] [Accepted: 06/05/2018] [Indexed: 01/13/2023] Open
Abstract
The internal environment of an organism must remain stable to ensure optimal performance and ultimately survival. The generation of motivated behaviors is an adaptive mechanism for defending homeostasis. Although physiological state modulates motivated behaviors, the influence of physiological state on phasic dopamine signaling, an underlying neurobiological substrate of reward-driven behavior, is underexplored. Here, we use sodium depletion and water restriction, manipulations of body fluid homeostasis, to determine the flexibility and specificity of dopamine responses. Changes in dopamine concentration were measured using fast-scan cyclic voltammetry in the nucleus accumbens shell of male rats in response to intraoral infusions of fluids that either satisfied or did not satisfy homeostatic need. Increases in dopamine concentration during intraoral infusions were observed only under conditions of physiological deficit. Furthermore, dopamine increases were selective and limited to those that satisfied the need state of the animal. Thus, dopamine neurons track fluid balance and respond to salt and water stimuli in a state- and taste-dependent manner. Using Fluoro-Gold tracing and immunohistochemistry for c-Fos and Foxp2, a marker of sodium-deprivation responsive neurons, we revealed brainstem populations of neurons that are activated by sodium depletion and project directly to the ventral tegmental area. The identified projections may modulate dopamine neuron excitability and consequently the state-specific dopamine release observed in our experiments. This work illustrates the impact of physiological state on mesolimbic dopamine signaling and a potential circuit by which homeostatic disruptions are communicated to mesolimbic circuitry to drive the selective reinforcement of biologically-required stimuli under conditions of physiological need.SIGNIFICANCE STATEMENT Motivated behaviors arise during physiological need and are highly selective for homeostasis-restoring stimuli. Although phasic dopamine signaling has been shown to contribute to the generation of motivated behaviors, the state and stimulus specificity of phasic dopamine signaling is less clear. These studies use thirst and sodium appetite to show that dopamine neurons dynamically track body fluid homeostasis and respond to water and salt stimuli in a state- and taste-dependent manner. We also identify hindbrain sodium deprivation-responsive neurons that project directly to the ventral tegmental area, where dopamine neuron cell bodies reside. This work demonstrates command of homeostasis over dopamine signaling and proposes a circuit by which physiological need drives motivated behavior by state- and taste-selective recruitment of phasic dopamine signaling.
Collapse
Affiliation(s)
| | - Mitchell F Roitman
- Graduate Program in Neuroscience and
- Department of Psychology, University of Illinois at Chicago (UIC), Chicago, Illinois, 60607
| |
Collapse
|
35
|
Considerations for the use of virally delivered genetic tools for in-vivo circuit analysis and behavior in mutant mice: a practical guide to optogenetics. Behav Pharmacol 2018; 28:598-609. [PMID: 29099403 DOI: 10.1097/fbp.0000000000000361] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Optogenetics was the method of the year in 2010 according to Nature Neuroscience. Since then, this method has become widespread, the use of virally delivered genetic tools has extended to other fields such as pharmacogenetics, and optogenetic techniques have become frequently applied in genetically manipulated animals for in-vivo circuit analysis and behavioral studies. However, several issues should be taken into consideration when planning such experiments. We aimed to summarize the critical points concerning optogenetic manipulation of a specific brain area in mutant mice. First, the appropriate vector should be chosen to allow optimal optogenetic manipulation. Adeno-associated viral vectors are the most common carriers with different available serotypes. Light-sensitive channels are available in many forms, and the expression of the delivered genetic material can be influenced in many ways. Second, selecting the adequate stimulation protocol is also essential. The pattern, intensity, and timing could be determinative parameters. Third, the mutant strain might have a phenotype that influences the observed behavior. In conclusion, detailed preliminary experiments and numerous control groups are required to choose the best vector and stimulation protocol and to ensure that the mutant animals do not have a specific phenotype that can influence the examined behavior.
Collapse
|
36
|
Phasic Stimulation of Midbrain Dopamine Neuron Activity Reduces Salt Consumption. eNeuro 2018; 5:eN-NWR-0064-18. [PMID: 29766048 PMCID: PMC5952649 DOI: 10.1523/eneuro.0064-18.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 03/12/2018] [Accepted: 03/13/2018] [Indexed: 01/13/2023] Open
Abstract
Salt intake is an essential dietary requirement, but excessive consumption is implicated in hypertension and associated conditions. Little is known about the neural circuit mechanisms that control motivation to consume salt, although the midbrain dopamine system, which plays a key role in other reward-related behaviors, has been implicated. We, therefore, examined the effects on salt consumption of either optogenetic excitation or chemogenetic inhibition of ventral tegmental area (VTA) dopamine neurons in male mice. Strikingly, optogenetic excitation of dopamine neurons decreased salt intake in a rapid and reversible manner, despite a strong salt appetite. Importantly, optogenetic excitation was not aversive, did not induce hyperactivity, and did not alter salt concentration preferences in a need-free state. In addition, we found that chemogenetic inhibition of dopamine neurons had no effect on salt intake. Lastly, optogenetic excitation of dopamine neurons reduced consumption of sucrose following an overnight fast, suggesting a more general role of VTA dopamine neuron excitation in organizing motivated behaviors.
Collapse
|
37
|
Gioia DA, Xu M, Wayman WN, Woodward JJ. Effects of drugs of abuse on channelrhodopsin-2 function. Neuropharmacology 2018; 135:316-327. [PMID: 29580953 DOI: 10.1016/j.neuropharm.2018.03.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 01/16/2023]
Abstract
Channelrhodopsins are light activated ion channels used extensively over the past decade to probe the function of genetically defined neuronal populations and distinct neural circuits with high temporal and spatial precision. The widely used Channelrhodopsin-2 variant (ChR2) is an excitatory opsin that undergoes conformational changes in response to blue light, allowing non-selective passage of protons and cations across the plasma membrane thus leading to depolarization. In the addiction neuroscience field, opsins such as ChR2 provide a means to disambiguate the overlapping circuitry involved in mediating the reinforcing and aversive effects of drugs of abuse as well as to determine the plasticity that can occur in these circuits during the development of dependence. Although ChR2 has been widely used in animal models of drug and alcohol self-administration, direct effects of drugs of abuse on ChR2 function may confound its use and lead to misinterpretation of data. As a variety of neuronal ion channels are primary targets of various drugs of abuse, it is critical to determine whether ChR2-mediated currents are modulated by these drugs. In this study, we performed whole-cell electrophysiological recordings in HEK293 cells expressing the commonly used ChR2(H134R) variant and examined the effects of various drugs of abuse and other commonly used agents on light-induced currents. We found no differences in ChR2-mediated currents in the presence of 30 μM nicotine, 30 μM cocaine, 100 μM methamphetamine or 3 mM toluene. Similarly, ChR2 currents were insensitive to 30 mM ethanol but higher concentrations (100-300 mM) produced significant effects on the desensitization and amplitude of light-evoked currents. Tetrahydrocannabinol (1-10 μM) and morphine (30-100 μM) significantly inhibited ChR2 currents while the cannabinoid receptor antagonist AM-251 had no effect. The sodium channel blocker tetrodotoxin (5 μM) and the generic channel blocker/contrast agent gadolinium chloride (10 mM) also reduced ChR2 currents while the divalent ion magnesium (10 mM) had no effect. Together, the results from this study highlight the importance of conducting appropriate control experiments when testing new compounds in combination with optogenetic approaches.
Collapse
Affiliation(s)
- Dominic A Gioia
- Medical University of South Carolina, Institute of Psychiatry, 67 President St, Charleston, SC, 29425, United States.
| | - Minfu Xu
- Medical University of South Carolina, Institute of Psychiatry, 67 President St, Charleston, SC, 29425, United States.
| | - Wesley N Wayman
- Medical University of South Carolina, Institute of Psychiatry, 67 President St, Charleston, SC, 29425, United States.
| | - John J Woodward
- Medical University of South Carolina, Institute of Psychiatry, 67 President St, Charleston, SC, 29425, United States.
| |
Collapse
|
38
|
The Effects of the Monoamine Stabilizer (-)-OSU6162 on Binge-Like Eating and Cue-Controlled Food-Seeking Behavior in Rats. Neuropsychopharmacology 2018; 43:617-626. [PMID: 28895569 PMCID: PMC5770763 DOI: 10.1038/npp.2017.215] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/28/2017] [Accepted: 09/06/2017] [Indexed: 02/02/2023]
Abstract
Binge-eating disorder (BED) is characterized by recurring episodes of excessive consumption of palatable food and an increased sensitivity to food cues. Patients with BED display an addiction-like symptomatology and the dopamine system might be a potential treatment target. The clinically safe monoamine stabilizer (-)-OSU6162 (OSU6162) restores dopaminergic dysfunction in long-term alcohol-drinking rats and shows promise as a novel treatment for alcohol use disorder. Here, the effects of OSU6162 on consummatory (binge-like eating) and appetitive (cue-controlled seeking) behavior motivated by chocolate-flavored sucrose pellets were evaluated in non-food-restricted male Lister Hooded rats. OSU6162 significantly reduced binge-like intake of chocolate-flavored sucrose pellets without affecting prior chow intake. Furthermore, OSU6162 significantly reduced the cue-controlled seeking of chocolate-flavored sucrose pellets under a second-order schedule of reinforcement before, but not after, the delivery and ingestion of reward, indicating a selective effect on incentive motivational processes. In contrast, the dopamine D2/D3 receptor antagonist raclopride reduced the seeking of chocolate-flavored sucrose pellets both pre- and post reward ingestion and also reduced responding under simpler schedules of seeking behavior. The D1/5 receptor antagonist SCH23390 had no effect on instrumental behavior under any reinforcement schedule tested. Finally, local administration of OSU6162 into the nucleus accumbens core, but not dorsolateral striatum, selectively reduced cue-controlled sucrose seeking. In conclusion, the present results show that OSU6162 reduces binge-like eating behavior and attenuates the impact of cues on seeking of palatable food. This indicates that OSU6162 might serve as a novel BED medication.
Collapse
|
39
|
Millan EZ, Kim HA, Janak PH. Optogenetic activation of amygdala projections to nucleus accumbens can arrest conditioned and unconditioned alcohol consummatory behavior. Neuroscience 2017; 360:106-117. [PMID: 28757250 PMCID: PMC5752133 DOI: 10.1016/j.neuroscience.2017.07.044] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/13/2017] [Accepted: 07/19/2017] [Indexed: 11/25/2022]
Abstract
Following a Pavlovian pairing procedure, alcohol-paired cues come to elicit behavioral responses that lead to alcohol consumption. Here we used an optogenetic approach to activate basolateral amygdala (BLA) axonal terminals targeting the shell of nucleus accumbens (AcbSh) and investigated a possible influence over cue-conditioned alcohol seeking and alcohol drinking, based on the demonstrated roles of these areas in behavioral responding to Pavlovian cues and in feeding behavior. Rats were trained to anticipate alcohol or sucrose following the onset of a discrete conditioned stimulus (CS). Channelrhodopsin-mediated activation of the BLA-to-AcbSh pathway concurrent with each CS disrupted cued alcohol seeking. Activation of the same pathway caused rapid cessation of alcohol drinking from a sipper tube. Neither effect was accompanied by an overall change in locomotion. Finally, the suppressive effect of photoactivation on cued-triggered seeking was also evidenced in animals trained with sucrose. Together these findings suggest that photoactivation of BLA terminals in the AcbSh can override the conditioned motivational properties of reward-predictive cues as well as unconditioned consummatory responses necessary for alcohol drinking. The findings provide evidence for a limbic-striatal influence over motivated behavior for orally consumed rewards, including alcohol.
Collapse
Affiliation(s)
- E Zayra Millan
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore MD 21218, United States.
| | - H Amy Kim
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore MD 21218, United States
| | - Patricia H Janak
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore MD 21218, United States; Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore MD 21205, United States.
| |
Collapse
|
40
|
The Relevance of AgRP Neuron-Derived GABA Inputs to POMC Neurons Differs for Spontaneous and Evoked Release. J Neurosci 2017; 37:7362-7372. [PMID: 28667175 DOI: 10.1523/jneurosci.0647-17.2017] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 06/14/2017] [Accepted: 06/21/2017] [Indexed: 12/25/2022] Open
Abstract
Hypothalamic agouti-related peptide (AgRP) neurons potently stimulate food intake, whereas proopiomelanocortin (POMC) neurons inhibit feeding. Whether AgRP neurons exert their orexigenic actions, at least in part, by inhibiting anorexigenic POMC neurons remains unclear. Here, the connectivity between GABA-releasing AgRP neurons and POMC neurons was examined in brain slices from male and female mice. GABA-mediated spontaneous IPSCs (sIPSCs) in POMC neurons were unaffected by disturbing GABA release from AgRP neurons either by cell type-specific deletion of the vesicular GABA transporter or by expression of botulinum toxin in AgRP neurons to prevent vesicle-associated membrane protein 2-dependent vesicle fusion. Additionally, there was no difference in the ability of μ-opioid receptor (MOR) agonists to inhibit sIPSCs in POMC neurons when MORs were deleted from AgRP neurons, and activation of the inhibitory designer receptor hM4Di on AgRP neurons did not affect sIPSCs recorded from POMC neurons. These approaches collectively indicate that AgRP neurons do not significantly contribute to the strong spontaneous GABA input to POMC neurons. Despite these observations, optogenetic stimulation of AgRP neurons reliably produced evoked IPSCs in POMC neurons, leading to the inhibition of POMC neuron firing. Thus, AgRP neurons can potently affect POMC neuron function without contributing a significant source of spontaneous GABA input to POMC neurons. Together, these results indicate that the relevance of GABAergic inputs from AgRP to POMC neurons is state dependent and highlight the need to consider different types of transmitter release in circuit mapping and physiologic regulation.SIGNIFICANCE STATEMENT Agouti-related peptide (AgRP) neurons play an important role in driving food intake, while proopiomelanocortin (POMC) neurons inhibit feeding. Despite the importance of these two well characterized neuron types in maintaining metabolic homeostasis, communication between these cells remains poorly understood. To provide clarity to this circuit, we made electrophysiological recordings from mouse brain slices and found that AgRP neurons do not contribute spontaneously released GABA onto POMC neurons, although when activated with channelrhodopsin AgRP neurons inhibit POMC neurons through GABA-mediated transmission. These findings indicate that the relevance of AgRP to POMC neuron GABA connectivity depends on the state of AgRP neuron activity and suggest that different types of transmitter release should be considered when circuit mapping.
Collapse
|
41
|
Altered sucrose self-administration following injection of melanocortin receptor agonists and antagonists into the ventral tegmental area. Psychopharmacology (Berl) 2017; 234:1683-1692. [PMID: 28243712 DOI: 10.1007/s00213-017-4570-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 02/10/2017] [Indexed: 12/26/2022]
Abstract
RATIONALE AND OBJECTIVES Alpha-melanocyte stimulating hormone (αMSH) and agouti-related protein (AgRP) are antagonistic neuropeptides that play an important role in the control of feeding and body weight through their central actions on the melanocortin-3 and melanocortin-4 receptors. Increasing evidence indicates that αMSH and AgRP can interact with the mesolimbic dopamine system to regulate feeding as well as other behaviors. For example, we have shown previously that injection of melanocortin receptor agonists and antagonists into the ventral tegmental area (VTA) alters both normal home-cage feeding and the intake of sucrose solutions, but it remains unknown whether αMSH and AgRP can also act in the VTA to affect reward-related feeding. METHODS We tested whether injection of the melanocortin receptor agonist, MTII, or the melanocortin receptor antagonist, SHU9119, directly into the VTA affected operant responding maintained by sucrose pellets in self-administration assays. RESULTS Injection of MTII into the VTA decreased operant responding maintained by sucrose pellets on both fixed ratio and progressive ratio schedules of reinforcement, whereas SHU9119 increased operant responding under fixed ratio, but not progressive ratio schedules. MTII also increased and SHU9119 decreased 24-h home-cage food intake. CONCLUSIONS This study demonstrates that αMSH and AgRP act in the VTA to affect sucrose self-administration. Thus, it adds critical information to the growing literature showing that in addition to their well-characterized role in controlling "need-based" feeding, αMSH and AgRP can also act on the mesolimbic dopamine system to control reward-related behavior.
Collapse
|
42
|
Cardozo Pinto DF, Lammel S. Viral vector strategies for investigating midbrain dopamine circuits underlying motivated behaviors. Pharmacol Biochem Behav 2017; 174:23-32. [PMID: 28257849 DOI: 10.1016/j.pbb.2017.02.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/07/2017] [Accepted: 02/23/2017] [Indexed: 12/21/2022]
Abstract
Midbrain dopamine (DA) neurons have received significant attention in brain research because of their central role in reward processing and their dysfunction in neuropsychiatric disorders such as Parkinson's disease, drug addiction, depression and schizophrenia. Until recently, it has been thought that DA neurons form a homogeneous population whose primary function is the computation of reward prediction errors. However, through the implementation of viral vector strategies, an unexpected complexity and diversity has been revealed at the anatomical, molecular and functional level. In this review, we discuss recent viral vector approaches that have been leveraged to dissect how different circuits involving distinct DA neuron subpopulations may contribute to the role of DA in reward- and aversion-related behaviors. We focus on studies that have used cell type- and projection-specific optogenetic manipulations, discuss the strengths and limitations of each approach, and critically examine emergent organizational principles that have led to a reclassification of midbrain DA neurons.
Collapse
Affiliation(s)
- Daniel F Cardozo Pinto
- Department of Molecular and Cell Biology, University of California, Berkeley, 142 Life Science Addition #3200, CA 94720, USA
| | - Stephan Lammel
- Department of Molecular and Cell Biology, University of California, Berkeley, 142 Life Science Addition #3200, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, 142 Life Science Addition #3200, CA 94720, USA.
| |
Collapse
|
43
|
Abstract
Many of the neurocircuits and hormones known to underlie the sensations of hunger and satiety also substantially alter the activity of the dopaminergic reward system. Much interest lies in the ways that hunger, satiety, and reward tie together, as the epidemic of obesity seems tied to the recent development and mass availability of highly palatable foods. In this review, we will first discuss the basic neurocircuitry of the midbrain and basal forebrain reward system. We will elaborate how several important mediators of hunger-the agouti-related protein neurons of the arcuate nucleus, the lateral hypothalamic nucleus, and ghrelin-enhance the sensitivity of the dopaminergic reward system. Then, we will elaborate how mediators of satiety-the nucleus tractus solitarius, pro-opiomelanocortin neurons of the arcuate nucleus, and its peripheral hormonal influences such as leptin-reduce the reward system sensitivity. We hope to provide a template by which future research may identify the ways in which highly rewarding foods bypass this balanced system to produce excessive food consumption.
Collapse
Affiliation(s)
- Ryan Michael Cassidy
- Brown Foundation of the Institute of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, Neuroscience Program MD Anderson Cancer Center and UTHealth Graduate School of Biological Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
- *Correspondence: Ryan Michael Cassidy,
| | - Qingchun Tong
- Brown Foundation of the Institute of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, Neuroscience Program MD Anderson Cancer Center and UTHealth Graduate School of Biological Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
44
|
Bell S, Kolobova I, Crapper L, Ernst C. Lesch-Nyhan Syndrome: Models, Theories, and Therapies. Mol Syndromol 2016; 7:302-311. [PMID: 27920633 PMCID: PMC5131334 DOI: 10.1159/000449296] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2016] [Indexed: 12/30/2022] Open
Abstract
Lesch-Nyhan syndrome (LNS) is a rare X-linked disorder caused by mutations in HPRT1, an important enzyme in the purine salvage pathway. Symptoms of LNS include dystonia, gout, intellectual disability, and self-mutilation. Despite having been characterized over 50 years ago, it remains unclear precisely how deficits in hypoxanthine and guanine recycling can lead to such a profound neurological phenotype. Several studies have proposed different hypotheses regarding the etiology of this disease, and several treatments have been tried in patients, though none have led to a satisfactory explanation of the disease. New technologies such as next-generation sequencing, optogenetics, genome editing, and induced pluripotent stem cells provide a unique opportunity to map the precise sequential pathways leading from genotype to phenotype.
Collapse
Affiliation(s)
| | | | | | - Carl Ernst
- Department of Psychiatry, Douglas Hospital Research Institute, McGill University, Montreal, Que., Canada
| |
Collapse
|
45
|
Namboodiri VMK, Stuber GD. Cell-Type-Specific Optogenetics in Monkeys. Cell 2016; 166:1366-1368. [PMID: 27610562 DOI: 10.1016/j.cell.2016.08.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The recent advent of technologies enabling cell-type-specific recording and manipulation of neuronal activity spurred tremendous progress in neuroscience. However, they have been largely limited to mice, which lack the richness in behavior of primates. Stauffer et al. now present a generalizable method for achieving cell-type specificity in monkeys.
Collapse
Affiliation(s)
- Vijay Mohan K Namboodiri
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Garret D Stuber
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA.
| |
Collapse
|