1
|
Wallace CW, Holleran KM, Slinkard CY, Centanni SW, Lapish CC, Jones SR. Kappa opioid receptors diminish spontaneous dopamine signals in awake mice through multiple mechanisms. Neuropharmacology 2025; 273:110458. [PMID: 40204058 DOI: 10.1016/j.neuropharm.2025.110458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 03/06/2025] [Accepted: 04/03/2025] [Indexed: 04/11/2025]
Abstract
The role of the dynorphin/kappa opioid receptor (KOR) system in dopamine (DA) regulation has been extensively investigated. KOR activation reduces extracellular DA concentrations, but the exact mechanism(s) through which this is accomplished are not fully elucidated. To explore KOR influences on real-time DA fluctuations, we used the photosensor dLight1.2 with fiber photometry in the nucleus accumbens (NAc) core of freely moving male and female C57BL/6J mice. First, we established that the rise and fall of spontaneously arising DA signals were due to DA release and reuptake, respectively. Next, mice were systemically administered the KOR agonist U50,488H in the presence or absence of the KOR antagonist aticaprant. U50,488H reduced both the amplitude and width of spontaneous signals in both sexes. Further, the slope of the correlation between amplitude and width was increased, indicating that DA uptake rates were increased. U50,488H also reduced the frequency of occurrence of signals in males and females. The effects of KOR activation were stronger in males, while effects of KOR antagonism were stronger in females. Overall, KORs exerted significant inhibitory control over spontaneous DA signaling, acting through at least three mechanisms - inhibiting DA release, promoting DA transporter-mediated uptake, and reducing the frequency of signals.
Collapse
Affiliation(s)
- Conner W Wallace
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Katherine M Holleran
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Clare Y Slinkard
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Samuel W Centanni
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Christopher C Lapish
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sara R Jones
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
2
|
McCoy K, Reed F, Conn K, Foldi CJ. Separate or inseparable? Serotonin and dopamine system interactions may underlie the therapeutic potential of psilocybin for anorexia nervosa. Physiol Behav 2025; 298:114957. [PMID: 40403997 DOI: 10.1016/j.physbeh.2025.114957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 04/16/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025]
Abstract
Psilocybin, a serotonergic psychedelic, has emerged as a promising treatment for a range of mental health conditions, including anorexia nervosa. Recent insights from animal models and human imaging studies suggest psilocybin enhances cognitive flexibility and modifies reward processing - two core processes disrupted in anorexia nervosa. Both cognitive flexibility and reward processing are highly dependent on interactions between serotonin (5-HT) and dopamine (DA) systems in key brain regions such as the prefrontal cortex and nucleus accumbens. Psilocybin's influence on neuroplasticity, particularly in promoting structural and functional changes in neural circuits, underpins its therapeutic potential. While its effects are predominantly attributed to activity of the 5-HT2A receptor subtype, recent evidence suggests a broader network of brain receptor interactions, particularly those with dopaminergic pathways, plays a crucial role. Investigations using rodent models reveal that psilocybin induces both rapid and enduring neuroplastic changes, improving cognitive flexibility through these complex neurochemical mechanisms. Advances in real-time in vivo neurochemical recording now allow simultaneous monitoring of 5-HT and DA signalling, which will provide essential insights into their distinct and coordinated actions during cognitive performance. This integrative framework highlights the need for further research into psilocybin's dual modulation of 5-HT and DA systems to optimize its therapeutic applications for anorexia nervosa, a life-threatening condition that is characterized by impairments in cognitive flexibility and reward processing.
Collapse
Affiliation(s)
- Kaspar McCoy
- Monash University, Department of Physiology, 26 Innovation Walk, 3800 Clayton, Australia; Biomedicine Discovery Institute, Monash University, 23 Innovation Walk, 3800 Clayton, Australia
| | - Felicia Reed
- Monash University, Department of Physiology, 26 Innovation Walk, 3800 Clayton, Australia; Biomedicine Discovery Institute, Monash University, 23 Innovation Walk, 3800 Clayton, Australia; Australian Eating Disorders Research & Translation Centre (AEDRTC), Sydney, NSW, Australia
| | - Kyna Conn
- Monash University, Department of Physiology, 26 Innovation Walk, 3800 Clayton, Australia; Biomedicine Discovery Institute, Monash University, 23 Innovation Walk, 3800 Clayton, Australia
| | - Claire J Foldi
- Monash University, Department of Physiology, 26 Innovation Walk, 3800 Clayton, Australia; Biomedicine Discovery Institute, Monash University, 23 Innovation Walk, 3800 Clayton, Australia.
| |
Collapse
|
3
|
Zhou S, Zhu Y, Du A, Niu S, Du Y, Yang Y, Chen W, Du S, Sun L, Liu Y, Wu H, Lou H, Li XM, Duan S, Yang H. A midbrain circuit mechanism for noise-induced negative valence coding. Nat Commun 2025; 16:4610. [PMID: 40382338 PMCID: PMC12085634 DOI: 10.1038/s41467-025-59956-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 05/06/2025] [Indexed: 05/20/2025] Open
Abstract
Unpleasant sounds elicit a range of negative emotional reactions, yet the underlying neural mechanisms remain largely unknown. Here we show that glutamatergic neurons in the central inferior colliculus (CICglu) relay noise information to GABAergic neurons in the ventral tegmental area (VTAGABA) via the cuneiform nucleus (CnF), encoding negative emotions in mice. In contrast, the CICglu→medial geniculate (MG) canonical auditory pathway processes salient stimuli. By combining viral tracing, calcium imaging, and optrode recording, we demonstrate that the CnF acts downstream of CICglu to convey negative valence to the mesolimbic dopamine system by activating VTAGABA neurons. Optogenetic or chemogenetic inhibition of any connection within the CICglu→CnFglu → VTAGABA circuit, or direct excitation of the mesolimbic dopamine (DA) system is sufficient to alleviate noise-induced negative emotion perception. Our findings highlight the significance of the CICglu→CnFglu → VTAGABA circuit in coping with acoustic stressors.
Collapse
Affiliation(s)
- Siyao Zhou
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Yuebin Zhu
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Ana Du
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Shuai Niu
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Yonglan Du
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Yan Yang
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenqiang Chen
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
- Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Siyu Du
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Li Sun
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Yijun Liu
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Hangjun Wu
- Department of Pathology of Sir Run Run Shaw Hospital; Center of Cryo-Electron Microscopy, Zhejiang University, Hangzhou, China
| | - Huifang Lou
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Xiao-Ming Li
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Nanhu Brain-Computer Interface Institute, Hangzhou, China
| | - Shumin Duan
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Hongbin Yang
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China.
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China.
| |
Collapse
|
4
|
Sköld J, Lazzarino GP, Nett M, Wiskerke J, Engblom D. Melanocortin 4 receptor-expressing neurons in the lateral stripe of the striatum regulate affect and motor control. iScience 2025; 28:112456. [PMID: 40352729 PMCID: PMC12063154 DOI: 10.1016/j.isci.2025.112456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/04/2024] [Accepted: 04/11/2025] [Indexed: 05/14/2025] Open
Abstract
The dopaminergic system is crucial for affect regulation. Melanocortin 4 receptors (MC4R) in the ventral striatum have been shown to be necessary for establishing aversive states. Here, we functionally characterize MC4R-expressing striatal neurons in mice. MC4Rs were enriched in atypical dopamine receptor 1 (D1) neurons in the lateral stripe of the striatum (LSS), an understudied area in the ventrolateral striatum. Fiber photometry recordings showed that MC4R neuron activity and local dopamine release in the LSS increased in response to both rewarding and aversive stimuli. Moreover, MC4R neuronal activity and glutamate release in the LSS correlated with body movement. Optogenetic activation of MC4R-LSS neurons was rewarding in a real-time place preference test and a self-stimulation paradigm, increased locomotor activity, and induced striatal dopamine release. Collectively, our findings suggest that MC4R-LSS neurons are activated by stimuli of both rewarding and aversive character and that they induce positive affect, dopamine release and locomotion.
Collapse
Affiliation(s)
- Johan Sköld
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Gisela Paola Lazzarino
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Myra Nett
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Joost Wiskerke
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - David Engblom
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
5
|
Yang MD, Cun XF, Wu N, Li J, Song R. Dopamine D3 receptor in the nucleus accumbens modulates opioid taking and seeking in mice. Prog Neuropsychopharmacol Biol Psychiatry 2025; 139:111389. [PMID: 40324674 DOI: 10.1016/j.pnpbp.2025.111389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/26/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
Accumulating preclinical evidence suggests that selective antagonists of dopamine receptor D3 (Drd3) affects opioid-induced addictive behaviors across various animal models, highlighting Drd3 as a potential therapeutic target for opioid use disorders. However, the cellular type and neural circuit mechanisms by which Drd3 mediates these effects remains unclear. We employed YQA14, a selective antagonist and knock-out to selectively block or delete Drd3 in the nucleus accumbens (NAc) or ventral tegmental area (VTA). We utilized a battery of morphine-induced self-administration assays, fiber photometry, RNAscope in situ hybridization and RT-PCR to functionally characterize the roles of antagonists of Drd3s in the morphine actions. Our results revealed Drd3 mRNA expression in approximately 80 % of vesicular GABA transporter 1 (VGAT1)-positive GABA neurons in the NAc and approximately 50 % of tyrosine hydroxylase (TH)-positive dopamine neurons in the VTA. Strikingly, microinjections of YQA14 into the NAc, rather than the VTA, inhibited morphine taking and cue-induced drug-seeking. Transgenic down-regulation of Drd3 gene expression in the NAc yielded similar results. To explore the dopamine-dependent mechanism underlying Drd3's action, we found that intra-NAc microinjections of YQA14 significantly reduced morphine- or cue-induced activation of dopamine neurons in the VTA during morphine self-administration or cue-induced drug-seeking tests. These results suggest that YQA14 effectively reduces opioid taking and seeking, mainly by blocking Drd3 in the NAc, which subsequently inhibits VTA dopamine neuron activity and opioid action in dopamine transmission.
Collapse
Affiliation(s)
- Meng-Die Yang
- Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China
| | - Xing-Fang Cun
- Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China
| | - Ning Wu
- Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China
| | - Jin Li
- Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China.
| | - Rui Song
- Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China.
| |
Collapse
|
6
|
Vieitas-Gaspar N, Soares-Cunha C, Rodrigues AJ. From valence encoding to motivated behavior: A focus on the nucleus accumbens circuitry. Neurosci Biobehav Rev 2025; 172:106125. [PMID: 40154653 DOI: 10.1016/j.neubiorev.2025.106125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/21/2025] [Accepted: 03/23/2025] [Indexed: 04/01/2025]
Abstract
How do our brains determine whether something is good or bad? The brain's ability to evaluate stimuli as positive or negative - by attributing valence - is fundamental to survival and decision-making. Different brain regions have been associated with valence encoding, including the nucleus accumbens (NAc). The NAc is predominantly composed of GABAergic medium spiny neurons (MSNs), which segregate into two distinct populations based on their dopamine receptor expression: D1-receptor-expressing (D1-MSNs) and D2-receptor-expressing neurons (D2-MSNs). Classical models propose a binary functional role, where D1-MSNs exclusively mediated reward and positive valence, while D2-MSNs processed aversion and negative valence. However, we now recognize that NAc MSN subpopulations operate in a more complex manner than previously thought, often working cooperatively rather than antagonistically in valence-related behaviors. This review synthesizes our current knowledge of valence-encoding neurocircuitry, with emphasis on the NAc. We examine electrophysiological, calcium imaging, optogenetic, chemogenetic and pharmacological studies detailing the contribution of NAc medium spiny neurons for rewarding and aversive responses. Finally, we explore emerging technical innovations that promise to advance our understanding of how the mammalian brain encodes valence and translates it into behavior.
Collapse
Affiliation(s)
- Natacha Vieitas-Gaspar
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Carina Soares-Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Ana João Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| |
Collapse
|
7
|
Xie W, Li Y, Wang X, Blokhina E, Krupitsky E, Vetrova M, Hu J, Yuan T, Chen J, Wang H, Chen X. GABA B Receptor: Structure, Biological Functions, and Therapy for Diseases. MedComm (Beijing) 2025; 6:e70163. [PMID: 40242161 PMCID: PMC12000685 DOI: 10.1002/mco2.70163] [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: 07/09/2024] [Revised: 12/29/2024] [Accepted: 02/19/2025] [Indexed: 04/18/2025] Open
Abstract
Gamma-aminobutyric acid (GABA) B receptors (GABABRs) that acts slowly and maintains the inhibitory tone are versatile regulators in the complex nervous behaviors and their involvement in various neuropsychiatric disorders, such as anxiety, epilepsy, pain, drug addiction, and Alzheimer's disease. Additional study advances have implied the crucial roles of GABABRs in regulating feeding-related behaviors, yet their therapeutic potential in addressing the neuropsychiatric disorders, binge eating, and feeding-related disorders remains underutilized. This general review summarized the physiological structure and functions of GABABR, explored the regulation in various psychiatric disorders, feeding behaviors, binge eating, and metabolism disorders, and fully discussed the potential of targeting GABABRs and its regulator-binding sites for the treatment of different psychiatric disorders, binge eating and even obesity. While agonists that directly bind to GABABR1 have some negative side effects, positive allosteric modulators (PAMs) that bind to GABABR2 demonstrate excellent therapeutic efficacy and tolerability and have better safety and therapeutic indexes. Moreover, phosphorylation sites of downstream GABABRs regulators may be novel therapeutic targets for psychiatric disorders, binge eating, and obesity. Further studies, clinical trials in particular, will be essential for confirming the therapeutic value of PAMs and other agents targeting the GABABR pathways in a clinical setting.
Collapse
Affiliation(s)
- Weijie Xie
- Clinical Research Center for Mental Disorders, Shanghai Pudong New Area Mental Health CenterTongji University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health CenterShanghai Jiaotong University School of MedicineShanghaiChina
| | - Yuan Li
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health CenterShanghai Jiaotong University School of MedicineShanghaiChina
| | - Xinyue Wang
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health CenterShanghai Jiaotong University School of MedicineShanghaiChina
| | - Elena Blokhina
- Valdman Institute of PharmacologyPavlov UniversitySt. PetersburgRussia
| | - Evgeny Krupitsky
- Valdman Institute of PharmacologyPavlov UniversitySt. PetersburgRussia
- Bekhterev National Medical Research Center for Psychiatry and NeurologySt. PetersburgRussia
| | - Marina Vetrova
- Valdman Institute of PharmacologyPavlov UniversitySt. PetersburgRussia
| | - Ji Hu
- ShanghaiTech UniversityShanghaiChina
| | - Ti‐Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health CenterShanghai Jiaotong University School of MedicineShanghaiChina
- Co‐innovation Center of NeuroregenerationNantong UniversityNantongJiangsuChina
| | - Jue Chen
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health CenterShanghai Jiaotong University School of MedicineShanghaiChina
| | - Hua Wang
- Department of OncologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Xiangfang Chen
- Department of EndocrinologySecond Affiliated Hospital of Naval Medical UniversityShanghaiChina
| |
Collapse
|
8
|
Gazit Shimoni N, Tose AJ, Seng C, Jin Y, Lukacsovich T, Yang H, Verharen JPH, Liu C, Tanios M, Hu E, Read J, Tang LW, Lim BK, Tian L, Földy C, Lammel S. Changes in neurotensin signalling drive hedonic devaluation in obesity. Nature 2025; 641:1238-1247. [PMID: 40140571 PMCID: PMC12119351 DOI: 10.1038/s41586-025-08748-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 02/06/2025] [Indexed: 03/28/2025]
Abstract
Calorie-rich foods, particularly those that are high in fat and sugar, evoke pleasure in both humans and animals1. However, prolonged consumption of such foods may reduce their hedonic value, potentially contributing to obesity2-4. Here we investigated this phenomenon in mice on a chronic high-fat diet (HFD). Although these mice preferred high-fat food over regular chow in their home cages, they showed reduced interest in calorie-rich foods in a no-effort setting. This paradoxical decrease in hedonic feeding has been reported previously3-7, but its neurobiological basis remains unclear. We found that in mice on regular diet, neurons in the lateral nucleus accumbens (NAcLat) projecting to the ventral tegmental area (VTA) encoded hedonic feeding behaviours. In HFD mice, this behaviour was reduced and uncoupled from neural activity. Optogenetic stimulation of the NAcLat→VTA pathway increased hedonic feeding in mice on regular diet but not in HFD mice, though this behaviour was restored when HFD mice returned to a regular diet. HFD mice exhibited reduced neurotensin expression and release in the NAcLat→VTA pathway. Furthermore, neurotensin knockout in the NAcLat and neurotensin receptor blockade in the VTA each abolished optogenetically induced hedonic feeding behaviour. Enhancing neurotensin signalling via overexpression normalized aspects of diet-induced obesity, including weight gain and hedonic feeding. Together, our findings identify a neural circuit mechanism that links the devaluation of hedonic foods with obesity.
Collapse
Affiliation(s)
- Neta Gazit Shimoni
- Department of Neuroscience and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Amanda J Tose
- Department of Neuroscience and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Charlotte Seng
- Brain Research Institute, Faculties of Medicine and Science, University of Zurich, Zürich, Switzerland
| | - Yihan Jin
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Davis, CA, USA
- Max Planck Florida Institute For Neuroscience, Jupiter, FL, USA
| | - Tamás Lukacsovich
- Brain Research Institute, Faculties of Medicine and Science, University of Zurich, Zürich, Switzerland
| | - Hongbin Yang
- Department of Neurobiology and Department of Affiliated Mental Health Center of Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Jeroen P H Verharen
- Department of Neuroscience and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Christine Liu
- Department of Neuroscience and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Michael Tanios
- Department of Neuroscience and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Eric Hu
- Department of Neuroscience and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Jonathan Read
- Department of Neuroscience and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Lilly W Tang
- Department of Neuroscience and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Byung Kook Lim
- Division of Biological Sciences, University of California San Diego, San Diego, CA, USA
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Davis, CA, USA
- Max Planck Florida Institute For Neuroscience, Jupiter, FL, USA
| | - Csaba Földy
- Brain Research Institute, Faculties of Medicine and Science, University of Zurich, Zürich, Switzerland
| | - Stephan Lammel
- Department of Neuroscience and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA.
| |
Collapse
|
9
|
De Paolis ML, Loffredo G, Krashia P, La Barbera L, Nobili A, Cauzzi E, Babicola L, Di Segni M, Coccurello R, Puglisi-Allegra S, Latagliata EC, D'Amelio M. Repetitive prefrontal tDCS activates VTA dopaminergic neurons, resulting in attenuation of Alzheimer's Disease-like deficits in Tg2576 mice. Alzheimers Res Ther 2025; 17:94. [PMID: 40301905 PMCID: PMC12039073 DOI: 10.1186/s13195-025-01736-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 04/07/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND Emerging evidence implicates early dysfunction of dopaminergic neurons in the Ventral Tegmental Area (VTA) as a key contributor to Alzheimer's Disease (AD) pathophysiology. Specifically, the VTA dopaminergic neurodegeneration and the consequent reduction of dopamine (DA) in mesocorticolimbic targets are associated with the onset of cognitive impairments and neuropsychiatric-like manifestations in AD animal models. Moreover, decreased midbrain volume and functional VTA disconnection are identified as predictors of accelerated progression from Mild Cognitive Impairment to AD-dementia in clinical populations. Given these findings, interventions capable of directly modulating VTA activity and augmenting DA release, despite the ongoing neurodegeneration, may hold therapeutic potential for mitigating DA-related deficits in AD. This study aims at evaluating the therapeutic potential of prefrontal transcranial Direct Current Stimulation (tDCS) in the Tg2576 mouse model of AD, exhibiting early VTA dopaminergic neurodegeneration. METHODS Repeated tDCS was applied to assess its ability to activate VTA DA neurons. We also evaluated tDCS effects on synaptic plasticity, cognitive and non-cognitive behaviours and AD-related pathology. Hippocampal DA release and Nucleus Accumbens (NAc) DA transporter (DAT) expression were measured. With immunohistochemistry we examined microglial density and morphological complexity at different disease stages. Additionally, intracellular amyloid-β (Aβ) levels and plaque burden were evaluated to determine the impact of tDCS on AD pathology. RESULTS Prefrontal tDCS enhanced the activity of VTA dopaminergic neurons, leading to increased hippocampal DA release and higher DAT levels in the NAc. The enhanced DA outflow is associated with restored CA3-CA1 synaptic plasticity and improvements in recognition memory and motivational behaviours. tDCS reduced microglial numbers and morphological complexity in Tg2576 mice at both pre-plaque stage (7-months) and at an advanced stage characterized by plaque accumulation (12-months). Notably, tDCS also decreased Aβ plaque burden, although no changes in intracellular Aβ levels were observed in younger Tg2576 mice. CONCLUSIONS These findings highlight the multifaceted therapeutic potential of prefrontal tDCS in targeting key AD pathophysiological hallmarks, including dopaminergic dysfunction, synaptic impairments, neuroinflammation and plaque deposition. As a non-invasive neuromodulatory approach, prefrontal tDCS emerges as a promising early intervention strategy to complement existing AD treatments, with the potential to improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Maria Luisa De Paolis
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
| | - Gilda Loffredo
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
| | - Paraskevi Krashia
- Department of Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy
| | - Livia La Barbera
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy
| | - Annalisa Nobili
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy
| | - Emma Cauzzi
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
| | - Lucy Babicola
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy
- Department of Psychology, Sapienza University of Rome, P.Le Aldo Moro, 5, 00185, Rome, Italy
| | - Matteo Di Segni
- Child Psychopathology Unit, IRCCS Eugenio Medea, Via Don Luigi Monza, 20, 23842, Bosisio Parini, Italy
| | - Roberto Coccurello
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy
- National Research Council (CNR), Institute for Complex System (ISC), Via Dei Taurini, 19, 00185, Rome, Italy
| | - Stefano Puglisi-Allegra
- Istituto Di Ricovero E Cura a Carattere Scientifico (IRCCS) Neuromed, Via Atinense, 18, 86077, Pozzilli, Italy
| | - Emanuele Claudio Latagliata
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy.
- Department of Psychology, International Telematic University Uninettuno, Corso Vittorio Emanuele II, 39, 00186, Rome, Italy.
| | - Marcello D'Amelio
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy.
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy.
| |
Collapse
|
10
|
Valenti O, Rekawek KA, Wieser S, Bulut H, Scholze P, Boehm S. Plasticity of ventral tegmental area disturbance during abstinence after repeated amphetamine exposure: restoration by selective activation of group II metabotropic glutamate receptors. Front Pharmacol 2025; 16:1534101. [PMID: 40337518 PMCID: PMC12055554 DOI: 10.3389/fphar.2025.1534101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/07/2025] [Indexed: 05/09/2025] Open
Abstract
Background and aims The psychostimulant actions of amphetamine (AMPH) have been correlated with its ability to orchestrate ventral tegmental area (VTA) dopamine (DA) neuron activity states and, thus, DA release in output regions: in rats, a single exposure is sufficient to reduce the fraction of spontaneously active DA neurons, i.e., DA neuron population activity, whereas AMPH abstinence after repeated exposure leads to an increase. Here, this switch in DA neuron activity was resolved in detail in mice, and its sensitivity towards activation of group II metabotropic glutamate receptor (mGluR2 and mGluR3) was investigated. Experimental procedure All experiments were conducted on C57BL/6J male mice. After repeated AMPH administration (2 mg/kg), the amine was withdrawn for up to 15 days and VTA DA neuron activity was assessed. The involvement VTA afferent regions with respect to AMPH actions was analyzed either by local instillation of drugs or through inactivation by tetrodotoxin. Selective agonists or allosteric modulators of mGluR2 and mGluR3 were used to explore whether group II mGluR might interfere with VTA disturbances caused by the amine. Results After repeated AMPH exposure, VTA DA neuron activity remained reduced for 4 days and then rose to a hyperdopaminergic state within 15 days. The initial hypodopaminergia was coordinated by an amygdala (AMG) - nucleus accumbens (NAc) -VTA pathway, whereas the hyperactivity relied on ventral hippocampus (vHPC). Hypodopaminergic VTA activity was recovered towards physiological levels by activation of mGluR2, but not mGluR3, and this remission was contingent on glutamatergic transmission within NAc and propagation via the ventral pallidum. Results of a light-dark transition task confirmed anxiolytic efficaciousness of mGluR2 activation. The hyperdopaminergic VTA activity, in contrast, was normalized by selective activation of mGluR3, but not mGluR2, within vHPC. AMPH re-exposure after abstinence turned VTA activity down, but this suppression involved alternative circuits and could no longer be rescued by mGluR activation. Conclusion Thus, abstinence from repeated AMPH intake drives VTA activity from hypo-into hyperdopaminergic states, and both can be readjusted towards physiological levels via different members of group II mGluRs.
Collapse
Affiliation(s)
- Ornella Valenti
- Division of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Katarzyna Anna Rekawek
- Division of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Sophie Wieser
- Division of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- Molecular Biotechnology, Fachhochschule (FH) Campus Wien, Vienna, Austria
| | - Hilal Bulut
- Division of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Petra Scholze
- Division of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Stefan Boehm
- Division of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
11
|
Yu YM, Xia SH, Xu Z, Zhao WN, Song L, Pan X, Zhong CC, Wang D, Gao YH, Yang JX, Wu P, Zhang H, An S, Cao JL, Ding HL. An accumbal microcircuit for the transition from acute to chronic pain. Curr Biol 2025; 35:1730-1749.e5. [PMID: 40112811 DOI: 10.1016/j.cub.2025.02.055] [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: 10/27/2024] [Revised: 01/28/2025] [Accepted: 02/25/2025] [Indexed: 03/22/2025]
Abstract
Persistent nociceptive inputs arising from peripheral tissues or/and nerve injuries cause maladaptive changes in neurons or neural circuits in the central nervous system, which further confer acute injury into chronic pain transitions (pain chronification) even after the injury is resolved. However, the critical brain regions and their neural mechanisms involved in this transition have not yet been elucidated. Here, we reveal an accumbal microcircuit that is essential for pain chronification. Notably, the increase of neuronal activity in the nucleus accumbens shell (NAcS) in the acute phase (<7 days) and in core (NAcC) in the chronic phase (14-21 days) was detected in a neuropathic pain mouse model. Importantly, we demonstrated that the NAcS neuronal activation in the acute phase of injury was necessary and sufficient for the development of chronic neuropathic pain. This process was mediated by the accumbal dopamine D2 receptor-expressing neuronal microcircuit from NAcS to NAcC. Thus, our findings reveal an accumbal microcircuit mechanism for pain chronification and suggest that the early intervention targeting this microcircuit may provide a therapeutic approach to pain chronification.
Collapse
Affiliation(s)
- Yu-Mei Yu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| | - Sun-Hui Xia
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Zheng Xu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Wei-Nan Zhao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Lingzhen Song
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Xiangyu Pan
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Chao-Chao Zhong
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Di Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Yi-Hong Gao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Jun-Xia Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Peng Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Hongxing Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Shuming An
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, Jiangsu, China.
| | - Hai-Lei Ding
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| |
Collapse
|
12
|
Sun Q, Liu M, Guan W, Xiao X, Dong C, Bruchas MR, Zweifel LS, Li Y, Tian L, Li B. Dynorphin modulates reward-seeking actions through a pallido-amygdala cholinergic circuit. Neuron 2025:S0896-6273(25)00218-1. [PMID: 40239651 DOI: 10.1016/j.neuron.2025.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/07/2025] [Accepted: 03/13/2025] [Indexed: 04/18/2025]
Abstract
The endogenous opioid peptide dynorphin and its receptor κ-opioid receptor (KOR) have been implicated in divergent behaviors, but the underlying mechanisms remain elusive. Here, we show that dynorphin released from nucleus accumbens dynorphinergic neurons exerts powerful modulation over a ventral pallidum (VP) disinhibitory circuit, thereby controlling cholinergic transmission to the amygdala and reward-seeking behavior in mice. On one hand, dynorphin acts postsynaptically via KORs on VP GABAergic neurons to promote disinhibition of cholinergic neurons, which release acetylcholine into the amygdala to facilitate learning and invigorate actions. On the other hand, dynorphin also acts presynaptically via KORs on dynorphinergic terminals to limit its own release. Such autoinhibition keeps cholinergic neurons from prolonged activation and release of acetylcholine and prevents perseverant reward seeking. Our study reveals how dynorphin exquisitely modulates behavior through the cholinergic system and provides an explanation for why these neuromodulators are involved in motivational disorders, including depression and addiction.
Collapse
Affiliation(s)
- Qingtao Sun
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| | - Mingzhe Liu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Wuqiang Guan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Xiong Xiao
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chunyang Dong
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA, USA; Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Michael R Bruchas
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Larry S Zweifel
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA
| | - Yulong Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA, USA; Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Bo Li
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| |
Collapse
|
13
|
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
|
14
|
Morris LS, Beltrán JM, Murrough JW, Morel C. Cross-species dissection of the modular role of the ventral tegmental area in depressive disorders. Neuroscience 2025; 569:248-266. [PMID: 39914519 PMCID: PMC11885014 DOI: 10.1016/j.neuroscience.2025.02.008] [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: 09/19/2024] [Revised: 01/17/2025] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
Depressive disorders, including major depressive disorder (MDD), represent one of the most prevalent set of disorders worldwide. MDD is characterized by a range of cognitive, behavioral, and neurobiological changes that contribute to the vast array of symptom profiles that make this disorder particularly difficult to treat. A multitude of established evidence suggests a role for the dopamine system, stemming in part from the ventral tegmental area (VTA), in mediating symptoms and behavioral changes that underlie depression. Developments in cutting-edge technologies in pre-clinical models of depressive phenotypes, such as retrograde tracing, electrophysiological recordings, immunohistochemistry, and molecular profiling, have allowed a deeper characterization of singular VTA neuron molecular, physiological, and projection properties. These developments have highlighted that the VTA is not a homogenous cell population but instead comprises vast cellular diversity that underscores its modular role across various functions related to reward processing, aversion, salience processing, learning and motivation. In this review, we begin by introducing the various cell types and brain regions that comprise the VTA circuitry. Then, we introduce the role of the VTA in reward processing as it compares to aversion processing. Next, we characterize distinct neural pathways within the VTA circuitry to understand the effects of chronic social and non-social stress and tie together how these neurobiological changes manifest into specific behavioral phenotypes. Finally, we relate these preclinical findings to clinical findings to parse the heterogeneity of depressive phenotypes and explain the efficacy of recent novel pharmacological interventions that may target the VTA in MDD.
Collapse
Affiliation(s)
- L S Morris
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai New York NY United States; Nuffield Department of Clinical Neurosciences, University of Oxford, UK; Department of Experimental Psychology, University of Oxford, UK.
| | - J M Beltrán
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai New York NY United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai New York NY United States
| | - J W Murrough
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai New York NY United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai New York NY United States; VISN 2 Mental Illness Research, Education, and Clinical Center (MIRECC), James J. Peters VA Medical Center Bronx NY United States
| | - C Morel
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai New York NY United States.
| |
Collapse
|
15
|
Cardozo Pinto DF, Pomrenze MB, Guo MY, Touponse GC, Chen APF, Bentzley BS, Eshel N, Malenka RC. Opponent control of reinforcement by striatal dopamine and serotonin. Nature 2025; 639:143-152. [PMID: 39586475 DOI: 10.1038/s41586-024-08412-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 11/14/2024] [Indexed: 11/27/2024]
Abstract
The neuromodulators dopamine (DA) and serotonin (5-hydroxytryptamine; 5HT) powerfully regulate associative learning1-8. Similarities in the activity and connectivity of these neuromodulatory systems have inspired competing models of how DA and 5HT interact to drive the formation of new associations9-14. However, these hypotheses have not been tested directly because it has not been possible to interrogate and manipulate multiple neuromodulatory systems in a single subject. Here we establish a mouse model that enables simultaneous genetic access to the brain's DA and 5HT neurons. Anterograde tracing revealed the nucleus accumbens (NAc) to be a putative hotspot for the integration of convergent DA and 5HT signals. Simultaneous recording of DA and 5HT axon activity, together with genetically encoded DA and 5HT sensor recordings, revealed that rewards increase DA signalling and decrease 5HT signalling in the NAc. Optogenetically dampening DA or 5HT reward responses individually produced modest behavioural deficits in an appetitive conditioning task, while blunting both signals together profoundly disrupted learning and reinforcement. Optogenetically reproducing DA and 5HT reward responses together was sufficient to drive the acquisition of new associations and supported reinforcement more potently than either manipulation did alone. Together, these results demonstrate that striatal DA and 5HT signals shape learning by exerting opponent control of reinforcement.
Collapse
Affiliation(s)
- Daniel F Cardozo Pinto
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Matthew B Pomrenze
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Michaela Y Guo
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Gavin C Touponse
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Allen P F Chen
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Neir Eshel
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Robert C Malenka
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
16
|
Medina S, Forero CG, Sanabria‐Mazo JP, Rodríguez‐Freire C, Navarrete J, O'Daly OG, Howard MA, Luciano JV. Baseline Functional Connectivity of the Mesolimbic, Salience, and Sensorimotor Systems Predicts Responses to Psychological Therapies for Chronic Low Back Pain With Comorbid Depression: A Functional MRI Study. Brain Behav 2025; 15:e70380. [PMID: 40022281 PMCID: PMC11870833 DOI: 10.1002/brb3.70380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/03/2025] Open
Abstract
INTRODUCTION Chronic low back pain (CLBP) is a prevalent and debilitating condition. Cognitive behavioral therapy (CBT) can improve coping mechanisms for CLBP and pain-related outcomes. However, the mechanisms by which they do so remain undetermined. We explored the neural correlates of CLBP symptoms and CBT action using functional magnetic resonance imaging (fMRI) in women with CLBP and comorbid depression. METHODS Forty individuals underwent fMRI followed by 8 weeks of either treatment as usual (TAU) or one of two CBT in addition to TAU: acceptance and commitment therapy (ACT) or behavioral activation treatment for depression (BATD). Pain intensity, depression, psychological inflexibility, and pain catastrophizing scores were obtained at baseline and follow-up. Functional connectivity (FC) patterns of the salience network (SN), sensorimotor network (SMN), and the mesolimbic pathway (MLP), derived from resting-state fMRI examination were correlated with both baseline and delta (baseline-follow-up) pain-related psychological measures. RESULTS Individuals receiving ACT and BATD showed reduced depression, psychological inflexibility, and pain catastrophizing. Strong baseline connectivity of the SN and SMN corresponded with higher pain intensity, but strong connectivity of the MLP and precuneus corresponded with lower pain intensity. Pain intensity changes correlated with mesolimbic-salience connectivity following ACT, and with sensorimotor connectivity following BATD. Specifically, stronger baseline FC between the MLP and posterior insula predicted greater pain intensity reduction with ACT, while stronger FC between the SMN and secondary somatosensory cortex predicted greater pain intensity reduction with BATD. FC of the SN correlated with changes in psychological inflexibility across both therapies. CONCLUSIONS We illustrate the potential of FC as a biomarker of CLBP plus depression and the response to CBT. Our data suggest ACT and BATD have differing underlying brain mechanisms. These findings indicate that FC biomarkers could guide personalized treatment, improving individual outcomes.
Collapse
Affiliation(s)
- Sonia Medina
- Department of NeuroimagingKing's College LondonLondonUK
- Exeter Medical SchoolUniversity of ExeterExeterUK
| | - Carlos G. Forero
- School of MedicineUniversitat Internacional de CatalunyaSant Cugat del VallèsSpain
| | - Juan P. Sanabria‐Mazo
- Teaching, Research & Innovation UnitParc Sanitari Sant Joan de DéuSant Boi de LlobregatSpain
- CIBER of Epidemiology and Public Health (CIBERESP)MadridSpain
| | - Carla Rodríguez‐Freire
- Teaching, Research & Innovation UnitParc Sanitari Sant Joan de DéuSant Boi de LlobregatSpain
| | - Jaime Navarrete
- Teaching, Research & Innovation UnitParc Sanitari Sant Joan de DéuSant Boi de LlobregatSpain
- CIBER of Epidemiology and Public Health (CIBERESP)MadridSpain
| | | | | | - Juan V. Luciano
- Teaching, Research & Innovation UnitParc Sanitari Sant Joan de DéuSant Boi de LlobregatSpain
- CIBER of Epidemiology and Public Health (CIBERESP)MadridSpain
- Department of Clinical & Health PsychologyAutonomous University of BarcelonaBellaterraSpain
| |
Collapse
|
17
|
Gu S, Park D, Seo S, Kim S, Kim Y, Webster M, Eom H, Lee D, Hong J, Han S, Cha H, Yun J. Crystallin Alpha B Inhibits Cocaine-Induced Conditioned Place Preference via the Modulation of Dopaminergic Neurotransmission. Addict Biol 2025; 30:e70028. [PMID: 40095747 PMCID: PMC11912016 DOI: 10.1111/adb.70028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 12/04/2024] [Accepted: 02/25/2025] [Indexed: 03/19/2025]
Abstract
Nonneuronal cells mediate neurotransmission and drug addiction. However, the role of oligodendrocytes in stress-induced cocaine relapses remains unclear. In the present study, we investigated the role of the oligodendrocyte-abundant molecule crystallin alpha B (CRYAB) in cocaine-induced conditioned place preference (CPP) relapsed by restraint stress. RNA sequencing (RNA-seq) was performed to identify oligodendrocytes and stress-associated molecules in the nucleus accumbens (NAcc) of both drug users and cocaine-treated animals. Further, we studied which cell subtypes in the brain express CRYAB. The effects of stress hormones and cocaine on CRYAB expression were evaluated in vitro in human oligodendrocytes. CRYAB is upregulated in the NAcc of both cocaine-treated animals and drug users. CRYAB levels in the NAcc of mice increased during CPP development but decreased following stress-induced relapse. Interestingly, CRYAB is expressed in oligodendrocytes in the NAcc of mice. Extracellular CRYAB levels are regulated by cocaine and stress hormone treatments in oligodendrocyte cultures. Dopamine levels in the NAcc and CPP development of CPP are significantly increased by cocaine in CRYAB knockout (KO) mice. Further, we demonstrated that CRYAB binds to the excitatory amino acid transporter 2 (EAAT2) in the NAcc of mice treated with cocaine. We suggest that oligodendrocyte-derived CRYAB regulates dopamine neurotransmission and stress-evoked cocaine reward behaviour via the modulation of EAAT2 in the NAcc.
Collapse
Affiliation(s)
- Sun Mi Gu
- College of PharmacyChungbuk National UniversityCheongjuChungcheongbukRepublic of Korea
| | - Daejin Park
- College of PharmacyChungbuk National UniversityCheongjuChungcheongbukRepublic of Korea
| | - Sowoon Seo
- College of PharmacyChungbuk National UniversityCheongjuChungcheongbukRepublic of Korea
| | - Sanghyeon Kim
- Stanley Brain Research LaboratoryStanley Medical Research InstituteRockvilleMarylandUSA
| | - Young Eun Kim
- College of PharmacyChungbuk National UniversityCheongjuChungcheongbukRepublic of Korea
| | - Maree J. Webster
- Stanley Brain Research LaboratoryStanley Medical Research InstituteRockvilleMarylandUSA
| | - Heejong Eom
- Laboratory Animal CenterOsong Medical Innovation FoundationCheongjuChungcheongbukRepublic of Korea
| | - Dohyun Lee
- Laboratory Animal CenterOsong Medical Innovation FoundationCheongjuChungcheongbukRepublic of Korea
| | - Jin Tae Hong
- College of PharmacyChungbuk National UniversityCheongjuChungcheongbukRepublic of Korea
| | - Sang‐Bae Han
- College of PharmacyChungbuk National UniversityCheongjuChungcheongbukRepublic of Korea
| | - Hye Jin Cha
- College of Veterinary MedicineGyeongsang National UniversityJinjuGyeongsangnamRepublic of Korea
| | - Jaesuk Yun
- College of PharmacyChungbuk National UniversityCheongjuChungcheongbukRepublic of Korea
| |
Collapse
|
18
|
Matsunaga D, Nakagawa H, Ishiwata T. Comparison of forced and voluntary exercise types on male rat brain monoamine levels, anxiety-like behaviour, and physiological indexes under light and dark phases. Behav Brain Res 2025; 479:115321. [PMID: 39510330 DOI: 10.1016/j.bbr.2024.115321] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 10/22/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024]
Abstract
PURPOSE Physical exercise improves physical and mental health; however, the differences between voluntary and forced exercise protocols are unclear. In addition, knowledge regarding the consequences of differences in testing timing, such as light and dark phases, in response to exercise type is limited. We investigated the effects of chronic forced and voluntary wheel running on the changes in brain monoamine levels (5-HT: serotonin, DA: dopamine, NA: noradrenaline), anxiety-like behaviours, and physiological stress responses in the light and dark phases. METHODS Adult male Wistar rats were equally and randomly assigned to four groups: sedentary control, voluntary exercise (free running on a wheel, V-EX), voluntary limited exercise (wheel available only 1 h/day, VL-EX), and forced exercise (running on a motorised wheel, F-EX). Each group was further divided into dark- or light-experimental condition groups. After 4 weeks, the rats underwent an open-field test. The monoamines and their metabolite levels were measured in the major neural cell bodies and the projection areas related to behaviour, cognition, anxiety, and stress in the brain. RESULTS Adrenal hypertrophy and elevated body temperature, except during the exercise period, were observed in the F-EX rats that exhibited anxiety-like behaviour. The levels of monoamines and their metabolites, particularly the 5-HTergic and DAergic systems, in specific areas, were significantly altered in the rats in the V-EX group compared to those in the VL-EX and other groups. These differences were observed only in the dark phase. CONCLUSION The results suggest that V-EX mainly stimulates the 5-HTergic and DAergic systems, while F-EX induces physiological stress and increases anxiety-like behaviour during the dark phase. This study highlights the importance of accounting for exercise types and light/dark phases in behavioural neuroscience experiments.
Collapse
Affiliation(s)
- Daisuke Matsunaga
- Department of Health-Promotion and Sports Science, Osaka Electro-Communication University, 1130-70 Kiyotaki, Shijonawate-shi, Osaka 575-0063, Japan; Graduate School of Community & Human Services, Rikkyo University, 1-2-26 Kitano, Niiza, Saitama 352-8558, Japan.
| | - Hikaru Nakagawa
- College of Sport &Wellness, Rikkyo University, 1-2-26 Kitano, Niiza, Saitama 352-8558, Japan
| | - Takayuki Ishiwata
- Graduate School of Community & Human Services, Rikkyo University, 1-2-26 Kitano, Niiza, Saitama 352-8558, Japan; College of Sport &Wellness, Rikkyo University, 1-2-26 Kitano, Niiza, Saitama 352-8558, Japan
| |
Collapse
|
19
|
Fraser KM, Collins V, Wolff AR, Ottenheimer DJ, Bornhoft KN, Pat F, Chen BJ, Janak PH, Saunders BT. Contextual cues facilitate dynamic value encoding in the mesolimbic dopamine system. Curr Biol 2025; 35:746-760.e5. [PMID: 39855205 PMCID: PMC11859769 DOI: 10.1016/j.cub.2024.12.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 11/04/2024] [Accepted: 12/11/2024] [Indexed: 01/27/2025]
Abstract
Adaptive behavior in a dynamic environmental context often requires rapid revaluation of stimuli that deviates from well-learned associations. The divergence between stable value-encoding and appropriate behavioral output remains a critical component of theories of dopamine's function in learning, motivation, and motor control. Yet, how dopamine neurons are involved in the revaluation of cues when the world changes, to alter our behavior, remains unclear. Here, we make use of a complementary set of in vivo approaches to clarify the contributions of the mesolimbic dopamine system to the dynamic reorganization of reward- seeking behavior. Male and female rats were trained to discriminate when a conditioned stimulus would be followed by a sucrose reward by exploiting the prior, non-overlapping presentation of a another discrete cue-an occasion setter. Only when the occasion setter's presentation preceded the conditioned stimulus did the conditioned stimulus predict sucrose delivery, dissociating the average value of the conditioned stimulus from its immediate value, on a trial-to-trial basis. Activity of ventral tegmental area dopamine neurons was essential for rats to successfully update behavioral response to the occasion setter. Moreover, dopamine release in the nucleus accumbens following the conditioned stimulus only occurred when the occasion setter indicated it would predict reward and did not reflect its average expected value. Downstream of dopamine release, we found that neurons in the nucleus accumbens dynamically tracked the value of the conditioned stimulus. Together, these results help refine notions of dopamine function, revealing a prominent contribution of the mesolimbic dopamine system to the rapid revaluation of motivation.
Collapse
Affiliation(s)
- Kurt M Fraser
- Department of Psychological & Brain Sciences, Johns Hopkins University, Baltimore, MD 21218, USA.
| | - Val Collins
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Amy R Wolff
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - David J Ottenheimer
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Kaisa N Bornhoft
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Fiona Pat
- Department of Psychological & Brain Sciences, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Bridget J Chen
- Department of Psychological & Brain Sciences, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Patricia H Janak
- Department of Psychological & Brain Sciences, Johns Hopkins University, Baltimore, MD 21218, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Benjamin T Saunders
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
20
|
Muroi Y, Ishii T. Neuronal stress-coping mechanisms in postpartum females. Neurosci Res 2025:S0168-0102(25)00032-X. [PMID: 39978735 DOI: 10.1016/j.neures.2025.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/05/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025]
Abstract
Animals exhibit a wide range of stress responses aimed at restoring homeostasis and promoting adaptation. In response to stress, they employ coping mechanisms to maintain physiological balance. Dysregulated stress-coping strategies have been associated with mental disorders, including depression, anxiety, and post-traumatic stress disorder. Understanding the neuronal mechanisms that regulate stress-coping is critical for elucidating normal physiological responses and addressing the pathological processes underlying these disorders. Stress responses are influenced by sex and life stage, with notable variability in the prevalence and severity of mental disorders based on these factors. Stress-coping mechanisms are pivotal in determining the vulnerability or resilience of an individual to stress. Thus, identifying differences in stress-coping strategies between sexes and across life stages is essential for advancing prevention and treatment strategies for stress-related mental disorders. This review explores the neuronal mechanisms underlying stress responses, emphasizing the distinct stress-coping strategies utilized by postpartum females. Highlighting these differences underscores the need for targeted prevention and treatment approaches that consider sex- and life stage-specific variations in stress-coping mechanisms.
Collapse
Affiliation(s)
- Yoshikage Muroi
- Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, National University Cooperation Hokkaido Higher Education and Research, Hokkaido 080-8555, Japan.
| | - Toshiaki Ishii
- Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, National University Cooperation Hokkaido Higher Education and Research, Hokkaido 080-8555, Japan
| |
Collapse
|
21
|
Requejo-Mendoza N, Arias-Montaño JA, Gutierrez R. Nucleus accumbens D2-expressing neurons: Balancing reward and licking disruption through rhythmic optogenetic stimulation. PLoS One 2025; 20:e0317605. [PMID: 39919051 PMCID: PMC11805367 DOI: 10.1371/journal.pone.0317605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/31/2024] [Indexed: 02/09/2025] Open
Abstract
Nucleus accumbens (NAc) dopamine D1 receptor-expressing neurons are known to be critical for processing reward and regulating food intake. However, the role of D2-expressing neurons in this nucleus remains less understood. This study employed optogenetic manipulations to investigate the role of NAc D2-expressing neurons in reward processing and sucrose consumption. Optogenetic activation of these neurons decreased sucrose preference (at 20 Hz), disrupted licking patterns (particularly at 8 and 20 Hz), and increased self-stimulation. Conversely, synchronizing stimulation with the animal licking rhythm mitigated licking disruption and even increased sucrose intake, suggesting a rewarding effect. Furthermore, 20 Hz stimulation (but not 8 Hz) induced place preference in a real-time place preference (RTPP) test. In contrast, inhibiting D2 neurons produced a negative hedonic state, although not reaching complete aversion, influencing food choices in specific contexts. For instance, while the RTPP test per se was not sensitive enough to observe place aversion when mice could choose between consuming a high-fat diet (HFD) pellet in a context associated with or without inhibition of D2 neurons, they preferred to consume HFD on the non-inhibited side. This suggests that the palatability of HFD can unmask (but also overshadow) the negative hedonic state associated with D2 neuron inhibition. A negative reinforcement paradigm further confirmed the active avoidance behavior induced by D2 neuron inhibition. In conclusion, NAc D2 neuron inhibition induces a negative hedonic state, while activation has a dual effect-it is rewarding yet disrupts licking behavior-highlighting its complex role in reward and consummatory behavior. Importantly, self-paced stimulation, where the animal controls the timing of the stimulation through its licking behavior, offers a more efficient and natural approach for stimulating NAc activity.
Collapse
Affiliation(s)
- Nikte Requejo-Mendoza
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados (Cinvestav), Ciudad de México, México
- Laboratory Neurobiology of Appetite; Departamento Farmacología, Centro de Investigación y de Estudios Avanzados (Cinvestav), Ciudad de México, México
| | - José-Antonio Arias-Montaño
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados (Cinvestav), Ciudad de México, México
| | - Ranier Gutierrez
- Laboratory Neurobiology of Appetite; Departamento Farmacología, Centro de Investigación y de Estudios Avanzados (Cinvestav), Ciudad de México, México
- Laboratory Neurobiology of Appetite; Centro de Investigación sobre el Envejecimiento, Centro de Investigación y de Estudios Avanzados (CIE, Cinvestav Sede sur), Ciudad de México, México
| |
Collapse
|
22
|
Akyuz E, Arulsamy A, Aslan FS, Sarisözen B, Guney B, Hekimoglu A, Yilmaz BN, Retinasamy T, Shaikh MF. An Expanded Narrative Review of Neurotransmitters on Alzheimer's Disease: The Role of Therapeutic Interventions on Neurotransmission. Mol Neurobiol 2025; 62:1631-1674. [PMID: 39012443 PMCID: PMC11772559 DOI: 10.1007/s12035-024-04333-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 06/24/2024] [Indexed: 07/17/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease. The accumulation of amyloid-β (Aβ) plaques and tau neurofibrillary tangles are the key players responsible for the pathogenesis of the disease. The accumulation of Aβ plaques and tau affect the balance in chemical neurotransmitters in the brain. Thus, the current review examined the role of neurotransmitters in the pathogenesis of Alzheimer's disease and discusses the alterations in the neurochemical activity and cross talk with their receptors and transporters. In the presence of Aβ plaques and neurofibrillary tangles, changes may occur in the expression of neuronal receptors which in turn triggers excessive release of glutamate into the synaptic cleft contributing to cell death and neuronal damage. The GABAergic system may also be affected by AD pathology in a similar way. In addition, decreased receptors in the cholinergic system and dysfunction in the dopamine neurotransmission of AD pathology may also contribute to the damage to cognitive function. Moreover, the presence of deficiencies in noradrenergic neurons within the locus coeruleus in AD suggests that noradrenergic stimulation could be useful in addressing its pathophysiology. The regulation of melatonin, known for its effectiveness in enhancing cognitive function and preventing Aβ accumulation, along with the involvement of the serotonergic system and histaminergic system in cognition and memory, becomes remarkable for promoting neurotransmission in AD. Additionally, nitric oxide and adenosine-based therapeutic approaches play a protective role in AD by preventing neuroinflammation. Overall, neurotransmitter-based therapeutic strategies emerge as pivotal for addressing neurotransmitter homeostasis and neurotransmission in the context of AD. This review discussed the potential for neurotransmitter-based drugs to be effective in slowing and correcting the neurodegenerative processes in AD by targeting the neurochemical imbalance in the brain. Therefore, neurotransmitter-based drugs could serve as a future therapeutic strategy to tackle AD.
Collapse
Affiliation(s)
- Enes Akyuz
- Department of Biophysics, International School of Medicine, University of Health Sciences, Istanbul, Turkey
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Alina Arulsamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia.
| | | | - Bugra Sarisözen
- School of Medicine, Tekirdağ Namık Kemal University, Tekirdağ, Turkey
| | - Beyzanur Guney
- International School of Medicine, University of Health Sciences, Istanbul, Turkey
| | | | - Beyza Nur Yilmaz
- International School of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Thaarvena Retinasamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia.
- School of Dentistry and Medical Sciences, Charles Sturt University, Orange, New South Wales, 2800, Australia.
| |
Collapse
|
23
|
Li WQ, Liu SN, Yang SC, Lin X, Zhang ZJ. Nitrous oxide exerts rewarding effect via regulating D1 receptor and BDNF pathway in ventral tegmental area-nucleus accumbens dopamine circuit. Transl Psychiatry 2025; 15:34. [PMID: 39875366 PMCID: PMC11775255 DOI: 10.1038/s41398-025-03257-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 12/09/2024] [Accepted: 01/23/2025] [Indexed: 01/30/2025] Open
Abstract
Recreational use of nitrous oxide (N2O) has risen dramatically over the past decades. This study aimed to examine its rewarding effect and the underlying mechanisms. The exposure of mice to a subanesthetic concentration (20%) of N2O for 30 min for 4 consecutive days paired with N2O in the morning and paired with the air in the afternoon produced apparent rewarding behavior in the conditioned place preference (CPP) paradigm. This was abrogated by microinjection into the nucleus accumbens (NAc) of the dopamine (DA) D1 receptor antagonist SCH23390, but not the D2 antagonist haloperidol. N2O robustly enhanced DAergic neuronal activity of the ventral tegmental area (VTA) and the concentration of DA in the NAc. The repeated N2O exposure also upregulated the expression of brain-derived neurotrophic factor (BDNF) in the VTA and its multiple downstream mediators in the NAc. Conversely, VTA focal knockdown of BDNF and the inhibition of the downstream mediators suppressed the N2O-induced rewarding effect and the DAergic neuronal activity of the VTA. Further, the combined intervention of BDNF knockdown and D1 antagonist significantly inhibited the N2O-induced rewarding effect in mice, which was greater than that of BDNF knockdown alone, but was not significantly different from that of D1 antagonist alone. These results indicate that the rewarding properties of N2O at subanesthetic concentration are associated with its upregulation of the VTA-NAc DA reward pathway probably via mediation of D1 receptor and BDNF/TrkB signaling. Among them, the modulation of BDNF may be the upstream of D1 receptor involved in N2O rewarding effect.
Collapse
Affiliation(s)
- Wen-Qi Li
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Sheng-Nan Liu
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Si-Chang Yang
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xiang Lin
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Zhang-Jin Zhang
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
24
|
Fritz M, Rosa PB, Wilhelms D, Jaarola M, Ruud J, Engblom D, Klawonn AM. Nicotinic α7 receptors on cholinergic neurons in the striatum mediate cocaine-reinforcement, but not food reward. Front Mol Neurosci 2025; 17:1418686. [PMID: 39906479 PMCID: PMC11790553 DOI: 10.3389/fnmol.2024.1418686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 12/30/2024] [Indexed: 02/06/2025] Open
Abstract
The neurotransmitter acetylcholine has since long been implicated in reward learning and drug addiction. However, the role of specific cholinergic receptor subtypes on different neuronal populations remain elusive. Here, we studied the function of nicotinic acetylcholinergic alpha 7 receptors (α7 nAChRs) in cocaine and food-enforced behaviors. We found that global deletion of α7 nAChRs in mice attenuates cocaine seeking in a Pavlovian conditioned place preference paradigm and decreases operant responding to cocaine in a runway task and in self-administration, without influencing responding to palatable food. This effect can be attributed to alpha 7 receptor signaling in the striatum, as selective deletion of striatal α7 nAChRs using a viral vector approach resulted in a similar decrease in cocaine-preference as that of global deletion. To investigate which type of striatal neurons are responsible for this effect, we selectively targeted Cholinergic (ChAT-expressing) neurons and dopamine D1-receptor (D1R) expressing neurons. Mice with conditional deletion of α7 nAChRs in ChAT-neurons (α7 nAChR-ChATCre) exhibited decreased cocaine place preference and intact place preference for food, while α7 nAChR-D1RCre mice had no changes in reward learning to neither food nor cocaine. Cocaine induction of striatal immediate early gene expression of cFos, FosB, Arc and EGR2 was blocked in α7 nAChR-ChATCre mice, demonstrating the importance of α7 nAChRs on cholinergic neurons for striatal neuronal activity changes. Collectively, our findings show that α7 nAChRs on cholinergic interneurons in the striatum are pivotal for learning processes related to cocaine, but not food reward.
Collapse
Affiliation(s)
- Michael Fritz
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- School of Health and Social Sciences, AKAD University of Applied Sciences, Stuttgart, Germany
- Department for Forensic Psychiatry and Psychotherapy, Ulm University, Ulm, Germany
| | - Priscila Batista Rosa
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Daniel Wilhelms
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Emergency Medicine, Linköping University Hospital, Linköping, Sweden
| | - Maarit Jaarola
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Johan Ruud
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - David Engblom
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Anna M. Klawonn
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Danish Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
| |
Collapse
|
25
|
Wang J, Zhang M, Sun Y, Su X, Hui R, Zhang L, Xie B, Cong B, Luo Y, Wen D, Ma C. The modulation of cholecystokinin receptor 1 in the NAc core input from VTA on METH-induced CPP acquisition. Life Sci 2025; 361:123290. [PMID: 39638282 DOI: 10.1016/j.lfs.2024.123290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/14/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Methamphetamine (METH) is a potent psychostimulant that interferes the functionality of various brain regions and nervous connections, leading to addiction. The nucleus accumbens core (NAcC), primarily composed of gamma-aminobutyric acid (GABAergic) neurons, serves as a critical nucleus intimately related to addictive behavior. Previous research has indicated the involvement of cholecystokinin (CCK) receptors in drug addiction, yet the precise function of CCK receptors within the neural circuitry mediating METH-induced addiction remains elusive. METHODS METH-induced conditioned place preference (CPP) model was established in mice. In CCK receptor 1 conditional knockout (CCK1Rflox/flox) or CCK receptor 2 conditional knockout (CCK2Rflox/flox) mice, we then utilized the adeno-associated virus (AAV) transfection system to knock out the specific CCK receptor subtype and explore the function of the CCK receptors in the ventral tegmental area (VTA) to NAcC circuit during METH-induced CPP acquisition. RESULTS During the acquisition of METH-induced CPP, the expression of CCK1R, but not CCK2R, was upregulated specifically in NAcC. Genetic disruption of either CCK1R in the NAcC effectively hindered METH-induced CPP acquisition and prevented the hyper-excitability of neurons triggered by METH. Furthermore, CCK is released by dopaminergic neurons in the VTA, projecting to the NAcC. Notably, specifically knocking out CCK1R in the VTADA → NAcCGABA circuit blocked the presynaptic release and synaptic plasticity enhancement induced by METH. CONCLUSIONS These discoveries highlight the critical effect of CCK1R in the VTADA → NAcCGABA circuit on METH-induced CPP acquisition and provide a more comprehensive understanding of the mechanisms underlying CCK receptors contributing to the METH-induced addictive behavior.
Collapse
Affiliation(s)
- Jian Wang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province, China
| | - Minglong Zhang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province, China; Department of Genetics, Qiqihar Medical University, Qiqihar, Heilongjiang Province, China
| | - Yufei Sun
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province, China
| | - Xiaorui Su
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province, China
| | - Rongji Hui
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province, China
| | - Ludi Zhang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province, China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, Hebei Province, China
| | - Bing Xie
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province, China
| | - Bin Cong
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province, China; Hainan Tropical Forensic Medicine Academician Workstation, Haikou, Hainan Province, China
| | - Yixiao Luo
- NHC Key Laboratory of Birth Defect for Research and Prevention (Hunan Provincial Maternal and Child Health Care Hospital), Hunan Province People's Hospital, Changsha, Hunan Province, China; The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Di Wen
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province, China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, Hebei Province, China; Hainan Tropical Forensic Medicine Academician Workstation, Haikou, Hainan Province, China.
| | - Chunling Ma
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province, China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, Hebei Province, China; Hainan Tropical Forensic Medicine Academician Workstation, Haikou, Hainan Province, China.
| |
Collapse
|
26
|
Magnard R, Cheng Y, Zhou J, Province H, Thiriet N, Janak PH, Vandaele Y. Sequence termination cues drive habits via dopamine-mediated credit assignment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.16.618735. [PMID: 39463939 PMCID: PMC11507917 DOI: 10.1101/2024.10.16.618735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Mesolimbic dopamine (DA) neurons are central to sequence learning and habit formation. Yet, the mechanisms by which cue-induced DA neural activity drives goal-directed or habitual sequence execution remain unknown. We designed two novel tasks to investigate how sequence initiation and termination cues influence DA-driven behavioral strategies and learning. We found that sequence initiation and termination cues differentially affect reward expectation during action sequences, with only the termination cue contributing to greater outcome devaluation insensitivity, automaticity and behavioral chunking. Mesolimbic fiber photometry recording revealed that this habit-like behavior was associated with a rapid backpropagation in DA signals from the reward to the immediately preceding cue and with attenuated DA reward prediction error signals, which reflected greater behavioral inflexibility. Finally, in absence of external cues, brief optogenetic stimulation of VTA DA neurons at sequence termination was sufficient to drive automaticity and behavioral chunking. Our results highlight the critical role of cue-evoked DA signals at sequence termination in mediating credit assignment and driving the development of habitual action sequence execution.
Collapse
Affiliation(s)
- Robin Magnard
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD
| | - Yifeng Cheng
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD
| | - Joanna Zhou
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD
| | - Haley Province
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD
| | - Nathalie Thiriet
- Université de Poitiers, INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Patricia H. Janak
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD
| | - Youna Vandaele
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD
- Université de Poitiers, INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Poitiers, France
| |
Collapse
|
27
|
Li J, Wei Y, Xiang J, Zhang D. Role of the ventral tegmental area in general anesthesia. Eur J Pharmacol 2025; 986:177145. [PMID: 39566814 DOI: 10.1016/j.ejphar.2024.177145] [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: 08/31/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 11/22/2024]
Abstract
The ventral tegmental area (VTA), located in the midbrain, plays a pivotal role in the regulation of many important behaviors, such as reward, addiction, aversion, memory, learning, and sleep-wakefulness cycles. The majority of VTA neurons are dopaminergic neurons, although there is a significant proportion of GABAergic neurons and few glutamatergic neurons. These neuronal types project to different brain regions, thus mediating various biological functions. Therefore, the diverse roles of the VTA might depend on its heterogeneous neuronal types and projecting circuits. General anesthesia and sleep-wakefulness cycles share the feature of reversible loss of consciousness, and several common neural mechanisms underlie these two conditions. In addition to the well-known regulatory role of VTA in sleep-wakefulness, emerging evidence has demonstrated that VTA activity is also associated with promoting emergence from general anesthesia. Herein, we reviewed the literature and summarized the evidence regarding the modulation of the VTA by general anesthesia in rodents, which will improve the understanding of the modulatory mechanism of the VTA in general anesthesia.
Collapse
Affiliation(s)
- Jia Li
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710000, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430048, China.
| | - Yiyong Wei
- Department of Anesthesiology, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, 518100, China
| | - Jiaxin Xiang
- Department of Anesthesiology, Weill Cornell Medicine, New York, 10065, USA
| | - Donghang Zhang
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430048, China; Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
28
|
Dvorak NM, Wadsworth PA, Aquino-Miranda G, Wang P, Engelke DS, Zhou J, Nguyen N, Singh AK, Aceto G, Haghighijoo Z, Smith II, Goode N, Zhou M, Avchalumov Y, Troendle EP, Tapia CM, Chen H, Powell RT, Baumgartner TJ, Singh J, Koff L, Di Re J, Wadsworth AE, Marosi M, Azar MR, Elias K, Lehmann P, Mármol Contreras YM, Shah P, Gutierrez H, Green TA, Ulmschneider MB, D'Ascenzo M, Stephan C, Cui G, Do Monte FH, Zhou J, Laezza F. Enhanced motivated behavior mediated by pharmacological targeting of the FGF14/Na v1.6 complex in nucleus accumbens neurons. Nat Commun 2025; 16:110. [PMID: 39747162 PMCID: PMC11696184 DOI: 10.1038/s41467-024-55554-7] [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: 03/30/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
Protein/protein interactions (PPI) play crucial roles in neuronal functions. Yet, their potential as drug targets for brain disorders remains underexplored. The fibroblast growth factor 14 (FGF14)/voltage-gated Na+ channel 1.6 (Nav1.6) complex regulates excitability of medium spiny neurons (MSN) of the nucleus accumbens (NAc), a central hub of reward circuitry that controls motivated behaviors. Here, we identified compound 1028 (IUPAC: ethyl 3-(2-(3-(hydroxymethyl)-1H-indol-1-yl)acetamido)benzoate), a brain-permeable small molecule that targets FGF14R117, a critical residue located within a druggable pocket at the FGF14/Nav1.6 PPI interface. We found that 1028 modulates FGF14/Nav1.6 complex assembly and depolarizes the voltage-dependence of Nav1.6 channel inactivation with nanomolar potency by modulating the intramolecular interaction between the III-IV linker and C-terminal domain of the Nav1.6 channel. Consistent with the compound's effects on Nav1.6 channel inactivation, 1028 enhances MSN excitability ex vivo and accumbal neuron firing rate in vivo in murine models. Systemic administration of 1028 maintains behavioral motivation preferentially during motivationally deficient conditions in murine models. These behavioral effects were abrogated by in vivo gene silencing of Fgf14 in the NAc and were accompanied by a selective reduction in accumbal dopamine levels during reward consumption in murine models. These findings underscore the potential to selectively regulate complex behaviors associated with neuropsychiatric disorders through targeting of PPIs in neurons.
Collapse
Affiliation(s)
- Nolan M Dvorak
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Paul A Wadsworth
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
- Department of Pathology, Stanford Medicine, Stanford, CA, USA
| | - Guillermo Aquino-Miranda
- Department of Neurobiology and Anatomy, University of Texas Health Science Center, Houston, TX, USA
| | - Pingyuan Wang
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Douglas S Engelke
- Department of Neurobiology and Anatomy, University of Texas Health Science Center, Houston, TX, USA
| | - Jingheng Zhou
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, USA
| | - Nghi Nguyen
- High-Throughput Research and Screening Center, Texas A&M Health Science Center, Houston, TX, USA
| | - Aditya K Singh
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Giuseppe Aceto
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Zahra Haghighijoo
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Isabella I Smith
- Department of Neurobiology and Anatomy, University of Texas Health Science Center, Houston, TX, USA
| | - Nana Goode
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mingxiang Zhou
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Yosef Avchalumov
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Evan P Troendle
- Department of Chemistry, King's College London 7 Trinity Street, London, UK
| | - Cynthia M Tapia
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Haiying Chen
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Reid T Powell
- High-Throughput Research and Screening Center, Texas A&M Health Science Center, Houston, TX, USA
| | - Timothy J Baumgartner
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jully Singh
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Leandra Koff
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jessica Di Re
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Ann E Wadsworth
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mate Marosi
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Marc R Azar
- Behavioral Pharma Inc., 505 Coast Blvd. South, Suite 212, La Jolla, CA, USA
| | - Kristina Elias
- Behavioral Pharma Inc., 505 Coast Blvd. South, Suite 212, La Jolla, CA, USA
| | - Paul Lehmann
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | | | - Poonam Shah
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Hector Gutierrez
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Thomas A Green
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | | | - Marcello D'Ascenzo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Clifford Stephan
- High-Throughput Research and Screening Center, Texas A&M Health Science Center, Houston, TX, USA
| | - Guohong Cui
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, USA
| | - Fabricio H Do Monte
- Department of Neurobiology and Anatomy, University of Texas Health Science Center, Houston, TX, USA
| | - Jia Zhou
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
29
|
Johnson NL, Cotelo-Larrea A, Stetzik LA, Akkaya UM, Zhang Z, Gadziola MA, Varga AG, Ma M, Wesson DW. Dopaminergic signaling to ventral striatum neurons initiates sniffing behavior. Nat Commun 2025; 16:336. [PMID: 39747223 PMCID: PMC11696867 DOI: 10.1038/s41467-024-55644-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
Sniffing is a motivated behavior displayed by nearly all terrestrial vertebrates. While sniffing is associated with acquiring and processing odors, sniffing is also intertwined with affective and motivated states. The systems which influence the display of sniffing are unclear. Here, we report that dopamine release into the ventral striatum in mice is coupled with bouts of sniffing and that stimulation of dopaminergic terminals in these regions drives increases in respiratory rate to initiate sniffing whereas inhibition of these terminals reduces respiratory rate. Both the firing of individual neurons and the activity of post-synaptic D1 and D2 dopamine receptor-expressing neurons are coupled with sniffing and local antagonism of D1 and D2 receptors squelches sniffing. Together, these results support a model whereby sniffing can be initiated by dopamine's actions upon ventral striatum neurons. The nature of sniffing being integral to both olfaction and motivated behaviors implicates this circuit in a wide array of functions.
Collapse
Affiliation(s)
- Natalie L Johnson
- Department of Pharmacology and Therapeutics, Florida Chemical Senses Institute, Center for Addiction Research and Education; University of Florida College of Medicine, Gainesville, FL, USA
| | - Anamaria Cotelo-Larrea
- Department of Pharmacology and Therapeutics, Florida Chemical Senses Institute, Center for Addiction Research and Education; University of Florida College of Medicine, Gainesville, FL, USA
| | - Lucas A Stetzik
- Department of Pharmacology and Therapeutics, Florida Chemical Senses Institute, Center for Addiction Research and Education; University of Florida College of Medicine, Gainesville, FL, USA
| | - Umit M Akkaya
- Department of Computer Engineering, Gebze Technical University, Kocaeli, Turkey
| | - Zihao Zhang
- Department of Pharmacology and Therapeutics, Florida Chemical Senses Institute, Center for Addiction Research and Education; University of Florida College of Medicine, Gainesville, FL, USA
| | - Marie A Gadziola
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, Canada
| | - Adrienn G Varga
- Department of Neuroscience, Breathing Research and Therapeutics Center, McKnight Brain Institute; University of Florida College of Medicine, Gainesville, FL, USA
| | - Minghong Ma
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel W Wesson
- Department of Pharmacology and Therapeutics, Florida Chemical Senses Institute, Center for Addiction Research and Education; University of Florida College of Medicine, Gainesville, FL, USA.
| |
Collapse
|
30
|
Miguel Telega L, Ashouri Vajari D, Ramanathan C, Coenen VA, Döbrössy MD. Chronic in vivo sequelae of repetitive acute mfb-DBS on accumbal dopamine and midbrain neuronal activity. J Neurochem 2025; 169:e16223. [PMID: 39308085 DOI: 10.1111/jnc.16223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/08/2024] [Accepted: 08/26/2024] [Indexed: 12/20/2024]
Abstract
Medial Forebrain Bundle Deep Brain Stimulation (MFB-DBS) can have rapid and long lasting antidepressant effects in Treatment Resistant Depression (TRD) patients. The mechanisms are not well understood, but one hypothesis stipulates that modulation of the dopaminergic (DAergic) fibers contribute to the therapeutic outcome. Acute DBS effects on DA release have been studied; however, longitudinal studies with acute-repetitive DBS are lacking. Long-Evans accumbal DA release and Ventral Tegmental Area (VTA) calcium tonic and phasic signaling to different mfb-DBS parameters were measured using fiber photometry over 8 weeks, following acute and repetitive stimulation in behaving and non-behaving animals. DBS-induced release was observed in both targets, with increased frequency and DBS duration. 130 Hz stimulation increased phasic and tonic DA response over time, with the latter being a potential mechanism for its long-term clinical effectiveness. VTA calcium transients decreased, while phasic activity increased with frequency. Pulse width (PW)-mediated differential peak release timing also suggests potential parallel activation of diverse fiber types. Additionally, decreased DA transients rate during Elevated Plus Maze (EPM) suggests context and stimulation duration-dependent DA release. The data confirm chronic antidromic/orthodromic DAergic responses with stimulation parameter dependent variability, providing novel insights into temporal adaptations, connectivity and fiber recruitment on mfb DBS.
Collapse
Affiliation(s)
- Lidia Miguel Telega
- Laboratory of Stereotaxy and Interventional Neurosciences (SIN), Department of Stereotactic and Functional Neurosurgery, Medical Center, -University of Freiburg, Freiburg im Breisgau, Germany
- Department of Stereotactic and Functional Neurosurgery, Medical Center, -University of Freiburg, Freiburg im Breisgau, Germany
- Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
- BrainLinks-BrainTools, IMBIT (Institute for Machine-Brain Interfacing Technology), University of Freiburg, Freiburg im Breisgau, Germany
| | - Danesh Ashouri Vajari
- BrainLinks-BrainTools, IMBIT (Institute for Machine-Brain Interfacing Technology), University of Freiburg, Freiburg im Breisgau, Germany
- Laboratory for Biomedical Microtechnology, Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg im Breisgau, Germany
| | - Chockalingam Ramanathan
- Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
- Institute for Physiology I, Medical Faculty, Albert-Ludwigs-University Freiburg, Freiburg im Breisgau, Germany
- Bernstein Center Freiburg, University of Freiburg, Freiburg im Breisgau, Germany
| | - Volker A Coenen
- Laboratory of Stereotaxy and Interventional Neurosciences (SIN), Department of Stereotactic and Functional Neurosurgery, Medical Center, -University of Freiburg, Freiburg im Breisgau, Germany
- Department of Stereotactic and Functional Neurosurgery, Medical Center, -University of Freiburg, Freiburg im Breisgau, Germany
- BrainLinks-BrainTools, IMBIT (Institute for Machine-Brain Interfacing Technology), University of Freiburg, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
- Center for Basics in Neuromodulation, University of Freiburg, Freiburg im Breisgau, Germany
| | - Máté D Döbrössy
- Laboratory of Stereotaxy and Interventional Neurosciences (SIN), Department of Stereotactic and Functional Neurosurgery, Medical Center, -University of Freiburg, Freiburg im Breisgau, Germany
- Department of Stereotactic and Functional Neurosurgery, Medical Center, -University of Freiburg, Freiburg im Breisgau, Germany
- Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
- Center for Basics in Neuromodulation, University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
31
|
Niitani K, Nishida R, Futami Y, Nishitani N, Deyama S, Kaneda K. Activation of ventral pallidum-projecting neurons in the nucleus accumbens via 5-HT 2C receptor stimulation regulates motivation for wheel running in male mice. Neuropharmacology 2024; 261:110181. [PMID: 39393590 DOI: 10.1016/j.neuropharm.2024.110181] [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: 04/11/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
Rodents have a strong motivation for wheel running; however, the neural mechanisms that regulate their motivation remain unknown. We investigated the possible involvement of serotonin (5-HT) systems in regulating motivation for wheel running in male mice. Systemic administration of a 5-HT1A receptor antagonist (WAY100635) increased the number of wheel rotations, whereas administration of a 5-HT2A or 5-HT2C receptor antagonist (volinanserin or SB242084, respectively) decreased it. In the open field test, neither WAY100635 nor volinanserin affected locomotor activity, whereas SB242084 increased locomotor activity. To identify the brain regions on which these antagonists act, we locally injected these into the motivational circuitry, including the nucleus accumbens (NAc), dorsomedial striatum (DM-Str), and medial prefrontal cortex (mPFC). Injection of SB242084 into the NAc, but not the DM-Str or mPFC, reduced the number of wheel rotations without altering locomotor activity. The local administration of WAY100635 or volinanserin to these brain regions did not affect the number of wheel rotations. Immunohistochemical analyses revealed that wheel running increased the number of c-Fos-positive cells in the NAc medial shell (NAc-MS), which was reduced by systemic SB242084 administration. In vitro slice whole-cell recordings showed that bath application of the 5-HT2C receptor agonist lorcaserin increased the frequency of spontaneous excitatory and inhibitory postsynaptic currents in the ventral tegmental area (VTA)-projecting neurons, whereas it only increased the frequency of spontaneous excitatory postsynaptic currents in ventral pallidum (VP)-projecting neurons in the NAc-MS. These findings suggest that the activation of VP-projecting NAc-MS neurons via 5-HT2C receptor stimulation regulates motivation for wheel running.
Collapse
Affiliation(s)
- Kazuhei Niitani
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Ryoma Nishida
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Yusaku Futami
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Naoya Nishitani
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Satoshi Deyama
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Katsuyuki Kaneda
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan.
| |
Collapse
|
32
|
Yu Z, Fu Q, Qiu T, Yang C, Lu M, Peng Q, Yang J, Hu Z. Role of Rab10 in cocaine-induced behavioral effects is associated with GABAB receptor membrane expression in the nucleus accumbens. Front Pharmacol 2024; 15:1496657. [PMID: 39669198 PMCID: PMC11635607 DOI: 10.3389/fphar.2024.1496657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 11/11/2024] [Indexed: 12/14/2024] Open
Abstract
Aim Previous studies have demonstrated that Ras-related GTP-binding protein Rab10 (Rab10) plays a role in psychostimulant-induced behavioral effects. In this study, we showed that Rab10 in the nucleus accumbens (NAc) of male animals affects the development of cocaine-induced behavioral effects, which are associated with the plasma membrane expression of the GABAB heteroreceptor (GABABR). Methods We performed flow cytometry, immunoendocytosis, pHluorin activity analysis, electrophysiology analysis, and open-field testing to explore the role of Rab10 in modulating the membrane expression and function of GABABR and its regulatory effect on cocaine-induced behavioral effects. Results Transcriptomics analysis showed that Rab10 was elevated following acute cocaine treatment. Membrane levels of Rab10 increased within day 1 of the cocaine treatment, subsequently decreasing at later time points. Rab10 deficiency in NAc regions significantly increased cocaine-inhibited membrane GABABR levels and inhibited cocaine-induced hyperlocomotion and behavioral sensitization. In addition, GAD 67 + -expressing neurons from NAc regions treated with cocaine revealed a significant decrease in Rab10 membrane expression. Furthermore, NAc neuron-specific Rab10 knockout resulted in a significant increase in the cocaine-inhibited membrane expression of GABABR, along with increased miniature inhibitory postsynaptic current (mIPSC) amplitude and attenuation of baclofen-amplified Ca2+ influx. Conclusion These results uncover a new mechanism in which Rab10-GABABR signaling may serve as a potential pathway for regulating cocaine-induced behavioral effects.
Collapse
Affiliation(s)
- Zhuoxuan Yu
- The First Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Qiang Fu
- Department of Respiration, Department Two, Jiangxi Provincial People’s Hospital, Nanchang, Jiangxi, China
| | - Tianyun Qiu
- Department of Clinical Laboratory, Wuhan Hankou Hospital, Wuhan, Hubei, China
| | - Caidi Yang
- The First Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Mingfen Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Qinghua Peng
- Department of Anesthesiology, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jianhua Yang
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhenzhen Hu
- Department of Pathophysiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
33
|
Liu Q, Xiong J, Kim DW, Lee SS, Bell BJ, Alexandre C, Blackshaw S, Latremoliere A, Wu MN. An amygdalar oscillator coordinates cellular and behavioral rhythms. Neuron 2024; 112:3750-3767.e7. [PMID: 39303704 PMCID: PMC11581920 DOI: 10.1016/j.neuron.2024.08.013] [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: 11/08/2023] [Revised: 07/12/2024] [Accepted: 08/23/2024] [Indexed: 09/22/2024]
Abstract
Circadian rhythms are generated by the master pacemaker suprachiasmatic nucleus (SCN) in concert with local clocks throughout the body. Although many brain regions exhibit cycling clock gene expression, the identity of a discrete extra-SCN brain oscillator that produces rhythmic behavior has remained elusive. Here, we show that an extra-SCN oscillator in the lateral amygdala (LA) is defined by expression of the clock-output molecule mWAKE/ANKFN1. mWAKE is enriched in the anterior/dorsal LA (adLA), and, strikingly, selective disruption of clock function or excitatory signaling in adLAmWAKE neurons abolishes Period2 (PER2) rhythms throughout the LA. mWAKE levels rise at night and promote rhythmic excitability of adLAmWAKE neurons by upregulating Ca2+-activated K+ channel activity specifically at night. adLAmWAKE neurons coordinate rhythmic sensory perception and anxiety in a clock-dependent and WAKE-dependent manner. Together, these data reveal the cellular identity of an extra-SCN brain oscillator and suggest a multi-level hierarchical system organizing molecular and behavioral rhythms.
Collapse
Affiliation(s)
- Qiang Liu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jiali Xiong
- Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Dong Won Kim
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sang Soo Lee
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Benjamin J Bell
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chloe Alexandre
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Seth Blackshaw
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Alban Latremoliere
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Mark N Wu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
34
|
Woo J, Uprety A, Reid D, Chang I, Samuel AK, Schuch HDC, Swain CC, Ostroumov A. Dynamic Changes in Chloride Homeostasis Coordinate Midbrain Inhibitory Network Activity during Reward Learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.18.624156. [PMID: 39605586 PMCID: PMC11601619 DOI: 10.1101/2024.11.18.624156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The ability to associate environmental stimuli with positive outcomes is a fundamental form of learning. While extensive research has focused on the response profiles of midbrain dopamine neurons during associative learning, less is known about learning-mediated changes in the afferents that shape their responses. We demonstrate that during critical phases of learning, anion homeostasis in midbrain GABA neurons - a primary source of input to dopamine neurons - is disrupted due to downregulation of the chloride transporter KCC2. This alteration in GABA neurons preferentially impacted lateral mesoaccumbal dopamine pathways and was not observed after learning was established. At the network level, learning-mediated KCC2 downregulation was associated with enhanced synchronization between individual GABA neurons and increased dopamine responses to reward-related stimuli. Conversely, enhancing KCC2 function during learning reduced GABA synchronization, diminished relevant dopamine signaling, and prevented cue-reward associations. Thus, circuit-specific adaptations in midbrain GABA neurons are crucial for forming new reward-related behaviors.
Collapse
|
35
|
Sansalone L, Evans RC, Twedell E, Zhang R, Khaliq ZM. Corticonigral projections recruit substantia nigra pars lateralis dopaminergic neurons for auditory threat memories. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.621665. [PMID: 39574768 PMCID: PMC11580856 DOI: 10.1101/2024.11.04.621665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
Dopaminergic neurons (DANs) in the lateral substantia nigra project to the tail of striatum (TS), which is involved in threat conditioning. Auditory cortex also contributes to threatening behaviors, but whether it directly interacts with midbrain DANs and how these interactions might influence threat conditioning remain unclear. Here, functional mapping revealed robust excitatory input from auditory and temporal association cortexes to substantia nigra pars lateralis (SNL) DANs, but not to pars compacta (SNc) DANs. SNL DANs exhibited unique firing patterns, with irregular pacemaking and higher maximal firing, reflecting different channel complements than SNc DANs. Behaviorally, inhibiting cortex to SNL projections impaired memory retrieval during auditory threat conditioning. Thus, we demonstrate robust corticonigral projections to SNL DANs, contrasting with previous observations of sparse cortical input to substantia nigra DANs. These findings distinguish SNL DANs from other nigral populations, highlighting their role in threatening behaviors and expanding knowledge of cortex to midbrain interactions.
Collapse
Affiliation(s)
- Lorenzo Sansalone
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, 20892
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Rebekah C. Evans
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, 20892
| | - Emily Twedell
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, 20892
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Renshu Zhang
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, 20892
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Zayd M. Khaliq
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, 20892
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| |
Collapse
|
36
|
Xi ZX, Bocarsly ME, Galaj E, Hempel B, Teresi C, Shaw M, Bi GH, Jordan C, Linz E, Alton H, Tanda G, Freyberg Z, Alvarez VA, Newman AH. Presynaptic and Postsynaptic Mesolimbic Dopamine D 3 Receptors Play Distinct Roles in Cocaine Versus Opioid Reward in Mice. Biol Psychiatry 2024; 96:752-765. [PMID: 38838841 PMCID: PMC11446657 DOI: 10.1016/j.biopsych.2024.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 04/23/2024] [Accepted: 05/08/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Past research has illuminated pivotal roles of dopamine D3 receptors (D3R) in the rewarding effects of cocaine and opioids. However, the cellular and neural circuit mechanisms that underlie these actions remain unclear. METHODS We employed Cre-LoxP techniques to selectively delete D3R from presynaptic dopamine neurons or postsynaptic dopamine D1 receptor (D1R)-expressing neurons in male and female mice. We utilized RNAscope in situ hybridization, immunohistochemistry, real-time polymerase chain reaction, voltammetry, optogenetics, microdialysis, and behavioral assays (n ≥ 8 animals per group) to functionally characterize the roles of presynaptic versus postsynaptic D3R in cocaine and opioid actions. RESULTS Our results revealed D3R expression in ∼25% of midbrain dopamine neurons and ∼70% of D1R-expressing neurons in the nucleus accumbens. While dopamine D2 receptors (D2R) were expressed in ∼80% dopamine neurons, we found no D2R and D3R colocalization among these cells. Selective deletion of D3R from dopamine neurons increased exploratory behavior in novel environments and enhanced pulse-evoked nucleus accumbens dopamine release. Conversely, deletion of D3R from D1R-expressing neurons attenuated locomotor responses to D1-like and D2-like agonists. Strikingly, deletion of D3R from either cell type reduced oxycodone self-administration and oxycodone-enhanced brain-stimulation reward. In contrast, neither of these D3R deletions impacted cocaine self-administration, cocaine-enhanced brain-stimulation reward, or cocaine-induced hyperlocomotion. Furthermore, D3R knockout in dopamine neurons reduced oxycodone-induced hyperactivity and analgesia, while deletion from D1R-expressing neurons potentiated opioid-induced hyperactivity without affecting analgesia. CONCLUSIONS We dissected presynaptic versus postsynaptic D3R function in the mesolimbic dopamine system. D2R and D3R are expressed in different populations of midbrain dopamine neurons, regulating dopamine release. Mesolimbic D3R are critically involved in the actions of opioids but not cocaine.
Collapse
Affiliation(s)
- Zheng-Xiong Xi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland.
| | - Miriam E Bocarsly
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, Bethesda, Maryland
| | - Ewa Galaj
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland
| | - Briana Hempel
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland
| | - Catherine Teresi
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, Bethesda, Maryland
| | - Marlisa Shaw
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, Bethesda, Maryland
| | - Guo-Hua Bi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland; Medication Development Program, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland
| | - Chloe Jordan
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland
| | - Emily Linz
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland; Medication Development Program, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland
| | - Hannah Alton
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland; Medication Development Program, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland
| | - Gianluigi Tanda
- Medication Development Program, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Veronica A Alvarez
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, Bethesda, Maryland; National Institute of Mental Health, Center on Compulsive Behaviors, Intramural Research Program, Bethesda, Maryland
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland.
| |
Collapse
|
37
|
Jung K, Krüssel S, Yoo S, An M, Burke B, Schappaugh N, Choi Y, Gu Z, Blackshaw S, Costa RM, Kwon HB. Dopamine-mediated formation of a memory module in the nucleus accumbens for goal-directed navigation. Nat Neurosci 2024; 27:2178-2192. [PMID: 39333785 PMCID: PMC11537966 DOI: 10.1038/s41593-024-01770-9] [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: 11/05/2021] [Accepted: 08/23/2024] [Indexed: 09/30/2024]
Abstract
Spatial memories guide navigation efficiently toward desired destinations. However, the neuronal and circuit mechanisms underlying the encoding of goal locations and its translation into goal-directed navigation remain unclear. Here we demonstrate that mice rapidly form a spatial memory of a shelter during shelter experiences, guiding escape behavior toward the goal location-a shelter-when under threat. Dopaminergic neurons in the ventral tegmental area and their projection to the nucleus accumbens (NAc) encode safety signals associated with the shelter. Optogenetically induced phasic dopamine signals are sufficient to create a place memory that directs escape navigation. Converging dopaminergic and hippocampal glutamatergic inputs to the NAc mediate the formation of a goal-related memory within a subpopulation of NAc neurons during shelter experiences. Artificial co-activation of this goal-related NAc ensemble with neurons in the dorsal periaqueductal gray was sufficient to trigger memory-guided, rather than random, escape behavior. These findings provide causal evidence of cognitive circuit modules linking memory with goal-directed action.
Collapse
Affiliation(s)
- Kanghoon Jung
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA.
- Allen Institute for Neural Dynamics, Seattle, WA, USA.
- Allen Institute, Seattle, WA, USA.
| | - Sarah Krüssel
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Sooyeon Yoo
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Myungmo An
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Benjamin Burke
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Nicholas Schappaugh
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Youngjin Choi
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Zirong Gu
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
- Department of Neuroscience, The University of Texas at Dallas, Richardson, Texas, USA
| | - Seth Blackshaw
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Rui M Costa
- Allen Institute, Seattle, WA, USA
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Hyung-Bae Kwon
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA.
| |
Collapse
|
38
|
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
|
39
|
Tian G, Bartas K, Hui M, Chen L, Vasquez JJ, Azouz G, Derdeyn P, Manville RW, Ho EL, Fang AS, Li Y, Tyler I, Setola V, Aoto J, Abbott GW, Beier KT. Molecular and circuit determinants in the globus pallidus mediating control of cocaine-induced behavioral plasticity. Neuron 2024; 112:3470-3485.e12. [PMID: 39153478 PMCID: PMC11502257 DOI: 10.1016/j.neuron.2024.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 04/12/2024] [Accepted: 07/22/2024] [Indexed: 08/19/2024]
Abstract
The globus pallidus externus (GPe) is a central component of the basal ganglia circuit that acts as a gatekeeper of cocaine-induced behavioral plasticity. However, the molecular and circuit mechanisms underlying this function are unknown. Here, we show that GPe parvalbumin-positive (GPePV) cells mediate cocaine responses by selectively modulating ventral tegmental area dopamine (VTADA) cells projecting to the dorsomedial striatum (DMS). Interestingly, GPePV cell activity in cocaine-naive mice is correlated with behavioral responses following cocaine, effectively predicting cocaine sensitivity. Expression of the voltage-gated potassium channels KCNQ3 and KCNQ5 that control intrinsic cellular excitability following cocaine was downregulated, contributing to the elevation in GPePV cell excitability. Acutely activating channels containing KCNQ3 and/or KCNQ5 using the small molecule carnosic acid, a key psychoactive component of Salvia rosmarinus (rosemary) extract, reduced GPePV cell excitability and impaired cocaine reward, sensitization, and volitional cocaine intake, indicating its therapeutic potential to counteract psychostimulant use disorder.
Collapse
Affiliation(s)
- Guilian Tian
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Katrina Bartas
- Program in Mathematical, Computational, and Systems Biology, University of California, Irvine, Irvine, CA, USA
| | - May Hui
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Lingxuan Chen
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Jose J Vasquez
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Ghalia Azouz
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Pieter Derdeyn
- Program in Mathematical, Computational, and Systems Biology, University of California, Irvine, Irvine, CA, USA
| | - Rían W Manville
- Bioelectricity Laboratory, Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Erick L Ho
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Amanda S Fang
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Yuan Li
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Isabella Tyler
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Vincent Setola
- Department of Neuroscience, West Virginia University, Morgantown, WV, USA; Department of Behavioral Medicine and Psychiatry, West Virginia University, Morgantown, WV, USA
| | - Jason Aoto
- University of Colorado Anschutz School of Medicine, Department of Pharmacology, Aurora, CO, USA
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Kevin T Beier
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA; Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
40
|
Fricker BA, Murugan M, Seifert AW, Kelly AM. Cingulate to septal circuitry facilitates the preference to affiliate with large peer groups. Curr Biol 2024; 34:4452-4463.e4. [PMID: 39265570 PMCID: PMC11486304 DOI: 10.1016/j.cub.2024.08.019] [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: 03/19/2024] [Revised: 07/17/2024] [Accepted: 08/13/2024] [Indexed: 09/14/2024]
Abstract
Despite the prevalence of large-group living across the animal kingdom, no studies have examined the neural mechanisms that make group living possible. Spiny mice, Acomys, have evolved to live in large groups and exhibit a preference to affiliate with large over small groups. Here, we determine the neural circuitry that facilitates the drive to affiliate with large groups. We first identify an anterior cingulate cortex (ACC) to lateral septum (LS) circuit that is more responsive to large than small groups of novel same-sex peers. Using chemogenetics, we then demonstrate that this circuit is necessary for both male and female group investigation preferences but only males' preference to affiliate with larger peer groups. Furthermore, inhibition of the ACC-LS circuit specifically impairs social, but not nonsocial, affiliative grouping preferences. These findings reveal a key circuit for the regulation of mammalian peer group affiliation.
Collapse
Affiliation(s)
- Brandon A Fricker
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA 30322, USA
| | - Malavika Murugan
- Department of Biology, Emory University, 1510 Clifton Road NE, Atlanta, GA 30322, USA
| | - Ashley W Seifert
- Department of Biology, University of Kentucky, 211 Thomas Hunt Morgan Building, Lexington, KY 40506, USA
| | - Aubrey M Kelly
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA 30322, USA.
| |
Collapse
|
41
|
Kleven H, Schlegel U, Groenewegen HJ, Leergaard TB, Bjerke IE. Comparison of basal ganglia regions across murine brain atlases using metadata models and the Waxholm Space. Sci Data 2024; 11:1036. [PMID: 39333155 PMCID: PMC11437236 DOI: 10.1038/s41597-024-03863-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 09/04/2024] [Indexed: 09/29/2024] Open
Abstract
The murine basal ganglia regions are targets for research into complex brain functions such as motor control and habit formation. However, there are several ways to name and annotate these regions, posing challenges for interpretation and comparison of data across studies. Here, we give an overview of basal ganglia terms and boundaries in the literature and reference atlases, and describe the criteria used for annotating these regions in the Waxholm Space rat brain atlas. We go on to compare basal ganglia annotations in stereotaxic rat brain atlases and the Allen Mouse brain Common Coordinate Framework to those in the Waxholm Space rat brain atlas. We demonstrate and describe considerable differences in the terms and boundaries of most basal ganglia regions across atlases and their versions. We also register information about atlases and regions in the openMINDS metadata framework, facilitating integration of data in neuroscience databases. The comparisons of terms and boundaries across rat and mouse atlases support analysis and interpretation of existing and new data from the basal ganglia.
Collapse
Affiliation(s)
- H Kleven
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - U Schlegel
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - H J Groenewegen
- Department of Anatomy and Neurosciences, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - T B Leergaard
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - I E Bjerke
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
42
|
Elum JE, Szelenyi ER, Juarez B, Murry AD, Loginov G, Zamorano CA, Gao P, Wu G, Ng-Evans S, Yee JX, Xu X, Golden SA, Zweifel LS. Distinct dynamics and intrinsic properties in ventral tegmental area populations mediate reward association and motivation. Cell Rep 2024; 43:114668. [PMID: 39207900 PMCID: PMC11514737 DOI: 10.1016/j.celrep.2024.114668] [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: 03/07/2024] [Revised: 07/04/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Ventral tegmental area (VTA) dopamine neurons regulate reward-related associative learning and reward-driven motivated behaviors, but how these processes are coordinated by distinct VTA neuronal subpopulations remains unresolved. Here, we compare the contribution of two primarily dopaminergic and largely non-overlapping VTA subpopulations, all VTA dopamine neurons and VTA GABAergic neurons of the mouse midbrain, to these processes. We find that the dopamine subpopulation that projects to the nucleus accumbens (NAc) core preferentially encodes reward-predictive cues and prediction errors. In contrast, the subpopulation that projects to the NAc shell preferentially encodes goal-directed actions and relative reward anticipation. VTA GABA neuron activity strongly contrasts VTA dopamine population activity and preferentially encodes reward outcome and retrieval. Electrophysiology, targeted optogenetics, and whole-brain input mapping reveal multiple convergent sources that contribute to the heterogeneity among VTA dopamine subpopulations that likely underlies their distinct encoding of reward-related associations and motivation that defines their functions in these contexts.
Collapse
Affiliation(s)
- Jordan E Elum
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA
| | - Eric R Szelenyi
- Department of Biological Structure, University of Washington, Seattle, WA, USA; University of Washington Center of Excellence in Neurobiology of Addiction, Pain, and Emotion (NAPE), Seattle, WA, USA
| | - Barbara Juarez
- Department of Neurobiology, University of Maryland, Baltimore, MD, USA
| | - Alexandria D Murry
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Grigory Loginov
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Catalina A Zamorano
- Department of Pharmacology, University of Washington, Seattle, WA, USA; University of Washington Center of Excellence in Neurobiology of Addiction, Pain, and Emotion (NAPE), Seattle, WA, USA
| | - Pan Gao
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Ginny Wu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Scott Ng-Evans
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
| | - Joshua X Yee
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Sam A Golden
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA; Department of Biological Structure, University of Washington, Seattle, WA, USA; University of Washington Center of Excellence in Neurobiology of Addiction, Pain, and Emotion (NAPE), Seattle, WA, USA
| | - Larry S Zweifel
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA; Department of Pharmacology, University of Washington, Seattle, WA, USA; Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA; University of Washington Center of Excellence in Neurobiology of Addiction, Pain, and Emotion (NAPE), Seattle, WA, USA.
| |
Collapse
|
43
|
Kim HD, Wei J, Call T, Ma X, Quintus NT, Summers AJ, Carotenuto S, Johnson R, Nguyen A, Cui Y, Park JG, Qiu S, Ferguson D. SIRT1 Coordinates Transcriptional Regulation of Neural Activity and Modulates Depression-Like Behaviors in the Nucleus Accumbens. Biol Psychiatry 2024; 96:495-505. [PMID: 38575105 PMCID: PMC11338727 DOI: 10.1016/j.biopsych.2024.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/16/2024] [Accepted: 03/25/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Major depression and anxiety disorders are significant causes of disability and socioeconomic burden. Despite the prevalence and considerable impact of these affective disorders, their pathophysiology remains elusive. Thus, there is an urgent need to develop novel therapeutics for these conditions. We evaluated the role of SIRT1 in regulating dysfunctional processes of reward by using chronic social defeat stress to induce depression- and anxiety-like behaviors. Chronic social defeat stress induces physiological and behavioral changes that recapitulate depression-like symptomatology and alters gene expression programs in the nucleus accumbens, but cell type-specific changes in this critical structure remain largely unknown. METHODS We examined transcriptional profiles of D1-expressing medium spiny neurons (MSNs) lacking deacetylase activity of SIRT1 by RNA sequencing in a cell type-specific manner using the RiboTag line of mice. We analyzed differentially expressed genes using gene ontology tools including SynGO and EnrichR and further demonstrated functional changes in D1-MSN-specific SIRT1 knockout (KO) mice using electrophysiological and behavioral measurements. RESULTS RNA sequencing revealed altered transcriptional profiles of D1-MSNs lacking functional SIRT1 and showed specific changes in synaptic genes including glutamatergic and GABAergic (gamma-aminobutyric acidergic) receptors in D1-MSNs. These molecular changes may be associated with decreased excitatory and increased inhibitory neural activity in Sirt1 KO D1-MSNs, accompanied by morphological changes. Moreover, the D1-MSN-specific Sirt1 KO mice exhibited proresilient changes in anxiety- and depression-like behaviors. CONCLUSIONS SIRT1 coordinates excitatory and inhibitory synaptic genes to regulate the GABAergic output tone of D1-MSNs. These findings reveal a novel signaling pathway that has potential for the development of innovative treatments for affective disorders.
Collapse
Affiliation(s)
- Hee-Dae Kim
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona
| | - Jing Wei
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona
| | - Tanessa Call
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona
| | - Xiaokuang Ma
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona
| | - Nicole Teru Quintus
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona
| | - Alexander J Summers
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona
| | - Samantha Carotenuto
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona
| | - Ross Johnson
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona
| | - Angel Nguyen
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona
| | - Yuehua Cui
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona
| | - Jin G Park
- Virginia G. Piper Biodesign Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Shenfeng Qiu
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona
| | - Deveroux Ferguson
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona.
| |
Collapse
|
44
|
Zhang C, Kúkeľová D, Sigrist H, Hengerer B, Kratzer RF, Mracek P, Omrani A, von Heimendahl M, Pryce CR. Orphan receptor-GPR52 inverse agonist efficacy in ameliorating chronic stress-related deficits in reward motivation and phasic accumbal dopamine activity in mice. Transl Psychiatry 2024; 14:363. [PMID: 39242529 PMCID: PMC11379876 DOI: 10.1038/s41398-024-03081-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 08/22/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024] Open
Abstract
Reward processing dysfunctions e.g., anhedonia, apathy, are common in stress-related neuropsychiatric disorders including depression and schizophrenia, and there are currently no established therapies. One potential therapeutic approach is restoration of reward anticipation during appetitive behavior, deficits in which co-occur with attenuated nucleus accumbens (NAc) activity, possibly due to NAc inhibition of mesolimbic dopamine (DA) signaling. Targeting NAc regulation of ventral tegmental area (VTA) DA neuron responsiveness to reward cues could involve either the direct or indirect-via ventral pallidium (VP)-pathways. One candidate is the orphan G protein-coupled receptor GPR52, expressed by DA receptor 2 NAc neurons that project to VP. In mouse brain-slice preparations, GPR52 inverse agonist (GPR52-IA) attenuated evoked inhibitory postsynaptic currents at NAc-VP neurons, which could disinhibit VTA DA neurons. A mouse model in which chronic social stress leads to reduced reward learning and effortful motivation was applied to investigate GPR52-IA behavioral effects. Control and chronically stressed mice underwent a discriminative learning test of tone-appetitive behavior-sucrose reinforcement: stress reduced appetitive responding and discriminative learning, and these anticipatory behaviors were dose-dependently reinstated by GPR52-IA. The same mice then underwent an effortful motivation test of operant behavior-tone-sucrose reinforcement: stress reduced effortful motivation and GPR52-IA dose-dependently restored it. In a new cohort, GRABDA-sensor fibre photometry was used to measure NAc DA activity during the motivation test: in stressed mice, reduced motivation co-occurred with attenuated NAc DA activity specifically to the tone that signaled reinforcement of effortful behavior, and GPR52-IA ameliorated both deficits. These findings: (1) Demonstrate preclinical efficacy of GPR52 inverse agonism for stress-related deficits in reward anticipation during appetitive behavior. (2) Suggest that GPR52-dependent disinhibition of the NAc-VP-VTA-NAc circuit, leading to increased phasic NAc DA signaling of earned incentive stimuli, could account for these clinically relevant effects.
Collapse
Affiliation(s)
- Chenfeng Zhang
- Preclinical Laboratory, Department of Adult Psychiatry and Psychotherapy, Psychiatric University Clinic and University of Zurich, Zurich, Switzerland
- Zurich Neuroscience Center, University of Zurich and ETH, Zurich, Switzerland
| | - Diana Kúkeľová
- Preclinical Laboratory, Department of Adult Psychiatry and Psychotherapy, Psychiatric University Clinic and University of Zurich, Zurich, Switzerland
| | - Hannes Sigrist
- Preclinical Laboratory, Department of Adult Psychiatry and Psychotherapy, Psychiatric University Clinic and University of Zurich, Zurich, Switzerland
| | - Bastian Hengerer
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Ramona F Kratzer
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Philipp Mracek
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Azar Omrani
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Christopher R Pryce
- Preclinical Laboratory, Department of Adult Psychiatry and Psychotherapy, Psychiatric University Clinic and University of Zurich, Zurich, Switzerland.
- Zurich Neuroscience Center, University of Zurich and ETH, Zurich, Switzerland.
| |
Collapse
|
45
|
Kuiper LB, Dawes MH, West AM, DiMarco EK, Galante EV, Kishida KT, Jones SR. Comparison of dopamine release and uptake parameters across sex, species and striatal subregions. Eur J Neurosci 2024; 60:5113-5140. [PMID: 39161062 PMCID: PMC11632670 DOI: 10.1111/ejn.16495] [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: 10/14/2023] [Revised: 07/05/2024] [Accepted: 07/24/2024] [Indexed: 08/21/2024]
Abstract
For over four decades, fast-scan cyclic voltammetry (FSCV) has been used to selectively measure neurotransmitters such as dopamine (DA) with high spatial and temporal resolution, providing detailed information about the regulation of DA in the extracellular space. FSCV is an optimal method for determining concentrations of stimulus-evoked DA in brain tissue. When modelling diseases involving disturbances in DA transmission, preclinical rodent models are especially useful because of the availability of specialized tools and techniques that serve as a foundation for translational research. There is known heterogeneity in DA dynamics between and within DA-innervated brain structures and between males and females. However, systematic evaluations of sex- and species-differences across multiple areas are lacking. Therefore, using FSCV, we captured a broad range of DA dynamics across five sub-regions of the dorsal and ventral striatum of males and females of both rats and mice that reflect the functional heterogeneity of DA kinetics and dynamics within these structures. While numerous differences were found, in particular, we documented a strong, consistent pattern of increased DA transporter activity in females in all of the regions surveyed. The data herein are intended to be used as a resource for further investigation of DA terminal function.
Collapse
Affiliation(s)
- Lindsey B. Kuiper
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Monica H. Dawes
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Alyssa M. West
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Emily K. DiMarco
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Emma V. Galante
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Kenneth T. Kishida
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Department of Biomedical Engineering, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Department of Neurosurgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Sara R. Jones
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
46
|
Simon RC, Loveless MC, Yee JX, Goh B, Cho SG, Nasir Z, Hashikawa K, Stuber GD, Zweifel LS, Soden ME. Opto-seq reveals input-specific immediate-early gene induction in ventral tegmental area cell types. Neuron 2024; 112:2721-2731.e5. [PMID: 38901431 PMCID: PMC11343674 DOI: 10.1016/j.neuron.2024.05.026] [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: 06/22/2023] [Revised: 03/18/2024] [Accepted: 05/27/2024] [Indexed: 06/22/2024]
Abstract
The ventral tegmental area (VTA) is a critical node in circuits governing motivated behavior and is home to diverse populations of neurons that release dopamine, gamma-aminobutyric acid (GABA), glutamate, or combinations of these neurotransmitters. The VTA receives inputs from many brain regions, but a comprehensive understanding of input-specific activation of VTA neuronal subpopulations is lacking. To address this, we combined optogenetic stimulation of select VTA inputs with single-nucleus RNA sequencing (snRNA-seq) and highly multiplexed in situ hybridization to identify distinct neuronal clusters and characterize their spatial distribution and activation patterns. Quantification of immediate-early gene (IEG) expression revealed that different inputs activated select VTA subpopulations, which demonstrated cell-type-specific transcriptional programs. Within dopaminergic subpopulations, IEG induction levels correlated with differential expression of ion channel genes. This new transcriptomics-guided circuit analysis reveals the diversity of VTA activation driven by distinct inputs and provides a resource for future analysis of VTA cell types.
Collapse
Affiliation(s)
- Rhiana C Simon
- Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195, USA; Center for the Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA
| | - Mary C Loveless
- Center for the Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Joshua X Yee
- Center for the Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Brandon Goh
- Center for the Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Su G Cho
- Center for the Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Zainab Nasir
- Center for the Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Koichi Hashikawa
- Center for the Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Garret D Stuber
- Center for the Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Larry S Zweifel
- Center for the Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Marta E Soden
- Center for the Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
47
|
Johnson NL, Cotelo-Larrea A, Stetzik LA, Akkaya UM, Zhang Z, Gadziola MA, Varga AG, Ma M, Wesson DW. Sniffing can be initiated by dopamine's actions on ventral striatum neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.19.581052. [PMID: 39229099 PMCID: PMC11370338 DOI: 10.1101/2024.02.19.581052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Sniffing is a motivated behavior displayed by nearly all terrestrial vertebrates. While sniffing is associated with acquiring and processing odors, sniffing is also intertwined with affective and motivated states. The neuromodulatory systems which influence the display of sniffing are unclear. Here, we report that dopamine release into the ventral striatum is coupled with bouts of sniffing and that stimulation of dopaminergic terminals in these regions drives increases in respiratory rate to initiate sniffing whereas inhibition of these terminals reduces respiratory rate. Both the firing of individual neurons and the activity of post-synaptic D1 and D2 receptor-expressing neurons in the ventral striatum are also coupled with sniffing and local antagonism of D1 and D2 receptors squelches sniffing. Together, these results support a model whereby sniffing can be initiated by dopamine's actions upon ventral striatum neurons. The nature of sniffing being integral to both olfaction and motivated behaviors implicates this circuit in a wide array of functions.
Collapse
|
48
|
Xu Y, Lin Y, Yu M, Zhou K. The nucleus accumbens in reward and aversion processing: insights and implications. Front Behav Neurosci 2024; 18:1420028. [PMID: 39184934 PMCID: PMC11341389 DOI: 10.3389/fnbeh.2024.1420028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/26/2024] [Indexed: 08/27/2024] Open
Abstract
The nucleus accumbens (NAc), a central component of the brain's reward circuitry, has been implicated in a wide range of behaviors and emotional states. Emerging evidence, primarily drawing from recent rodent studies, suggests that the function of the NAc in reward and aversion processing is multifaceted. Prolonged stress or drug use induces maladaptive neuronal function in the NAc circuitry, which results in pathological conditions. This review aims to provide comprehensive and up-to-date insights on the role of the NAc in motivated behavior regulation and highlights areas that demand further in-depth analysis. It synthesizes the latest findings on how distinct NAc neuronal populations and pathways contribute to the processing of opposite valences. The review examines how a range of neuromodulators, especially monoamines, influence the NAc's control over various motivational states. Furthermore, it delves into the complex underlying mechanisms of psychiatric disorders such as addiction and depression and evaluates prospective interventions to restore NAc functionality.
Collapse
Affiliation(s)
| | | | | | - Kuikui Zhou
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| |
Collapse
|
49
|
Marinescu AM, Labouesse MA. The nucleus accumbens shell: a neural hub at the interface of homeostatic and hedonic feeding. Front Neurosci 2024; 18:1437210. [PMID: 39139500 PMCID: PMC11319282 DOI: 10.3389/fnins.2024.1437210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/16/2024] [Indexed: 08/15/2024] Open
Abstract
Feeding behavior is a complex physiological process regulated by the interplay between homeostatic and hedonic feeding circuits. Among the neural structures involved, the nucleus accumbens (NAc) has emerged as a pivotal region at the interface of these two circuits. The NAc comprises distinct subregions and in this review, we focus mainly on the NAc shell (NAcSh). Homeostatic feeding circuits, primarily found in the hypothalamus, ensure the organism's balance in energy and nutrient requirements. These circuits monitor peripheral signals, such as insulin, leptin, and ghrelin, and modulate satiety and hunger states. The NAcSh receives input from these homeostatic circuits, integrating information regarding the organism's metabolic needs. Conversely, so-called hedonic feeding circuits involve all other non-hunger and -satiety processes, i.e., the sensory information, associative learning, reward, motivation and pleasure associated with food consumption. The NAcSh is interconnected with hedonics-related structures like the ventral tegmental area and prefrontal cortex and plays a key role in encoding hedonic information related to palatable food seeking or consumption. In sum, the NAcSh acts as a crucial hub in feeding behavior, integrating signals from both homeostatic and hedonic circuits, to facilitate behavioral output via its downstream projections. Moreover, the NAcSh's involvement extends beyond simple integration, as it directly impacts actions related to food consumption. In this review, we first focus on delineating the inputs targeting the NAcSh; we then present NAcSh output projections to downstream structures. Finally we discuss how the NAcSh regulates feeding behavior and can be seen as a neural hub integrating homeostatic and hedonic feeding signals, via a functionally diverse set of projection neuron subpopulations.
Collapse
Affiliation(s)
- Alina-Măriuca Marinescu
- Brain, Wire and Behavior Group, Translational Nutritional Biology Laboratory, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Marie A. Labouesse
- Brain, Wire and Behavior Group, Translational Nutritional Biology Laboratory, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
50
|
Engel L, Wolff AR, Blake M, Collins VL, Sinha S, Saunders BT. Dopamine neurons drive spatiotemporally heterogeneous striatal dopamine signals during learning. Curr Biol 2024; 34:3086-3101.e4. [PMID: 38925117 PMCID: PMC11279555 DOI: 10.1016/j.cub.2024.05.069] [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: 03/25/2024] [Revised: 04/25/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024]
Abstract
Environmental cues, through Pavlovian learning, become conditioned stimuli that invigorate and guide animals toward rewards. Dopamine (DA) neurons in the ventral tegmental area (VTA) and substantia nigra (SNc) are crucial for this process, via engagement of a reciprocally connected network with their striatal targets. Critically, it remains unknown how dopamine neuron activity itself engages dopamine signals throughout the striatum, across learning. Here, we investigated how optogenetic Pavlovian cue conditioning of VTA or SNc dopamine neurons directs cue-evoked behavior and shapes subregion-specific striatal dopamine dynamics. We used a fluorescent biosensor to monitor dopamine in the nucleus accumbens (NAc) core and shell, dorsomedial striatum (DMS), and dorsolateral striatum (DLS). We demonstrate spatially heterogeneous, learning-dependent dopamine changes across striatal regions. Although VTA stimulation-evoked robust dopamine release in NAc core, shell, and DMS, predictive cues preferentially recruited dopamine release in NAc core, starting early in training, and DMS, late in training. Negative prediction error signals, reflecting a violation in the expectation of dopamine neuron activation, only emerged in the NAc core and DMS. Despite the development of vigorous movement late in training, conditioned dopamine signals did not emerge in the DLS, even during Pavlovian conditioning with SNc dopamine neuron activation, which elicited robust DLS dopamine release. Together, our studies show a broad dissociation in the fundamental prediction and reward-related information generated by VTA and SNc dopamine neuron populations and signaled by dopamine across the striatum. Further, they offer new insight into how larger-scale adaptations across the striatal network emerge during learning to coordinate behavior.
Collapse
Affiliation(s)
- Liv Engel
- Department of Neuroscience, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA; Medical Discovery Team on Addiction, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA
| | - Amy R Wolff
- Department of Neuroscience, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA; Medical Discovery Team on Addiction, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA
| | - Madelyn Blake
- Department of Neuroscience, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA
| | - Val L Collins
- Department of Neuroscience, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA; Medical Discovery Team on Addiction, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA
| | - Sonal Sinha
- Krieger School of Arts & Sciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Benjamin T Saunders
- Department of Neuroscience, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA; Medical Discovery Team on Addiction, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA.
| |
Collapse
|