1
|
Zhao B, Liang L, Li J, Schaefke B, Wang L, Tseng YT. An escape-enhancing circuit involving subthalamic CRH neurons mediates stress-induced anhedonia in mice. Neurobiol Dis 2024; 200:106649. [PMID: 39187210 DOI: 10.1016/j.nbd.2024.106649] [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: 05/25/2024] [Revised: 07/18/2024] [Accepted: 08/22/2024] [Indexed: 08/28/2024] Open
Abstract
Chronic predator stress (CPS) is an important and ecologically relevant tool for inducing anhedonia in animals, but the neural circuits underlying the associated neurobiological changes remain to be identified. Using cell-type-specific manipulations, we found that corticotropin-releasing hormone (CRH) neurons in the medial subthalamic nucleus (mSTN) enhance struggle behaviors in inescapable situations and lead to anhedonia, predominately through projections to the external globus pallidus (GPe). Recordings of in vivo neuronal activity revealed that CPS distorted mSTN-CRH neuronal responsivity to negative and positive stimuli, which may underlie CPS-induced behavioral despair and anhedonia. Furthermore, we discovered presynaptic inputs from the bed nucleus of the stria terminalis (BNST) to mSTN-CRH neurons projecting to the GPe that were enhanced following CPS, and these inputs may mediate such behaviors. This study identifies a neurocircuitry that co-regulates escape response and anhedonia in response to predator stress. This new understanding of the neural basis of defensive behavior in response to predator stress will likely benefit our understanding of neuropsychiatric diseases.
Collapse
Affiliation(s)
- Binghao Zhao
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Lisha Liang
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jingfei Li
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Bernhard Schaefke
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Liping Wang
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Yu-Ting Tseng
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|
2
|
Peczely L, Grace AA. The dose-dependent effect of the D2R agonist quinpirole microinjected into the ventral pallidum on information flow in the limbic system. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111059. [PMID: 38901759 PMCID: PMC11348604 DOI: 10.1016/j.pnpbp.2024.111059] [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: 01/16/2024] [Revised: 04/28/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
The ventral pallidum (VP) receives its primary inputs from the nucleus accumbens (NAC) and the basolateral amygdala (BLA). We demonstrated recently that in the VP, the D2 DA receptor (D2R) agonist quinpirole dose-dependently facilitates memory consolidation in inhibitory avoidance and spatial learning. In the VP, D2R can be found both on NAC and BLA terminals. According to our hypothesis, quinpirole microinjected into the VP can facilitate memory consolidation via modulation of synaptic plasticity on NAC and/or BLA terminals. The effect of intra-VP quinpirole on BLA-VP and NAC shell-VP synapses was investigated via a high frequency stimulation (HFS) protocol. Quinpirole was administered in three doses into the VP of male Sprague-Dawley rats after HFS; controls received vehicle. To examine whether an interaction between the NAC shell and the BLA at the level of the VP was involved, tetrodotoxin (TTX) was microinjected into one of the nuclei while stimulating the other nucleus. Our results showed that quinpirole dose-dependently modulates BLA-VP and NAC shell-VP synapses, similar to those observed in inhibitory avoidance and spatial learning, respectively. The lower dose inhibits BLA inputs, while the larger doses facilitates NAC shell inputs. The experiments with TTX demonstrates that the two nuclei do not influence each others' evoked responses in the VP. Power spectral density analysis demonstrated that independent from the synaptic facilitation, intra-VP quinpirole increases the amplitude of gamma frequency band after NAC HFS, and BLA tonically suppresses the NAC's HFS-induced gamma facilitation. In contrast, HFS of the BLA results in a delayed, transient increase in the amplitude of the gamma frequency band correlating with the LTP of the P1 component of the VP response to BLA stimulation. Furthermore, our results demonstrate that the BLA plays a prominent role in the generation of the delta oscillations: HFS of the BLA leads to a gradually increasing delta frequency band facilitation over time, while BLA inhibition blocks the NAC's HFS induced strong delta facilitation. These findings demonstrate that there is a complex interaction between the NAC shell region and the VP, as well as the BLA and the VP, and support the important role of VP D2Rs in the regulation of limbic information flow.
Collapse
Affiliation(s)
- Laszlo Peczely
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA 15260, USA; Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary; Learning in Biological and Artificial Systems Research Group, Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary; Centre for Neuroscience, University of Pécs, Pécs, Hungary.
| | - Anthony A Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
3
|
Hughes BW, Huebschman JL, Tsvetkov E, Siemsen BM, Snyder KK, Akiki RM, Wood DJ, Penrod RD, Scofield MD, Berto S, Taniguchi M, Cowan CW. NPAS4 supports cocaine-conditioned cues in rodents by controlling the cell type-specific activation balance in the nucleus accumbens. Nat Commun 2024; 15:5971. [PMID: 39117647 PMCID: PMC11310321 DOI: 10.1038/s41467-024-50099-1] [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: 09/26/2022] [Accepted: 06/28/2024] [Indexed: 08/10/2024] Open
Abstract
Powerful associations that link drugs of abuse with cues in the drug-paired environment often serve as prepotent relapse triggers. Drug-associated contexts and cues activate ensembles of nucleus accumbens (NAc) neurons, including D1-class medium spiny neurons (MSNs) that typically promote, and D2-class MSNs that typically oppose, drug seeking. We found that in mice, cocaine conditioning upregulated transiently the activity-regulated transcription factor, Neuronal PAS Domain Protein 4 (NPAS4), in a small subset of NAc neurons. The NPAS4+ NAc ensemble was required for cocaine conditioned place preference. We also observed that NPAS4 functions within NAc D2-, but not D1-, MSNs to support cocaine-context associations and cue-induced cocaine, but not sucrose, seeking. Together, our data show that the NPAS4+ ensemble of NAc neurons is essential for cocaine-context associations in mice, and that NPAS4 itself functions in NAc D2-MSNs to support cocaine-context associations by suppressing drug-induced counteradaptations that oppose relapse-related behaviour.
Collapse
Affiliation(s)
- Brandon W Hughes
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Jessica L Huebschman
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Evgeny Tsvetkov
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Benjamin M Siemsen
- Department of Anesthesiology, Medical University of South Carolina, Charleston, SC, USA
| | - Kirsten K Snyder
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Rose Marie Akiki
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
- Medical Scientist Training Program, Medical University of South Carolina, Charleston, SC, USA
| | - Daniel J Wood
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
- Medical Scientist Training Program, Medical University of South Carolina, Charleston, SC, USA
| | - Rachel D Penrod
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Michael D Scofield
- Department of Anesthesiology, Medical University of South Carolina, Charleston, SC, USA
| | - Stefano Berto
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Makoto Taniguchi
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
| | - Christopher W Cowan
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
4
|
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
|
5
|
Teague CD, Markovic T, Zhou X, Martinez-Rivera FJ, Minier-Toribio A, Zinsmaier A, Pulido NV, Schmidt KH, Lucerne KE, Godino A, van der Zee YY, Ramakrishnan A, Futamura R, Browne CJ, Holt LM, Yim YY, Azizian CH, Walker DM, Shen L, Dong Y, Zhang B, Nestler EJ. Circuit-Wide Gene Network Analysis Reveals Sex-Specific Roles for Phosphodiesterase 1b in Cocaine Addiction. J Neurosci 2024; 44:e1327232024. [PMID: 38637154 PMCID: PMC11154853 DOI: 10.1523/jneurosci.1327-23.2024] [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: 07/16/2023] [Revised: 03/21/2024] [Accepted: 04/01/2024] [Indexed: 04/20/2024] Open
Abstract
Cocaine use disorder is a significant public health issue without an effective pharmacological treatment. Successful treatments are hindered in part by an incomplete understanding of the molecular mechanisms that underlie long-lasting maladaptive plasticity and addiction-like behaviors. Here, we leverage a large RNA sequencing dataset to generate gene coexpression networks across six interconnected regions of the brain's reward circuitry from mice that underwent saline or cocaine self-administration. We identify phosphodiesterase 1b (Pde1b), a Ca2+/calmodulin-dependent enzyme that increases cAMP and cGMP hydrolysis, as a central hub gene within a nucleus accumbens (NAc) gene module that was bioinformatically associated with addiction-like behavior. Chronic cocaine exposure increases Pde1b expression in NAc D2 medium spiny neurons (MSNs) in male but not female mice. Viral-mediated Pde1b overexpression in NAc reduces cocaine self-administration in female rats but increases seeking in both sexes. In female mice, overexpressing Pde1b in D1 MSNs attenuates the locomotor response to cocaine, with the opposite effect in D2 MSNs. Overexpressing Pde1b in D1/D2 MSNs had no effect on the locomotor response to cocaine in male mice. At the electrophysiological level, Pde1b overexpression reduces sEPSC frequency in D1 MSNs and regulates the excitability of NAc MSNs. Lastly, Pde1b overexpression significantly reduced the number of differentially expressed genes (DEGs) in NAc following chronic cocaine, with discordant effects on gene transcription between sexes. Together, we identify novel gene modules across the brain's reward circuitry associated with addiction-like behavior and explore the role of Pde1b in regulating the molecular, cellular, and behavioral responses to cocaine.
Collapse
Affiliation(s)
- Collin D Teague
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Tamara Markovic
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Xianxiao Zhou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Freddyson J Martinez-Rivera
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Angelica Minier-Toribio
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Alexander Zinsmaier
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Nathalia V Pulido
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Kyra H Schmidt
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Kelsey E Lucerne
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Arthur Godino
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Yentl Y van der Zee
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Rita Futamura
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Caleb J Browne
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Leanne M Holt
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Yun Young Yim
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Corrine H Azizian
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Deena M Walker
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon 97239
| | - Li Shen
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| |
Collapse
|
6
|
Morais-Silva G, Lobo MK. Refining the circuits of drug addiction: The ventral pallidum. Curr Opin Neurobiol 2024; 86:102883. [PMID: 38815544 DOI: 10.1016/j.conb.2024.102883] [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: 03/15/2024] [Revised: 05/05/2024] [Accepted: 05/09/2024] [Indexed: 06/01/2024]
Abstract
The ventral pallidum is a prominent structure within the basal ganglia, regulating reward and motivational processes. Positioned at the interface between motor and limbic structures, its function is crucial to the development and maintenance of substance use disorders. Chronic drug use induces neuroplastic events in this structure, leading to long-term changes in VP neuronal activity and synaptic communication. Moreover, different neuronal populations within the VP drive drug-seeking behavior in opposite directions. This review explores the role of the VP as a hub for reward, motivation, and aversion, establishing it as an important contributor to the pathophysiology of substance use disorders.
Collapse
Affiliation(s)
- Gessynger Morais-Silva
- Sao Paulo State University (UNESP), School of Pharmaceutical Sciences, Laboratory of Pharmacology, Araraquara, SP, Brazil. https://twitter.com/gessynger
| | - Mary Kay Lobo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
7
|
Kobayashi H, Fuchikami M, Oga K, Miyagi T, Fujita S, Fujita S, Okada S, Okamoto Y, Morinobu S. Neonatal Isolation Increases the Susceptibility to Learned Helplessness through the Aberrant Neuronal Activity in the Ventral Pallidum of Rats. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2024; 22:354-363. [PMID: 38627082 PMCID: PMC11024705 DOI: 10.9758/cpn.23.1140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/26/2023] [Accepted: 12/27/2023] [Indexed: 04/20/2024]
Abstract
Objective : Environmental deprivation, a type of childhood maltreatment, has been reported to constrain the cognitive developmental processes such as associative learning and implicit learning, which may lead to functional and morphological changes in the ventral pallidum (VP) and pessimism, a well-known cognitive feature of major depression. We examined whether neonatal isolation (NI) could influence the incidence of learned helplessness (LH) in a rat model mimicking the pessimism, and the number of vesicular glutamate transporter 2 (VGLUT2)-expressing VP cells and Penk-expressing VP cells. Methods : The number of escape failures from foot-shocks in the LH test was measured to examine stress-induced depression-like behavior in rats. The number of VGLUT2-expressing VP cells and Penk-expressing VP cells was measured by immunohistochemistry. Results : In NI rats compared with Sham rats, the incidence of LH in adulthood was increased and VGLUT2-expressing VP cells but not Penk-expressing VP cells in adulthood were decreased. VGLUT2-expressing VP cells were decreased only in the LH group of NI rats and significantly correlated with the escape latency in the LH test. Conclusion : These findings suggest that the aberrant VP neuronal activity due to environmental deprivation early in life leads to pessimistic associative and implicit learning. Modulating VP neuronal activity could be a novel therapeutic and preventive strategy for the patients with this specific pathophysiology.
Collapse
Affiliation(s)
- Hironori Kobayashi
- Division of Graduate School of Biomedical and Health Sciences, Department of Psychiatry and Neuroscience, Hiroshima University, Hiroshima, Japan
| | - Manabu Fuchikami
- Division of Graduate School of Biomedical and Health Sciences, Department of Psychiatry and Neuroscience, Hiroshima University, Hiroshima, Japan
| | - Kenichi Oga
- Division of Graduate School of Biomedical and Health Sciences, Department of Psychiatry and Neuroscience, Hiroshima University, Hiroshima, Japan
| | - Tatsuhiro Miyagi
- Division of Graduate School of Biomedical and Health Sciences, Department of Psychiatry and Neuroscience, Hiroshima University, Hiroshima, Japan
| | - Sho Fujita
- Division of Graduate School of Biomedical and Health Sciences, Department of Psychiatry and Neuroscience, Hiroshima University, Hiroshima, Japan
| | - Satoshi Fujita
- Division of Graduate School of Biomedical and Health Sciences, Department of Psychiatry and Neuroscience, Hiroshima University, Hiroshima, Japan
| | - Satoshi Okada
- Division of Graduate School of Biomedical and Health Sciences, Department of Psychiatry and Neuroscience, Hiroshima University, Hiroshima, Japan
| | - Yasumasa Okamoto
- Division of Graduate School of Biomedical and Health Sciences, Department of Psychiatry and Neuroscience, Hiroshima University, Hiroshima, Japan
| | - Shigeru Morinobu
- Department of Occupational Therapy, School of Health Science and Social Welfare, Kibi International University, Takahashi, Japan
| |
Collapse
|
8
|
Faget L, Oriol L, Lee WC, Zell V, Sargent C, Flores A, Hollon NG, Ramanathan D, Hnasko TS. Ventral pallidum GABA and glutamate neurons drive approach and avoidance through distinct modulation of VTA cell types. Nat Commun 2024; 15:4233. [PMID: 38762463 PMCID: PMC11102457 DOI: 10.1038/s41467-024-48340-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: 05/19/2023] [Accepted: 04/26/2024] [Indexed: 05/20/2024] Open
Abstract
The ventral pallidum (VP) contains GABA and glutamate neurons projecting to ventral tegmental area (VTA) whose stimulation drives approach and avoidance, respectively. Yet little is known about the mechanisms by which VP cell types shape VTA activity and drive behavior. Here, we found that both VP GABA and glutamate neurons were activated during approach to reward or by delivery of an aversive stimulus. Stimulation of VP GABA neurons inhibited VTA GABA, but activated dopamine and glutamate neurons. Remarkably, stimulation-evoked activation was behavior-contingent such that VTA recruitment was inhibited when evoked by the subject's own action. Conversely, VP glutamate neurons activated VTA GABA, as well as dopamine and glutamate neurons, despite driving aversion. However, VP glutamate neurons evoked dopamine in aversion-associated ventromedial nucleus accumbens (NAc), but reduced dopamine release in reward-associated dorsomedial NAc. These findings show how heterogeneous VP projections to VTA can be engaged to shape approach and avoidance behaviors.
Collapse
Affiliation(s)
- Lauren Faget
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA.
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.
| | - Lucie Oriol
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Wen-Chun Lee
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Vivien Zell
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Cody Sargent
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Andrew Flores
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Nick G Hollon
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Dhakshin Ramanathan
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Thomas S Hnasko
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA.
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.
| |
Collapse
|
9
|
Yamada D, Davidson AM, Hige T. Cyclic nucleotide-induced bidirectional long-term synaptic plasticity in Drosophila mushroom body. J Physiol 2024; 602:2019-2045. [PMID: 38488688 PMCID: PMC11068490 DOI: 10.1113/jp285745] [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: 09/30/2023] [Accepted: 02/21/2024] [Indexed: 03/26/2024] Open
Abstract
Activation of the cAMP pathway is one of the common mechanisms underlying long-term potentiation (LTP). In the Drosophila mushroom body, simultaneous activation of odour-coding Kenyon cells (KCs) and reinforcement-coding dopaminergic neurons activates adenylyl cyclase in KC presynaptic terminals, which is believed to trigger synaptic plasticity underlying olfactory associative learning. However, learning induces long-term depression (LTD) at these synapses, contradicting the universal role of cAMP as a facilitator of transmission. Here, we developed a system to electrophysiologically monitor both short-term and long-term synaptic plasticity at KC output synapses and demonstrated that they are indeed an exception in which activation of the cAMP-protein kinase A pathway induces LTD. Contrary to the prevailing model, our cAMP imaging found no evidence for synergistic action of dopamine and KC activity on cAMP synthesis. Furthermore, we found that forskolin-induced cAMP increase alone was insufficient for plasticity induction; it additionally required simultaneous KC activation to replicate the presynaptic LTD induced by pairing with dopamine. On the other hand, activation of the cGMP pathway paired with KC activation induced slowly developing LTP, proving antagonistic actions of the two second-messenger pathways predicted by behavioural study. Finally, KC subtype-specific interrogation of synapses revealed that different KC subtypes exhibit distinct plasticity duration even among synapses on the same postsynaptic neuron. Thus, our work not only revises the role of cAMP in synaptic plasticity by uncovering the unexpected convergence point of the cAMP pathway and neuronal activity, but also establishes the methods to address physiological mechanisms of synaptic plasticity in this important model. KEY POINTS: Although presynaptic cAMP increase generally facilitates synapses, olfactory associative learning in Drosophila, which depends on dopamine and cAMP signalling genes, induces long-term depression (LTD) at the mushroom body output synapses. By combining electrophysiology, pharmacology and optogenetics, we directly demonstrate that these synapses are an exception where activation of the cAMP-protein kinase A pathway leads to presynaptic LTD. Dopamine- or forskolin-induced cAMP increase alone is not sufficient for LTD induction; neuronal activity, which has been believed to trigger cAMP synthesis in synergy with dopamine input, is required in the downstream pathway of cAMP. In contrast to cAMP, activation of the cGMP pathway paired with neuronal activity induces presynaptic long-term potentiation, which explains behaviourally observed opposing actions of transmitters co-released by dopaminergic neurons. Our work not only revises the role of cAMP in synaptic plasticity, but also provides essential methods to address physiological mechanisms of synaptic plasticity in this important model system.
Collapse
Affiliation(s)
- Daichi Yamada
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Andrew M. Davidson
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States
- Integrative Program for Biological and Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Toshihide Hige
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States
- Integrative Program for Biological and Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, United States
| |
Collapse
|
10
|
Bernat N, Campbell RR, Nam H, Basu M, Odesser T, Elyasaf G, Engeln M, Chandra R, Golden S, Ament S, Lobo MK, Kupchik YM. Multimodal Interrogation of Ventral Pallidum Projections Reveals Projection-Specific Signatures and Effects on Cocaine Reward. J Neurosci 2024; 44:e1469232024. [PMID: 38485256 PMCID: PMC11063828 DOI: 10.1523/jneurosci.1469-23.2024] [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: 08/02/2023] [Revised: 02/29/2024] [Accepted: 03/03/2024] [Indexed: 03/26/2024] Open
Abstract
The ventral pallidum (VP) is a central hub in the reward circuitry with diverse projections that have different behavioral roles attributed mostly to the connectivity with the downstream target. However, different VP projections may represent, as in the striatum, separate neuronal populations that differ in more than just connectivity. In this study, we performed in mice of both sexes a multimodal dissection of four major projections of the VP-to the lateral hypothalamus (VP→LH), ventral tegmental area (VP→VTA), lateral habenula (VP→LHb), and mediodorsal thalamus (VP→MDT)-with physiological, anatomical, genetic, and behavioral tools. We also tested for physiological differences between VP neurons receiving input from nucleus accumbens medium spiny neurons (MSNs) that express either the D1 (D1-MSNs) or the D2 (D2-MSNs) dopamine receptor. We show that each VP projection (1) when inhibited during a cocaine conditioned place preference (CPP) test affects performance differently, (2) receives a different pattern of inputs using rabies retrograde labeling, (3) shows differentially expressed genes using RNA sequencing, and (4) has projection-specific characteristics in excitability and synaptic input characteristics using whole-cell patch clamp. VP→LH and VP→VTA projections have different effects on CPP and show low overlap in circuit tracing experiments, as VP→VTA neurons receive more striatal input, while VP→LH neurons receive more olfactory input. Additionally, VP→VTA neurons are less excitable, while VP→LH neurons are more excitable than the average VP neuron, a difference driven mainly by D2-MSN-responding neurons. Thus, VP→VTA and VP→LH neurons may represent largely distinct populations of VP neurons.
Collapse
Affiliation(s)
- Nimrod Bernat
- Department of Medical Neurobiology, Faculty of Medicine, The Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
- IMRIC Center for Addiction Research (ICARe), The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Rianne R Campbell
- Departments of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Hyungwoo Nam
- Departments of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Mahashweta Basu
- Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Tal Odesser
- Department of Medical Neurobiology, Faculty of Medicine, The Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Gal Elyasaf
- Department of Medical Neurobiology, Faculty of Medicine, The Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Michel Engeln
- Departments of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
- CNRS, INCIA, UMR 5287, University of Bordeaux, Bordeaux F-33000, France
| | - Ramesh Chandra
- Departments of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Shana Golden
- Department of Medical Neurobiology, Faculty of Medicine, The Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Seth Ament
- Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Mary Kay Lobo
- Departments of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Yonatan M Kupchik
- Department of Medical Neurobiology, Faculty of Medicine, The Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
- IMRIC Center for Addiction Research (ICARe), The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| |
Collapse
|
11
|
Zachry JE, Kutlu MG, Yoon HJ, Leonard MZ, Chevée M, Patel DD, Gaidici A, Kondev V, Thibeault KC, Bethi R, Tat J, Melugin PR, Isiktas AU, Joffe ME, Cai DJ, Conn PJ, Grueter BA, Calipari ES. D1 and D2 medium spiny neurons in the nucleus accumbens core have distinct and valence-independent roles in learning. Neuron 2024; 112:835-849.e7. [PMID: 38134921 PMCID: PMC10939818 DOI: 10.1016/j.neuron.2023.11.023] [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: 12/20/2022] [Revised: 10/03/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023]
Abstract
At the core of value-based learning is the nucleus accumbens (NAc). D1- and D2-receptor-containing medium spiny neurons (MSNs) in the NAc core are hypothesized to have opposing valence-based roles in behavior. Using optical imaging and manipulation approaches in mice, we show that neither D1 nor D2 MSNs signal valence. D1 MSN responses were evoked by stimuli regardless of valence or contingency. D2 MSNs were evoked by both cues and outcomes, were dynamically changed with learning, and tracked valence-free prediction error at the population and individual neuron level. Finally, D2 MSN responses to cues were necessary for associative learning. Thus, D1 and D2 MSNs work in tandem, rather than in opposition, by signaling specific properties of stimuli to control learning.
Collapse
Affiliation(s)
- Jennifer E Zachry
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Munir Gunes Kutlu
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Hye Jean Yoon
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Michael Z Leonard
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Maxime Chevée
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Dev D Patel
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Anthony Gaidici
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Veronika Kondev
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Kimberly C Thibeault
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Rishik Bethi
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Jennifer Tat
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Patrick R Melugin
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Atagun U Isiktas
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA; Department of Neuroscience, Yale University, New Haven, CT 06520, USA
| | - Max E Joffe
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Denise J Cai
- Nash Family Department of Neuroscience, Icahn School of Medicine, Mount Sinai, New York, NY 10029, USA
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Brad A Grueter
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA; Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Erin S Calipari
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
12
|
Le Merrer J, Detraux B, Gandía J, De Groote A, Fonteneau M, de Kerchove d'Exaerde A, Becker JAJ. Balance Between Projecting Neuronal Populations of the Nucleus Accumbens Controls Social Behavior in Mice. Biol Psychiatry 2024; 95:123-135. [PMID: 37207936 DOI: 10.1016/j.biopsych.2023.05.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/06/2023] [Accepted: 05/02/2023] [Indexed: 05/21/2023]
Abstract
BACKGROUND Deficient social interactions are a hallmark of major neuropsychiatric disorders, and accumulating evidence points to altered social reward and motivation as key underlying mechanisms of these pathologies. In the present study, we further explored the role of the balance of activity between D1 and D2 receptor-expressing striatal projection neurons (D1R- and D2R-SPNs) in the control of social behavior, challenging the hypothesis that excessive D2R-SPN activity, rather than deficient D1R-SPN activity, compromises social behavior. METHODS We selectively ablated D1R- and D2R-SPNs using an inducible diphtheria toxin receptor-mediated cell targeting strategy and assessed social behavior as well as repetitive/perseverative behavior, motor function, and anxiety levels. We tested the effects of optogenetic stimulation of D2R-SPNs in the nucleus accumbens (NAc) and pharmacological compounds repressing D2R-SPN. RESULTS Targeted deletion of D1R-SPNs in the NAc blunted social behavior in mice, facilitated motor skill learning, and increased anxiety levels. These behaviors were normalized by pharmacological inhibition of D2R-SPN, which also repressed transcription in the efferent nucleus, the ventral pallidum. Ablation of D1R-SPNs in the dorsal striatum had no impact on social behavior but impaired motor skill learning and decreased anxiety levels. Deletion of D2R-SPNs in the NAc produced motor stereotypies but facilitated social behavior and impaired motor skill learning. We mimicked excessive D2R-SPN activity by optically stimulating D2R-SPNs in the NAc and observed a severe deficit in social interaction that was prevented by D2R-SPN pharmacological inhibition. CONCLUSIONS Repressing D2R-SPN activity may represent a promising therapeutic strategy to relieve social deficits in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Julie Le Merrer
- Physiologie de la Reproduction et des Comportements, Unité Mixte de Recherche Centre National de la Recherche Scientifique 7247, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement 0085, Institut National de la Santé et de la Recherche Médicale, Université de Tours, Nouzilly, France; iBrain, Unité Mixte de Recherche 1253 Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Faculté des Sciences et Techniques, Université de Tours, Tours, France.
| | - Bérangère Detraux
- Neurophy Lab, ULB Neuroscience Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Jorge Gandía
- Physiologie de la Reproduction et des Comportements, Unité Mixte de Recherche Centre National de la Recherche Scientifique 7247, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement 0085, Institut National de la Santé et de la Recherche Médicale, Université de Tours, Nouzilly, France
| | - Aurélie De Groote
- Neurophy Lab, ULB Neuroscience Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Mathieu Fonteneau
- iBrain, Unité Mixte de Recherche 1253 Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Faculté des Sciences et Techniques, Université de Tours, Tours, France
| | - Alban de Kerchove d'Exaerde
- Neurophy Lab, ULB Neuroscience Institute, Université Libre de Bruxelles, Brussels, Belgium; WELBIO, Wavre, Belgium.
| | - Jérôme A J Becker
- Physiologie de la Reproduction et des Comportements, Unité Mixte de Recherche Centre National de la Recherche Scientifique 7247, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement 0085, Institut National de la Santé et de la Recherche Médicale, Université de Tours, Nouzilly, France; iBrain, Unité Mixte de Recherche 1253 Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Faculté des Sciences et Techniques, Université de Tours, Tours, France
| |
Collapse
|
13
|
Yamada D, Davidson AM, Hige T. Cyclic nucleotide-induced bidirectional long-term synaptic plasticity in Drosophila mushroom body. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.28.560058. [PMID: 37808762 PMCID: PMC10557778 DOI: 10.1101/2023.09.28.560058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Activation of the cAMP pathway is one of the common mechanisms underlying long-term potentiation (LTP). In the Drosophila mushroom body, simultaneous activation of odor-coding Kenyon cells (KCs) and reinforcement-coding dopaminergic neurons activates adenylyl cyclase in KC presynaptic terminals, which is believed to trigger synaptic plasticity underlying olfactory associative learning. However, learning induces long-term depression (LTD) at these synapses, contradicting the universal role of cAMP as a facilitator of transmission. Here, we develop a system to electrophysiologically monitor both short-term and long-term synaptic plasticity at KC output synapses and demonstrate that they are indeed an exception where activation of the cAMP/protein kinase A pathway induces LTD. Contrary to the prevailing model, our cAMP imaging finds no evidence for synergistic action of dopamine and KC activity on cAMP synthesis. Furthermore, we find that forskolin-induced cAMP increase alone is insufficient for plasticity induction; it additionally requires simultaneous KC activation to replicate the presynaptic LTD induced by pairing with dopamine. On the other hand, activation of the cGMP pathway paired with KC activation induces slowly developing LTP, proving antagonistic actions of the two second-messenger pathways predicted by behavioral study. Finally, KC subtype-specific interrogation of synapses reveals that different KC subtypes exhibit distinct plasticity duration even among synapses on the same postsynaptic neuron. Thus, our work not only revises the role of cAMP in synaptic plasticity by uncovering the unexpected convergence point of the cAMP pathway and neuronal activity, but also establishes the methods to address physiological mechanisms of synaptic plasticity in this important model.
Collapse
Affiliation(s)
- Daichi Yamada
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Andrew M. Davidson
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States
- Integrative Program for Biological and Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Toshihide Hige
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States
- Integrative Program for Biological and Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, United States
| |
Collapse
|
14
|
Fang LZ, Creed MC. Updating the striatal-pallidal wiring diagram. Nat Neurosci 2024; 27:15-27. [PMID: 38057614 DOI: 10.1038/s41593-023-01518-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: 08/15/2021] [Accepted: 11/06/2023] [Indexed: 12/08/2023]
Abstract
The striatal and pallidal complexes are basal ganglia structures that orchestrate learning and execution of flexible behavior. Models of how the basal ganglia subserve these functions have evolved considerably, and the advent of optogenetic and molecular tools has shed light on the heterogeneity of subcircuits within these pathways. However, a synthesis of how molecularly diverse neurons integrate into existing models of basal ganglia function is lacking. Here, we provide an overview of the neurochemical and molecular diversity of striatal and pallidal neurons and synthesize recent circuit connectivity studies in rodents that takes this diversity into account. We also highlight anatomical organizational principles that distinguish the dorsal and ventral basal ganglia pathways in rodents. Future work integrating the molecular and anatomical properties of striatal and pallidal subpopulations may resolve controversies regarding basal ganglia network function.
Collapse
Affiliation(s)
- Lisa Z Fang
- Washington University Pain Center, Department of Anesthesiology, St. Louis, MO, USA
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, Newfoundland and Labrador, Canada
| | - Meaghan C Creed
- Washington University Pain Center, Department of Anesthesiology, St. Louis, MO, USA.
- Departments of Psychiatry, Neuroscience and Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
15
|
Swinford-Jackson SE, Rich MT, Huffman PJ, Knouse MC, Thomas AS, Mankame S, Worobey SJ, Pierce RC. Low frequency deep brain stimulation of nucleus accumbens shell neuronal subpopulations attenuates cocaine seeking selectively in male rats. ADDICTION NEUROSCIENCE 2023; 9:100133. [PMID: 38312329 PMCID: PMC10836638 DOI: 10.1016/j.addicn.2023.100133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
The present study examined the effect of deep brain stimulation (DBS) in the nucleus accumbens shell on cocaine seeking and neuronal plasticity in rats. Electrical DBS of the accumbens shell attenuated cocaine primed reinstatement across a range of frequencies as low as 12 Hz in male rats. Nucleus accumbens medium spiny neurons (MSNs) can be differentiated by expression of dopamine D1 receptors (D1DRs) or D2DRs. Low-frequency optogenetic-DBS in D1DR- or D2DR-containing neurons attenuated cocaine seeking in male but not female rats. In slice electrophysiology experiments, 12 Hz electrical stimulation evoked long term potentiation (LTP) in D1DR-MSNs and D2DR-MSNs from cocaine naive male and female rats. However, in cocaine-experienced rats, electrical and optical DBS only elicited LTP in D2DR-MSNs from male rats. These results suggest that low frequency DBS in the nucleus accumbens shell effectively, but sex-specifically, suppresses cocaine seeking, which may be associated with the reversal of synaptic plasticity deficits in D2DR-MSNs.
Collapse
Affiliation(s)
- Sarah E. Swinford-Jackson
- Brain Health Institute and Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 USA
- Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Matthew T. Rich
- Brain Health Institute and Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 USA
- Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Phillip J. Huffman
- Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Melissa C. Knouse
- Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Arthur S. Thomas
- Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Sharvari Mankame
- Brain Health Institute and Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 USA
| | - Samantha J. Worobey
- Brain Health Institute and Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 USA
| | - R. Christopher Pierce
- Brain Health Institute and Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 USA
- Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| |
Collapse
|
16
|
Serra GP, Guillaumin A, Vlcek B, Delgado-Zabalza L, Ricci A, Rubino E, Dumas S, Baufreton J, Georges F, Wallén-Mackenzie Å. A role for the subthalamic nucleus in aversive learning. Cell Rep 2023; 42:113328. [PMID: 37925641 DOI: 10.1016/j.celrep.2023.113328] [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: 02/27/2023] [Revised: 07/28/2023] [Accepted: 10/08/2023] [Indexed: 11/07/2023] Open
Abstract
The subthalamic nucleus (STN) is critical for behavioral control; its dysregulation consequently correlated with neurological and neuropsychiatric disorders, including Parkinson's disease. Deep brain stimulation (DBS) targeting the STN successfully alleviates parkinsonian motor symptoms. However, low mood and depression are affective side effects. STN is adjoined with para-STN, associated with appetitive and aversive behavior. DBS aimed at STN might unintentionally modulate para-STN, causing aversion. Alternatively, the STN mediates aversion. To investigate causality between STN and aversion, affective behavior is addressed using optogenetics in mice. Selective promoters allow dissociation of STN (e.g., Pitx2) vs. para-STN (Tac1). Acute photostimulation results in aversion via both STN and para-STN. However, only STN stimulation-paired cues cause conditioned avoidance and only STN stimulation interrupts on-going sugar self-administration. Electrophysiological recordings identify post-synaptic responses in pallidal neurons, and selective photostimulation of STN terminals in the ventral pallidum replicates STN-induced aversion. Identifying STN as a source of aversive learning contributes neurobiological underpinnings to emotional affect.
Collapse
Affiliation(s)
- Gian Pietro Serra
- Uppsala University, Department of Organism Biology, 752 36 Uppsala, Sweden
| | - Adriane Guillaumin
- Uppsala University, Department of Organism Biology, 752 36 Uppsala, Sweden; University of Bordeaux, CNRS, IMN, UMR 5293, 33000 Bordeaux, France
| | - Bianca Vlcek
- Uppsala University, Department of Organism Biology, 752 36 Uppsala, Sweden
| | | | - Alessia Ricci
- Uppsala University, Department of Organism Biology, 752 36 Uppsala, Sweden
| | - Eleonora Rubino
- Uppsala University, Department of Organism Biology, 752 36 Uppsala, Sweden
| | | | - Jérôme Baufreton
- University of Bordeaux, CNRS, IMN, UMR 5293, 33000 Bordeaux, France
| | - François Georges
- University of Bordeaux, CNRS, IMN, UMR 5293, 33000 Bordeaux, France
| | | |
Collapse
|
17
|
Campbell RR, Lobo MK. Pallidal circuits drive addiction behavior. Trends Neurosci 2023; 46:S0166-2236(23)00228-X. [PMID: 39492310 DOI: 10.1016/j.tins.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/11/2023] [Accepted: 10/02/2023] [Indexed: 11/05/2024]
Abstract
Understanding the neural mechanisms that control addiction processes, including drug-seeking and relapse, is key to finding new targets for substance use disorder (SUD) pharmacotherapies and circuit-based therapies. Addictive drugs alter activity in distinct neural circuits that can lead to SUD symptoms, including compulsive drug craving and taking. This includes the pallidum, a region in the basal ganglia that acts as an integrator of associative, sensorimotor, and limbic information to shape motor responses, promote reward-learning, and regulate habit formation. Here, we review key findings that demonstrate the sub-regional and circuit-specific functions of the pallidum that drive addiction-related behaviors in rodents. We also highlight newly discovered mechanisms within distinct cell types and circuits of the pallidum that drive drug-seeking. Overall, this review serves to emphasize the importance of the pallidum in addiction processes and underscore the need for studying circuit-specific mechanisms in SUD research.
Collapse
Affiliation(s)
- Rianne R Campbell
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mary Kay Lobo
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
18
|
Luo YJ, Ge J, Chen ZK, Liu ZL, Lazarus M, Qu WM, Huang ZL, Li YD. Ventral pallidal glutamatergic neurons regulate wakefulness and emotion through separated projections. iScience 2023; 26:107385. [PMID: 37609631 PMCID: PMC10440712 DOI: 10.1016/j.isci.2023.107385] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 06/30/2023] [Accepted: 07/11/2023] [Indexed: 08/24/2023] Open
Abstract
Insomnia is often comorbid with depression, but the underlying neuronal circuit mechanism remains elusive. Recently, we reported that GABAergic ventral pallidum (VP) neurons control wakefulness associated with motivation. However, whether and how other subtypes of VP neurons regulate arousal and emotion are largely unknown. Here, we report glutamatergic VP (VPVglut2) neurons control wakefulness and depressive-like behaviors. Physiologically, the calcium activity of VPVglut2 neurons was increased during both NREM sleep-to-wake transitions and depressive/anxiety-like behaviors in mice. Functionally, activation of VPVglut2 neurons was sufficient to increase wakefulness and induce anxiety/depressive-like behaviors, whereas inhibition attenuated both. Dissection of the circuit revealed that separated projections of VPVglut2 neurons to the lateral hypothalamus and lateral habenula promote arousal and depressive-like behaviors, respectively. Our results demonstrate a subtype of VP neurons is responsible for wakefulness and emotion through separated projections, and may provide new lines for the intervention of insomnia and depression in patients.
Collapse
Affiliation(s)
- Yan-Jia Luo
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai 200032, China
- Department of Anesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jing Ge
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Ze-Ka Chen
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Zi-Long Liu
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine (WPI-IIIS) and Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Wei-Min Qu
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai 200032, China
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ya-Dong Li
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai 200032, China
- Songjiang Research Institute, Shanghai Songjiang District Central Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201699, China
| |
Collapse
|
19
|
Macpherson T, Dixon CI, Robertson J, Sindarto MM, Janak PH, Belelli D, Lambert JJ, Stephens DN, King SL. α4-Containing GABA A Receptors on DRD2 Neurons of the Nucleus Accumbens Mediate Instrumental Responding for Conditioned Reinforcers and Its Potentiation by Cocaine. eNeuro 2023; 10:ENEURO.0236-23.2023. [PMID: 37553242 PMCID: PMC10470850 DOI: 10.1523/eneuro.0236-23.2023] [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: 07/05/2023] [Accepted: 07/11/2023] [Indexed: 08/10/2023] Open
Abstract
Extrasynaptic GABAA receptors (GABAARs) composed of α4, β, and δ subunits mediate GABAergic tonic inhibition and are potential molecular targets in the modulation of behavioral responses to natural and drug rewards. These GABAARs are highly expressed within the nucleus accumbens (NAc), where they influence the excitability of the medium spiny neurons. Here, we explore their role in modulating behavioral responses to food-conditioned cues and the behavior-potentiating effects of cocaine. α4-Subunit constitutive knock-out mice (α4-/-) showed higher rates of instrumental responding for reward-paired stimuli in a test of conditioned reinforcement (CRf). A similar effect was seen following viral knockdown of GABAAR α4 subunits within the NAc. Local infusion of the α4βδ-GABAAR-preferring agonist THIP (4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridin-3-ol; Gaboxadol) into the NAc had no effect on responding when given alone but reduced cocaine potentiation of responding for conditioned reinforcers in wild-type, but not α4-/- mice. Finally, specific deletion of α4-subunits from dopamine D2, but not D1, receptor-expressing neurons (DRD2 and DRD1 neurons), mimicked the phenotype of the constitutive knockout, potentiating CRf responding, and blocking intra-accumbal THIP attenuation of cocaine-potentiated CRf responding. These data demonstrate that α4-GABAAR-mediated inhibition of DRD2 neurons reduces instrumental responding for a conditioned reinforcer and its potentiation by cocaine and emphasize the importance of GABAergic signaling within the NAc in mediating the effects of cocaine.
Collapse
Affiliation(s)
- Tom Macpherson
- Sussex Neuroscience, School of Psychology, University of Sussex, Brighton BN1 9QG, United Kingdom
- Laboratory for Advanced Brain Functions, Institute for Protein Research, Osaka University, Suita 565-0871, Japan
| | - Claire I. Dixon
- Sussex Neuroscience, School of Psychology, University of Sussex, Brighton BN1 9QG, United Kingdom
| | - Jonathan Robertson
- Sussex Neuroscience, School of Psychology, University of Sussex, Brighton BN1 9QG, United Kingdom
| | - Marsha M. Sindarto
- Sussex Neuroscience, School of Psychology, University of Sussex, Brighton BN1 9QG, United Kingdom
| | - Patricia H. Janak
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, Maryland 21218
| | - Delia Belelli
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital & Medical School, University of Dundee, Dundee DD1 9SY, United Kingdom
| | - Jeremy J. Lambert
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital & Medical School, University of Dundee, Dundee DD1 9SY, United Kingdom
| | - David N. Stephens
- Sussex Neuroscience, School of Psychology, University of Sussex, Brighton BN1 9QG, United Kingdom
| | - Sarah L. King
- Sussex Neuroscience, School of Psychology, University of Sussex, Brighton BN1 9QG, United Kingdom
| |
Collapse
|
20
|
Faget L, Oriol L, Lee WC, Sargent C, Ramanathan D, Hnasko TS. Ventral pallidum GABA and glutamate neurons drive approach and avoidance through distinct modulation of VTA cell types. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.12.548750. [PMID: 37502884 PMCID: PMC10369949 DOI: 10.1101/2023.07.12.548750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The ventral pallidum (VP) contains GABA and glutamate (Glut) neurons projecting to ventral tegmental area (VTA) whose stimulation drives approach and avoidance, respectively. Yet little is known about the cell-type-specific mechanisms by which VP projections to VTA drive behavior. Here, we found that both VP GABA and Glut neurons were activated during approach to reward or delivery of an aversive stimulus. Stimulation of VP GABA neurons inhibited VTA GABA, but activated dopamine (DA) and glutamate neurons. Remarkably, this cell-type-specific recruitment was behavior-contingent such that VTA recruitment was inhibited when evoked by the subject's own action. Conversely, VP Glut neurons activated VTA GABA, as well as DA and Glut neurons, despite driving aversion. However, VP Glut neurons evoked DA in reward-associated ventromedial nucleus accumbens (NAc), but reduced DA in aversion-associated dorsomedial NAc. These findings show how heterogeneous VP cell types can engage VTA cell types to shape approach and avoidance behaviors.
Collapse
Affiliation(s)
- Lauren Faget
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA
| | - Lucie Oriol
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Wen-Chun Lee
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Cody Sargent
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Dhakshin Ramanathan
- Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093, USA
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA
| | - Thomas S. Hnasko
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA
| |
Collapse
|
21
|
Neuhofer D, Kalivas P. Differential Modulation of GABAergic and Glutamatergic Neurons in the Ventral Pallidum by GABA and Neuropeptides. eNeuro 2023; 10:ENEURO.0404-22.2023. [PMID: 37414552 PMCID: PMC10348443 DOI: 10.1523/eneuro.0404-22.2023] [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: 09/26/2022] [Revised: 02/23/2023] [Accepted: 03/13/2023] [Indexed: 07/08/2023] Open
Abstract
The ventral pallidum (VP) is an integral locus in the reward circuitry and a major target of GABAergic innervation of both D1-medium spiny neurons (MSNs) and D2-MSNs from the nucleus accumbens. The VP contains populations of GABAergic [VPGABA, GAD2(+), or VGluT(-)] and glutamatergic [VPGlutamate, GAD2(-), or VGluT(+)] cells that facilitate positive reinforcement and behavioral avoidance, respectively. MSN efferents to the VP exert opponent control over behavioral reinforcement with activation of D1-MSN afferents promoting and D2-MSN afferents inhibiting reward seeking. How this afferent-specific and cell type-specific control of reward seeking is integrated remains largely unknown. In addition to GABA, D1-MSNs corelease substance P to stimulate neurokinin 1 receptors (NK1Rs) and D2-MSNs corelease enkephalin to activate μ-opioid receptors (MORs) and δ-opioid receptors. These neuropeptides act in the VP to alter appetitive behavior and reward seeking. Using a combination of optogenetics and patch-clamp electrophysiology in mice, we found that GAD2(-) cells receive weaker GABA input from D1-MSN, but GAD2(+) cells receive comparable GABAergic input from both afferent types. Pharmacological activation of MORs induced an equally strong presynaptic inhibition of GABA and glutamate transmission on both cell types. Interestingly, MOR activation hyperpolarized VPGABA but not VGluT(+). NK1R activation inhibited glutamatergic transmission only on VGluT(+) cells. Our results indicate that the afferent-specific release of GABA and neuropeptides from D1-MSNs and D2-MSNs can differentially influence VP neuronal subtypes.
Collapse
Affiliation(s)
- Daniela Neuhofer
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Peter Kalivas
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina 29425
| |
Collapse
|
22
|
Soares-Cunha C, Heinsbroek JA. Ventral pallidal regulation of motivated behaviors and reinforcement. Front Neural Circuits 2023; 17:1086053. [PMID: 36817646 PMCID: PMC9932340 DOI: 10.3389/fncir.2023.1086053] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/06/2023] [Indexed: 02/05/2023] Open
Abstract
The interconnected nuclei of the ventral basal ganglia have long been identified as key regulators of motivated behavior, and dysfunction of this circuit is strongly implicated in mood and substance use disorders. The ventral pallidum (VP) is a central node of the ventral basal ganglia, and recent studies have revealed complex VP cellular heterogeneity and cell- and circuit-specific regulation of reward, aversion, motivation, and drug-seeking behaviors. Although the VP is canonically considered a relay and output structure for this circuit, emerging data indicate that the VP is a central hub in an extensive network for reward processing and the regulation of motivation that extends beyond classically defined basal ganglia borders. VP neurons respond temporally faster and show more advanced reward coding and prediction error processing than neurons in the upstream nucleus accumbens, and regulate the activity of the ventral mesencephalon dopamine system. This review will summarize recent findings in the literature and provide an update on the complex cellular heterogeneity and cell- and circuit-specific regulation of motivated behaviors and reinforcement by the VP with a specific focus on mood and substance use disorders. In addition, we will discuss mechanisms by which stress and drug exposure alter the functioning of the VP and produce susceptibility to neuropsychiatric disorders. Lastly, we will outline unanswered questions and identify future directions for studies necessary to further clarify the central role of VP neurons in the regulation of motivated behaviors. Significance: Research in the last decade has revealed a complex cell- and circuit-specific role for the VP in reward processing and the regulation of motivated behaviors. Novel insights obtained using cell- and circuit-specific interrogation strategies have led to a major shift in our understanding of this region. Here, we provide a comprehensive review of the VP in which we integrate novel findings with the existing literature and highlight the emerging role of the VP as a linchpin of the neural systems that regulate motivation, reward, and aversion. In addition, we discuss the dysfunction of the VP in animal models of neuropsychiatric disorders.
Collapse
Affiliation(s)
- 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
| | - Jasper A. Heinsbroek
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
23
|
Swinford-Jackson SE, Huffman PJ, Knouse MC, Thomas AS, Rich MT, Mankame S, Worobey SJ, Sarmiento M, Coleman A, Pierce RC. High frequency DBS-like optogenetic stimulation of nucleus accumbens dopamine D2 receptor-containing neurons attenuates cocaine reinstatement in male rats. Neuropsychopharmacology 2023; 48:459-467. [PMID: 36446928 PMCID: PMC9852282 DOI: 10.1038/s41386-022-01495-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/27/2022] [Accepted: 10/29/2022] [Indexed: 11/30/2022]
Abstract
Previous work indicated that deep brain stimulation (DBS) of the nucleus accumbens shell in male rats attenuated reinstatement of cocaine seeking, an animal model of craving. However, the potential differential impact of DBS on specific populations of neurons to drive the suppression of cocaine seeking is unknown. Medium spiny neurons in the nucleus accumbens are differentiated by expression of dopamine D1 receptors (D1DRs) or D2DRs, activation of which promotes or inhibits cocaine-related behaviors, respectively. The advent of transgenic rat lines expressing Cre recombinase selectively in D1DR-containing or D2DR-containing neurons, when coupled with Cre-dependent virally mediated gene transfer of channelrhodopsin (ChR2), enabled mimicry of DBS in a selective subpopulation of neurons during complex tasks. We tested the hypothesis that high frequency DBS-like optogenetic stimulation of D1DR-containing neurons in the accumbens shell would potentiate, whereas stimulation of D2DR-containing neurons in the accumbens shell would attenuate, cocaine-primed reinstatement of cocaine seeking. Results indicated that high frequency, DBS-like optogenetic stimulation of D2DR-containing neurons attenuated reinstatement of cocaine seeking in male rats, whereas DBS-like stimulation of D1DR-containing neurons did not alter cocaine-primed reinstatement. Surprisingly, DBS-like optogenetic stimulation did not alter reinstatement of cocaine seeking in female rats. In rats which only expressed eYFP, intra-accumbens optogenetic stimulation did not alter cocaine reinstatement, indicating that the effect of DBS-like stimulation to attenuate cocaine reinstatement is mediated specifically by ChR2 rather than by prolonged light delivery. These results suggest that DBS of the accumbens may attenuate cocaine-primed reinstatement in male rats through the selective manipulation of D2DR-containing neurons.
Collapse
Affiliation(s)
- Sarah E Swinford-Jackson
- Brain Health Institute and Department of Psychiatry, Rutgers University, Piscataway, NJ, 08854, USA.
- Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Brain Health Institute and Department of Psychiatry, Rutgers University, Piscataway, NJ, 08854, USA.
| | - Phillip J Huffman
- Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Melissa C Knouse
- Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA
| | - Arthur S Thomas
- Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Matthew T Rich
- Brain Health Institute and Department of Psychiatry, Rutgers University, Piscataway, NJ, 08854, USA
- Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Brain Health Institute and Department of Psychiatry, Rutgers University, Piscataway, NJ, 08854, USA
| | - Sharvari Mankame
- Brain Health Institute and Department of Psychiatry, Rutgers University, Piscataway, NJ, 08854, USA
| | - Samantha J Worobey
- Brain Health Institute and Department of Psychiatry, Rutgers University, Piscataway, NJ, 08854, USA
| | - Mateo Sarmiento
- Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ayanna Coleman
- Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - R Christopher Pierce
- Brain Health Institute and Department of Psychiatry, Rutgers University, Piscataway, NJ, 08854, USA
- Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Brain Health Institute and Department of Psychiatry, Rutgers University, Piscataway, NJ, 08854, USA
| |
Collapse
|
24
|
Swinford-Jackson SE, Rich MT, Huffman PJ, Knouse MC, Thomas AS, Mankame S, Worobey SJ, Pierce RC. Low frequency optogenetic deep brain stimulation of nucleus accumbens dopamine D1 or D2 receptor-containing neurons attenuates cocaine seeking selectively in male rats in part by reversing synaptic plasticity deficits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.23.524956. [PMID: 36747662 PMCID: PMC9900748 DOI: 10.1101/2023.01.23.524956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Background Clinically, deep brain stimulation (DBS) utilizes relatively high frequencies (>100 Hz). In preclinical models, 160 Hz stimulation of the nucleus accumbens in rodents prevents relapse of drug seeking. However, the ability of varied frequencies of accumbens DBS to attenuate drug seeking, and the neuronal subtype specificity of this effect, is unclear. Methods The present study examined the effect of DBS in the nucleus accumbens on neuronal plasticity and cocaine-primed reinstatement of cocaine seeking behavior in rats. Results Electrical DBS of the accumbens shell attenuated cocaine primed reinstatement across a range of frequencies in male rats, including as low as 12 Hz. The majority of nucleus accumbens neurons are medium spiny neurons (MSNs), which can be differentiated in terms of projections and effects on cocaine-related behaviors by expression of dopamine D1 receptors (D1DRs) or D2DRs. In slice electrophysiology experiments, 12 Hz electrical stimulation evoked long term potentiation (LTP) in eYFP labeled D1DR-MSNs and D2DR-MSNs from cocaine naive male and female rats. However, in rats that self-administered cocaine and underwent extinction training, a paradigm identical to our reinstatement experiments, electrical DBS only elicited LTP in D2DR-MSNs from male rats; this effect was replicated by optical stimulation in rats expressing Cre-dependent ChR2 in D2DR-MSNs. Low-frequency optogenetic-DBS in D1DR-containing or D2DR-containing neurons attenuated cocaine-primed reinstatement of cocaine seeking in male but not female rats. Conclusions These results suggest that administering DBS in the nucleus accumbens shell at lower frequencies effectively, but sex-specifically, suppresses cocaine craving, perhaps in part by reversing synaptic plasticity deficits selectively in D2DR-MSNs.
Collapse
Affiliation(s)
- Sarah E. Swinford-Jackson
- Brain Health Institute and Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 USA
- Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Matthew T. Rich
- Brain Health Institute and Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 USA
- Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Phillip J. Huffman
- Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Melissa C. Knouse
- Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Arthur S. Thomas
- Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Sharvari Mankame
- Brain Health Institute and Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 USA
| | - Samantha J. Worobey
- Brain Health Institute and Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 USA
| | - R. Christopher Pierce
- Brain Health Institute and Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854 USA
- Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| |
Collapse
|
25
|
Newman AH, Xi ZX, Heidbreder C. Current Perspectives on Selective Dopamine D 3 Receptor Antagonists/Partial Agonists as Pharmacotherapeutics for Opioid and Psychostimulant Use Disorders. Curr Top Behav Neurosci 2023; 60:157-201. [PMID: 35543868 PMCID: PMC9652482 DOI: 10.1007/7854_2022_347] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Over three decades of evidence indicate that dopamine (DA) D3 receptors (D3R) are involved in the control of drug-seeking behavior and may play an important role in the pathophysiology of substance use disorders (SUD). The expectation that a selective D3R antagonist/partial agonist would be efficacious for the treatment of SUD is based on the following key observations. First, D3R are distributed in strategic areas belonging to the mesolimbic DA system such as the ventral striatum, midbrain, and ventral pallidum, which have been associated with behaviors controlled by the presentation of drug-associated cues. Second, repeated exposure to drugs of abuse produces neuroadaptations in the D3R system. Third, the synthesis and characterization of highly potent and selective D3R antagonists/partial agonists have further strengthened the role of the D3R in SUD. Based on extensive preclinical and preliminary clinical evidence, the D3R shows promise as a target for the development of pharmacotherapies for SUD as reflected by their potential to (1) regulate the motivation to self-administer drugs and (2) disrupt the responsiveness to drug-associated stimuli that play a key role in reinstatement of drug-seeking behavior triggered by re-exposure to the drug itself, drug-associated environmental cues, or stress. The availability of PET ligands to assess clinically relevant receptor occupancy by selective D3R antagonists/partial agonists, the definition of reliable dosing, and the prospect of using human laboratory models may further guide the design of clinical proof of concept studies. Pivotal clinical trials for more rapid progression of this target toward regulatory approval are urgently required. Finally, the discovery that highly selective D3R antagonists, such as R-VK4-116 and R-VK4-40, do not adversely affect peripheral biometrics or cardiovascular effects alone or in the presence of oxycodone or cocaine suggests that this class of drugs has great potential in safely treating psychostimulant and/or opioid use disorders.
Collapse
Affiliation(s)
- Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, Baltimore, MD, USA.
| | - Zheng-Xiong Xi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, Baltimore, MD, USA
| | | |
Collapse
|
26
|
Saga Y, Galineau L, Tremblay L. Impulsive and compulsive behaviors can be induced by opposite GABAergic dysfunctions inside the primate ventral pallidum. Front Syst Neurosci 2022; 16:1009626. [PMID: 36567755 PMCID: PMC9774472 DOI: 10.3389/fnsys.2022.1009626] [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: 08/02/2022] [Accepted: 11/17/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction: The ventral pallidum (VP) is central in the limbic Basal Ganglia circuit, controlling both appetitive (approach) and aversive (avoidance) motivated behaviors. Nevertheless, VP involvement in pathological aspects remains unclear, especially in the behavioral expression of different motivational dysfunctions. This study aimed to investigate how the VP contributes to the expression of abnormal behaviors via opposite GABAergic dysfunctions. Methods: Opposite GABAergic dysfunctions were induced by injecting muscimol (a GABAA agonist) and bicuculline (a GABAA antagonist) into monkeys. We determined the effects of both substances on self-initiated behaviors in lab-chair and in free-moving home-cage contexts in six monkeys, and in two animals performing an approach-avoidance task in appetitive and aversive contexts. Results: While the self-initiated behaviors induced by bicuculline injections in VP were characterized by compulsive behaviors such as repetitive grooming and self-biting, muscimol injections induced impulsive behaviors including limb movements in a lab-chair context and exploration behaviors in a free-moving context. More specific behavioral effects were observed in the approach-avoidance task. The muscimol injections induced premature responses and erroneous screen touches, which characterize impulsive and attention disorders, while the bicuculline injections into the VP increased passive avoidance (non-initiated action) and task-escape in an aversive context, suggesting an anxiety disorder. Conclusions: These results show that activating or blocking GABAergic transmission in the VP impairs motivated behaviors. Furthermore, the behavioral expressions produced by these opposite disturbances show that the VP could be involved in anxiety-driven compulsive disorders, such as OCD, as well as in impulsive disorders motivated by attention deficits or reward-seeking, as seen in ADHD or impulse control disorders.
Collapse
Affiliation(s)
- Yosuke Saga
- Institut des Sciences Cognitives Marc Jeannerod, UMR-5229 CNRS, Bron Cedex, France,*Correspondence: Yosuke Saga Léon Tremblay
| | - Laurent Galineau
- UMR INSERM U1253, Université François Rabelais de Tours, Tours, France
| | - Léon Tremblay
- Institut des Sciences Cognitives Marc Jeannerod, UMR-5229 CNRS, Bron Cedex, France,Université Claude-Bernard Lyon1, Villeurbanne, France,*Correspondence: Yosuke Saga Léon Tremblay
| |
Collapse
|
27
|
Dusa D, Ollmann T, Kállai V, Lénárd L, Kertes E, Berta B, Szabó Á, László K, Gálosi R, Zagoracz O, Karádi Z, Péczely L. The antipsychotic drug sulpiride in the ventral pallidum paradoxically impairs learning and induces place preference. Sci Rep 2022; 12:19247. [PMID: 36357539 PMCID: PMC9649625 DOI: 10.1038/s41598-022-23450-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 10/31/2022] [Indexed: 11/12/2022] Open
Abstract
Sulpiride, as a D2-like dopamine (DA) receptor (D2R) antagonist, is an important antipsychotic drug in the treatment of schizophrenia. Recently, we have shown that the activation of D2Rs in the ventral pallidum (VP) modulates the activity of the ventral tegmental area (VTA) DAergic neurons. According to our hypothesis, intra-VP sulpiride can influence the motivational and learning processes, pervasively modifying the behavior of examined animals. In the present study, sulpiride was microinjected into the VP of male Wistar rats in three different doses. Morris water maze (MWM) test was applied to investigate the effects of sulpiride on spatial learning, while conditioned place preference (CPP) test was used to examine the potential rewarding effect of the drug. In order to show, whether the animals can associate the rewarding effect with an area which can be recognized only on its spatial location, we introduced a modified version of the CPP paradigm, the spatial CPP test. Our results show that the intra-VP sulpiride dose-dependently impairs learning processes. However, the largest dose of sulpiride induces place preference. Results of the spatial CPP paradigm demonstrate that the animals cannot associate the rewarding effect of the drug with the conditioning area based on its spatial location. In the CPP paradigm, locomotor activity decrease could be observed in the sulpiride-treated rats, likely because of a faster habituation with the conditioning environment. In summary, we can conclude that intra-VP sulpiride has a dual effect: it diminishes the hippocampus-dependent spatial learning processes, in addition, it has a dose-dependent rewarding effect.
Collapse
Affiliation(s)
- Daniella Dusa
- Learning in Biological and Artificial Systems Research Group, Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Institute of Physiology, Medical School, University of Pécs, Szigeti Str. 12, P.O. Box: 99, Pécs, 7602, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - Tamás Ollmann
- Learning in Biological and Artificial Systems Research Group, Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Neuropeptides, Cognition, Animal Models of Neuropsychiatric Disorders Research Group, Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Institute of Physiology, Medical School, University of Pécs, Szigeti Str. 12, P.O. Box: 99, Pécs, 7602, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - Veronika Kállai
- Learning in Biological and Artificial Systems Research Group, Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Neuropeptides, Cognition, Animal Models of Neuropsychiatric Disorders Research Group, Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Institute of Physiology, Medical School, University of Pécs, Szigeti Str. 12, P.O. Box: 99, Pécs, 7602, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - László Lénárd
- Institute of Physiology, Medical School, University of Pécs, Szigeti Str. 12, P.O. Box: 99, Pécs, 7602, Hungary
- Molecular Neuroendocrinology and Neurophysiology Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - Erika Kertes
- Learning in Biological and Artificial Systems Research Group, Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Neuropeptides, Cognition, Animal Models of Neuropsychiatric Disorders Research Group, Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Institute of Physiology, Medical School, University of Pécs, Szigeti Str. 12, P.O. Box: 99, Pécs, 7602, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - Beáta Berta
- Learning in Biological and Artificial Systems Research Group, Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Neuropeptides, Cognition, Animal Models of Neuropsychiatric Disorders Research Group, Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Institute of Physiology, Medical School, University of Pécs, Szigeti Str. 12, P.O. Box: 99, Pécs, 7602, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - Ádám Szabó
- Learning in Biological and Artificial Systems Research Group, Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Institute of Physiology, Medical School, University of Pécs, Szigeti Str. 12, P.O. Box: 99, Pécs, 7602, Hungary
| | - Kristóf László
- Neuropeptides, Cognition, Animal Models of Neuropsychiatric Disorders Research Group, Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Institute of Physiology, Medical School, University of Pécs, Szigeti Str. 12, P.O. Box: 99, Pécs, 7602, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - Rita Gálosi
- Institute of Physiology, Medical School, University of Pécs, Szigeti Str. 12, P.O. Box: 99, Pécs, 7602, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - Olga Zagoracz
- Neuropeptides, Cognition, Animal Models of Neuropsychiatric Disorders Research Group, Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Institute of Physiology, Medical School, University of Pécs, Szigeti Str. 12, P.O. Box: 99, Pécs, 7602, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - Zoltán Karádi
- Institute of Physiology, Medical School, University of Pécs, Szigeti Str. 12, P.O. Box: 99, Pécs, 7602, Hungary
- Molecular Neuroendocrinology and Neurophysiology Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - László Péczely
- Learning in Biological and Artificial Systems Research Group, Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary.
- Institute of Physiology, Medical School, University of Pécs, Szigeti Str. 12, P.O. Box: 99, Pécs, 7602, Hungary.
- Centre for Neuroscience, University of Pécs, Pécs, Hungary.
| |
Collapse
|
28
|
Linders LE, Supiot LF, Du W, D’Angelo R, Adan RAH, Riga D, Meye FJ. Studying Synaptic Connectivity and Strength with Optogenetics and Patch-Clamp Electrophysiology. Int J Mol Sci 2022; 23:ijms231911612. [PMID: 36232917 PMCID: PMC9570045 DOI: 10.3390/ijms231911612] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 02/07/2023] Open
Abstract
Over the last two decades the combination of brain slice patch clamp electrophysiology with optogenetic stimulation has proven to be a powerful approach to analyze the architecture of neural circuits and (experience-dependent) synaptic plasticity in such networks. Using this combination of methods, originally termed channelrhodopsin-assisted circuit mapping (CRACM), a multitude of measures of synaptic functioning can be taken. The current review discusses their rationale, current applications in the field, and their associated caveats. Specifically, the review addresses: (1) How to assess the presence of synaptic connections, both in terms of ionotropic versus metabotropic receptor signaling, and in terms of mono- versus polysynaptic connectivity. (2) How to acquire and interpret measures for synaptic strength and function, like AMPAR/NMDAR, AMPAR rectification, paired-pulse ratio (PPR), coefficient of variance and input-specific quantal sizes. We also address how synaptic modulation by G protein-coupled receptors can be studied with pharmacological approaches and advanced technology. (3) Finally, we elaborate on advances on the use of dual color optogenetics in concurrent investigation of multiple synaptic pathways. Overall, with this review we seek to provide practical insights into the methods used to study neural circuits and synapses, by combining optogenetics and patch-clamp electrophysiology.
Collapse
|
29
|
Dai KZ, Choi IB, Levitt R, Blegen MB, Kaplan AR, Matsui A, Shin JH, Bocarsly ME, Simpson EH, Kellendonk C, Alvarez VA, Dobbs LK. Dopamine D2 receptors bidirectionally regulate striatal enkephalin expression: Implications for cocaine reward. Cell Rep 2022; 40:111440. [PMID: 36170833 PMCID: PMC9620395 DOI: 10.1016/j.celrep.2022.111440] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 08/04/2022] [Accepted: 09/09/2022] [Indexed: 11/24/2022] Open
Abstract
Low dopamine D2 receptor (D2R) availability in the striatum can predispose for cocaine abuse; though how low striatal D2Rs facilitate cocaine reward is unclear. Overexpression of D2Rs in striatal neurons or activation of D2Rs by acute cocaine suppresses striatal Penk mRNA. Conversely, low D2Rs in D2-striatal neurons increases striatal Penk mRNA and enkephalin peptide tone, an endogenous mu-opioid agonist. In brain slices, met-enkephalin and inhibition of enkephalin catabolism suppresses intra-striatal GABA transmission. Pairing cocaine with intra-accumbens met-enkephalin during place conditioning facilitates acquisition of preference, while mu-opioid receptor antagonist blocks preference in wild-type mice. We propose that heightened striatal enkephalin potentiates cocaine reward by suppressing intra-striatal GABA to enhance striatal output. Surprisingly, a mu-opioid receptor antagonist does not block cocaine preference in mice with low striatal D2Rs, implicating other opioid receptors. The bidirectional regulation of enkephalin by D2R activity and cocaine offers insights into mechanisms underlying the vulnerability for cocaine abuse. Low striatal D2 receptor levels are associated with cocaine abuse. Dai et al. bidirectionally alter striatal D2 receptor levels to probe the downstream mechanisms underlying this abuse liability. They provide evidence that enhanced enkephalin tone resulting from low D2 receptors is associated with suppressed intra-striatal GABA and potentiated cocaine reward.
Collapse
Affiliation(s)
- Kathy Z Dai
- Laboratory on Neurobiology of Compulsive Behaviors, NIAAA, IRP, NIH, Bethesda, MD, USA
| | - In Bae Choi
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Ryan Levitt
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Mariah B Blegen
- Laboratory on Neurobiology of Compulsive Behaviors, NIAAA, IRP, NIH, Bethesda, MD, USA
| | - Alanna R Kaplan
- Laboratory on Neurobiology of Compulsive Behaviors, NIAAA, IRP, NIH, Bethesda, MD, USA
| | - Aya Matsui
- Laboratory on Neurobiology of Compulsive Behaviors, NIAAA, IRP, NIH, Bethesda, MD, USA
| | - J Hoon Shin
- Laboratory on Neurobiology of Compulsive Behaviors, NIAAA, IRP, NIH, Bethesda, MD, USA
| | - Miriam E Bocarsly
- Laboratory on Neurobiology of Compulsive Behaviors, NIAAA, IRP, NIH, Bethesda, MD, USA; Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Rutgers Brain Health Institute, Newark, NJ, USA
| | - Eleanor H Simpson
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA; Division of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, USA
| | - Christoph Kellendonk
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA; Department of Molecular Pharmacology and Therapeutics, Columbia University Medical Center, New York, NY, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Veronica A Alvarez
- Laboratory on Neurobiology of Compulsive Behaviors, NIAAA, IRP, NIH, Bethesda, MD, USA; Center on Compulsive Behaviors, IRP, NIH, Bethesda, MD, USA
| | - Lauren K Dobbs
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX, USA; Department of Neuroscience, Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
30
|
Farrell MR, Ye Q, Xie Y, Esteban JSD, Mahler SV. Ventral pallidum GABA neurons bidirectionally control opioid relapse across rat behavioral models. ADDICTION NEUROSCIENCE 2022; 3:100026. [PMID: 36156918 PMCID: PMC9494709 DOI: 10.1016/j.addicn.2022.100026] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Opioid addiction is a chronic, relapsing disorder. Whether addicted individuals are forced to abstain or they decide themselves to quit using drugs, relapse rates are high-especially upon encountering contexts and stimuli associated with prior opioid use. Rodents similarly show context- and cue-induced reinstatement of drug seeking following abstinence, and intriguingly, the neural circuits underlying these relapse-like behaviors differ when abstinence is involuntarily imposed, responding is extinguished, or animals decide themselves to cease taking drug. Here, we employ two complementary rat behavioral models of relapse-like behavior for the highly reinforcing opioid drug remifentanil, and asked whether GABAergic neurons in the ventral pallidum (VPGABA) control opioid seeking under these behavioral conditions. Specifically, we asked how chemogenetically stimulating VPGABA neurons with clozapine-N-oxide (CNO) influences the ability of contextual or discrete remifentanil-paired cues to reinstate drug seeking following either voluntary abstinence (punishment-induced; GroupPunish), or extinction training (GroupExt). In GroupPunish rats, we also chemogenetically inhibited VPGABA neurons, and examined spontaneous VP activity (Fos) during cued reinstatement. In both GroupPunish and GroupExt rats, stimulating Gq-signaling in VPGABA neurons augmented remifentanil reinstatement in a cue- and context-dependent manner. Conversely, engaging inhibitory Gi-signaling in VPGABA neurons in GroupPunish suppressed cue-induced reinstatement, and cue-triggered seeking was correlated with Fos expression in rostral, but not caudal VP. Neither stimulating nor inhibiting VPGABA neurons influenced unpunished remifentanil self-administration. We conclude that VPGABA neurons bidirectionally control opioid seeking regardless of the specific relapse model employed, highlighting their fundamental role in opioid relapse-like behavior across behavioral models, and potentially across species.
Collapse
Affiliation(s)
- Mitchell R. Farrell
- University of California, Irvine Department of Neurobiology and Behavior, 1203 McGaugh Hall Irvine, CA, 92697, USA
| | - Qiying Ye
- University of California, Irvine Department of Neurobiology and Behavior, 1203 McGaugh Hall Irvine, CA, 92697, USA
| | - Yiyan Xie
- University of California, Irvine Department of Neurobiology and Behavior, 1203 McGaugh Hall Irvine, CA, 92697, USA
| | - Jeanine Sandra D. Esteban
- University of California, Irvine Department of Neurobiology and Behavior, 1203 McGaugh Hall Irvine, CA, 92697, USA
| | - Stephen V. Mahler
- University of California, Irvine Department of Neurobiology and Behavior, 1203 McGaugh Hall Irvine, CA, 92697, USA
| |
Collapse
|
31
|
Zhao F, Cheng Z, Piao J, Cui R, Li B. Dopamine Receptors: Is It Possible to Become a Therapeutic Target for Depression? Front Pharmacol 2022; 13:947785. [PMID: 36059987 PMCID: PMC9428607 DOI: 10.3389/fphar.2022.947785] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Dopamine and its receptors are currently recognized targets for the treatment of several neuropsychiatric disorders, including Parkinson’s disease, schizophrenia, some drug use addictions, as well as depression. Dopamine receptors are widely distributed in various regions of the brain, but their role and exact contribution to neuropsychiatric diseases has not yet been thoroughly studied. Based on the types of dopamine receptors and their distribution in different brain regions, this paper reviews the current research status of the molecular, cellular and circuit mechanisms of dopamine and its receptors involved in depression. Multiple lines of investigation of these mechanisms provide a new future direction for understanding the etiology and treatment of depression and potential new targets for antidepressant treatments.
Collapse
Affiliation(s)
- Fangyi Zhao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
- Engineering Laboratory for Screening of Antidepressant Drugs, Jilin Province Development and Reform Commission, Changchun, China
| | - Ziqian Cheng
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
- Engineering Laboratory for Screening of Antidepressant Drugs, Jilin Province Development and Reform Commission, Changchun, China
| | - Jingjing Piao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
- Engineering Laboratory for Screening of Antidepressant Drugs, Jilin Province Development and Reform Commission, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
- Engineering Laboratory for Screening of Antidepressant Drugs, Jilin Province Development and Reform Commission, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
- Engineering Laboratory for Screening of Antidepressant Drugs, Jilin Province Development and Reform Commission, Changchun, China
- *Correspondence: Bingjin Li,
| |
Collapse
|
32
|
Inbar K, Levi LA, Kupchik YM. Cocaine induces input and cell-type-specific synaptic plasticity in ventral pallidum-projecting nucleus accumbens medium spiny neurons. Neuropsychopharmacology 2022; 47:1461-1472. [PMID: 35121830 PMCID: PMC9205871 DOI: 10.1038/s41386-022-01285-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 01/23/2022] [Accepted: 01/24/2022] [Indexed: 11/09/2022]
Abstract
Cocaine use and abstinence induce long-term synaptic alterations in the excitatory input to nucleus accumbens (NAc) medium spiny neurons (MSNs). The NAc regulates reward-related behaviors through two parallel projections to the ventral pallidum (VP)-originating in D1 or D2-expressing MSNs (D1-MSNs→VP; D2-MSNs→VP). The activity of these projections depends on their excitatory synaptic inputs, but it is not known whether and how abstinence from cocaine affects the excitatory transmission to D1-MSNs→VP and D2-MSNs→VP. Here we examined different forms of cocaine-induced synaptic plasticity in the inputs from the basolateral amygdala (BLA) and medial prefrontal cortex (mPFC) to NAc D1-MSNs→VP and putative D2-MSNs→VP (pD2-MSNs→VP) in the core and shell subcompartments of the NAc. We used the whole-cell patch-clamp technique to record excitatory postsynaptic currents from D1-tdTomato mice injected with ChR2 in either the BLA or the mPFC and retrograde tracer (RetroBeads) in the VP. We found that cocaine conditioned place preference (CPP) followed by abstinence potentiated the excitatory input from the BLA and mPFC to both D1-MSNs→VP and pD2-MSNs→VP. Interestingly, while the strengthening of the inputs to D1-MSNs→VP was of postsynaptic origin and manifested as increased AMPA to NMDA ratio, in pD2-MSNs→VP plasticity was predominantly presynaptic and was detected as changes in the paired-pulse ratio and coefficient of variation. Lastly, some of the changes were sex-specific. Overall our data show that abstinence from cocaine changes the excitatory inputs to both D1-MSNs→VP and pD2-MSNs→VP but with different mechanisms. This may help understand how circuits converging into the VP change after cocaine exposure.
Collapse
Affiliation(s)
- Kineret Inbar
- grid.9619.70000 0004 1937 0538Department of Medical Neurobiology, Faculty of Medicine, The Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Jerusalem, 9112102 Israel
| | - Liran A. Levi
- grid.9619.70000 0004 1937 0538Department of Medical Neurobiology, Faculty of Medicine, The Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Jerusalem, 9112102 Israel
| | - Yonatan M. Kupchik
- grid.9619.70000 0004 1937 0538Department of Medical Neurobiology, Faculty of Medicine, The Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Jerusalem, 9112102 Israel
| |
Collapse
|
33
|
Nall RW, Chalhoub RM, Kalivas PW. Drug versus non-drug behaviors: A dual-reward model of sex differences and neurobiological mechanisms in rats. J Exp Anal Behav 2022; 117:457-471. [PMID: 35297047 PMCID: PMC10775707 DOI: 10.1002/jeab.752] [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: 10/15/2021] [Revised: 02/02/2022] [Accepted: 02/09/2022] [Indexed: 11/05/2022]
Abstract
Substance Use Disorders (SUDs) are an impactful problem characterized by chronic relapse and engagement in drug-related behaviors at the expense of non-drug behaviors. Brain regions implicated in drug and non-drug-related behaviors often overlap, complicating investigations of neurobiological mechanisms underlying SUDs. Here we presented a within-subject model for studying self-administration, reinforcer competition, extinction, and cued reinstatement of cocaine- and food-seeking in rats. Due to differences in cocaine- and food-reinforced behavior, we transformed data to proportions of baseline, revealing increased resistance to extinction and disproportionately greater cued reinstatement of cocaine seeking relative to food seeking. Consistent with previous reports, females showed greater preference for cocaine reinforcement than males, though these findings failed to reach statistical significance. To demonstrate the model's utility for investigating neurobiological mechanisms, we included proof-of-concept calcium imaging data demonstrating the utility of the behavioral model for detecting cellular activity patterns associated with cocaine- and food-seeking behaviors. Future studies utilizing this model should improve understanding of the development and expression of pathological behaviors characteristic of SUDs in humans, sex differences in these behaviors, and their neurobiological correlates. Thus, the model has utility for improving understanding of SUDs, leading to novel treatments to reduce the pathological behaviors associated with SUDs.
Collapse
Affiliation(s)
- Rusty W. Nall
- Medical University of South Carolina
- Jacksonville State University
| | | | | |
Collapse
|
34
|
Doucette WT, Smedley EB, Ruiz-Jaquez M, Khokhar JY, Smith KS. Chronic Chemogenetic Manipulation of Ventral Pallidum Targeted Neurons in Male Rats Fed an Obesogenic Diet. Brain Res 2022; 1784:147886. [DOI: 10.1016/j.brainres.2022.147886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/14/2022] [Accepted: 03/16/2022] [Indexed: 11/26/2022]
|
35
|
Juarez B, Zweifel LS. Disinhibitory feedback loops for reward and aversion. Cell Res 2022; 32:115-116. [PMID: 34949785 PMCID: PMC8807610 DOI: 10.1038/s41422-021-00601-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Affiliation(s)
- Barbara Juarez
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
- Department of Pharmacology, University of Washington, Seattle, WA, USA
- Center for the Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
| | - Larry S Zweifel
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA.
- Department of Pharmacology, University of Washington, Seattle, WA, USA.
- Center for the Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA.
| |
Collapse
|
36
|
A distinct D1-MSN subpopulation down-regulates dopamine to promote negative emotional state. Cell Res 2022; 32:139-156. [PMID: 34848869 PMCID: PMC8807621 DOI: 10.1038/s41422-021-00588-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022] Open
Abstract
Dopamine (DA) level in the nucleus accumbens (NAc) is critical for reward and aversion encoding. DA released from the ventral mesencephalon (VM) DAergic neurons increases the excitability of VM-projecting D1-dopamine receptor-expressing medium spiny neurons (D1-MSNs) in the NAc to enhance DA release and augment rewards. However, how such a DA positive feedback loop is regulated to maintain DA homeostasis and reward-aversion balance remains elusive. Here we report that the ventral pallidum (VP) projection of NAc D1-MSNs (D1NAc-VP) is inhibited by rewarding stimuli and activated by aversive stimuli. In contrast to the VM projection of D1-MSN (D1NAc-VM), activation of D1NAc-VP projection induces aversion, but not reward. D1NAc-VP MSNs are distinct from the D1NAc-VM MSNs, which exhibit conventional functions of D1-MSNs. Activation of D1NAc-VP projection stimulates VM GABAergic transmission, inhibits VM DAergic neurons, and reduces DA release into the NAc. Thus, D1NAc-VP and D1NAc-VM MSNs cooperatively control NAc dopamine balance and reward-aversion states.
Collapse
|
37
|
Peart DR, Andrade AK, Logan CN, Knackstedt LA, Murray JE. Regulation of Cocaine-related Behaviors by Estrogen and Progesterone. Neurosci Biobehav Rev 2022; 135:104584. [DOI: 10.1016/j.neubiorev.2022.104584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/30/2022] [Accepted: 02/12/2022] [Indexed: 10/19/2022]
|
38
|
Kruyer A, Dixon D, Angelis A, Amato D, Kalivas PW. Astrocytes in the ventral pallidum extinguish heroin seeking through GAT-3 upregulation and morphological plasticity at D1-MSN terminals. Mol Psychiatry 2022; 27:855-864. [PMID: 34642457 PMCID: PMC9054673 DOI: 10.1038/s41380-021-01333-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 09/16/2021] [Accepted: 09/29/2021] [Indexed: 12/20/2022]
Abstract
GABAergic projections from the nucleus accumbens core to the dorsolateral ventral pallidum are necessary for drug-conditioned cues to initiate relapse-like drug seeking. Astrocytes in the ventral pallidum are situated perisynaptically and regulate GABA transmission through expression of GABA uptake transporters, but whether they are involved in regulating drug seeking is unknown. To determine the contribution of ventral pallidal astrocytes to heroin seeking, we labeled astrocytes in male and female rats with a membrane-bound fluorescent tag and used confocal microscopy to quantify astroglial expression of the GABA transporter GAT-3 and astrocyte synaptic proximity after withdrawal from heroin self-administration and during 15 min of cued heroin seeking. We found that GAT-3 was upregulated in rats that had extinguished heroin seeking, but not in animals that were withdrawn from heroin without extinction training or in rats that extinguished sucrose seeking. When GAT-3 upregulation was reversed using a vivo-morpholino oligo, heroin seeking was restored in the extinguished context and extinction of cued heroin seeking was disrupted compared to control animals. Although astrocyte synaptic proximity was not altered overall after heroin withdrawal, examination of astrocyte proximity to accumbens D1- or D2-expressing afferents revealed a selective increase in astrocyte proximity with D1-expressing terminals during extinction of heroin self-administration. Experimentally-induced reduction of astrocyte synaptic proximity through knockdown of the astrocyte-selective actin-binding protein ezrin also markedly disrupted extinction of heroin seeking. Notably, GAT-3 or ezrin knockdown had no impact on context- or cue-induced seeking in sucrose-trained animals. These data show that astrocytes in the ventral pallidum undergo plasticity after extinction of heroin use that reduces seeking and highlight the importance of astrocyte-neuron interactions in shaping behaviors associated with opioid use disorder.
Collapse
Affiliation(s)
- Anna Kruyer
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
| | - Danielle Dixon
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Ariana Angelis
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Davide Amato
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
39
|
Allichon MC, Ortiz V, Pousinha P, Andrianarivelo A, Petitbon A, Heck N, Trifilieff P, Barik J, Vanhoutte P. Cell-Type-Specific Adaptions in Striatal Medium-Sized Spiny Neurons and Their Roles in Behavioral Responses to Drugs of Abuse. Front Synaptic Neurosci 2022; 13:799274. [PMID: 34970134 PMCID: PMC8712310 DOI: 10.3389/fnsyn.2021.799274] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/26/2021] [Indexed: 12/21/2022] Open
Abstract
Drug addiction is defined as a compulsive pattern of drug-seeking- and taking- behavior, with recurrent episodes of abstinence and relapse, and a loss of control despite negative consequences. Addictive drugs promote reinforcement by increasing dopamine in the mesocorticolimbic system, which alters excitatory glutamate transmission within the reward circuitry, thereby hijacking reward processing. Within the reward circuitry, the striatum is a key target structure of drugs of abuse since it is at the crossroad of converging glutamate inputs from limbic, thalamic and cortical regions, encoding components of drug-associated stimuli and environment, and dopamine that mediates reward prediction error and incentive values. These signals are integrated by medium-sized spiny neurons (MSN), which receive glutamate and dopamine axons converging onto their dendritic spines. MSN primarily form two mostly distinct populations based on the expression of either DA-D1 (D1R) or DA-D2 (D2R) receptors. While a classical view is that the two MSN populations act in parallel, playing antagonistic functional roles, the picture seems much more complex. Herein, we review recent studies, based on the use of cell-type-specific manipulations, demonstrating that dopamine differentially modulates dendritic spine density and synapse formation, as well as glutamate transmission, at specific inputs projecting onto D1R-MSN and D2R-MSN to shape persistent pathological behavioral in response to drugs of abuse. We also discuss the identification of distinct molecular events underlying the detrimental interplay between dopamine and glutamate signaling in D1R-MSN and D2R-MSN and highlight the relevance of such cell-type-specific molecular studies for the development of innovative strategies with potential therapeutic value for addiction. Because drug addiction is highly prevalent in patients with other psychiatric disorders when compared to the general population, we last discuss the hypothesis that shared cellular and molecular adaptations within common circuits could explain the co-occurrence of addiction and depression. We will therefore conclude this review by examining how the nucleus accumbens (NAc) could constitute a key interface between addiction and depression.
Collapse
Affiliation(s)
- Marie-Charlotte Allichon
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Vanesa Ortiz
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Paula Pousinha
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Andry Andrianarivelo
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Anna Petitbon
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Nicolas Heck
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Pierre Trifilieff
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Jacques Barik
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Peter Vanhoutte
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| |
Collapse
|
40
|
Zinsmaier AK, Dong Y, Huang YH. Cocaine-induced projection-specific and cell type-specific adaptations in the nucleus accumbens. Mol Psychiatry 2022; 27:669-686. [PMID: 33963288 PMCID: PMC8691189 DOI: 10.1038/s41380-021-01112-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 03/29/2021] [Accepted: 04/09/2021] [Indexed: 02/03/2023]
Abstract
Cocaine craving, seeking, and relapse are mediated, in part, by cocaine-induced adaptive changes in the brain reward circuits. The nucleus accumbens (NAc) integrates and prioritizes different emotional and motivational inputs to the reward system by processing convergent glutamatergic projections from the medial prefrontal cortex, basolateral amygdala, ventral hippocampus, and other limbic and paralimbic brain regions. Medium spiny neurons (MSNs) are the principal projection neurons in the NAc, which can be divided into two major subpopulations, namely dopamine receptor D1- versus D2-expressing MSNs, with complementing roles in reward-associated behaviors. After cocaine experience, NAc MSNs exhibit complex and differential adaptations dependent on cocaine regimen, withdrawal time, cell type, location (NAc core versus shell), and related input and output projections, or any combination of these factors. Detailed characterization of these cellular adaptations has been greatly facilitated by the recent development of optogenetic/chemogenetic techniques combined with transgenic tools. In this review, we discuss such cell type- and projection-specific adaptations induced by cocaine experience. Specifically, (1) D1 and D2 NAc MSNs frequently exhibit differential adaptations in spinogenesis, glutamatergic receptor trafficking, and intrinsic membrane excitability, (2) cocaine experience differentially changes the synaptic transmission at different afferent projections onto NAc MSNs, (3) cocaine-induced NAc adaptations exhibit output specificity, e.g., being different at NAc-ventral pallidum versus NAc-ventral tegmental area synapses, and (4) the input, output, subregion, and D1/D2 cell type may together determine cocaine-induced circuit plasticity in the NAc. In light of the projection- and cell-type specificity, we also briefly discuss ensemble and circuit mechanisms contributing to cocaine craving and relapse after drug withdrawal.
Collapse
Affiliation(s)
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15219,Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219
| | - Yanhua H. Huang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219
| |
Collapse
|
41
|
The role of the nucleus accumbens and ventral pallidum in feeding and obesity. Prog Neuropsychopharmacol Biol Psychiatry 2021; 111:110394. [PMID: 34242717 DOI: 10.1016/j.pnpbp.2021.110394] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 05/31/2021] [Accepted: 06/29/2021] [Indexed: 02/04/2023]
Abstract
Obesity is a growing global epidemic that stems from the increasing availability of highly-palatable foods and the consequent enhanced calorie consumption. Extensive research has shown that brain regions that are central to reward seeking modulate feeding and evidence linking obesity to pathology in such regions have recently started to accumulate. In this review we focus on the contribution of two major interconnected structures central to reward processing, the nucleus accumbens and the ventral pallidum, to obesity. We first review the known literature linking these structures to feeding behavior, then discuss recent advances connecting pathology in the nucleus accumbens and ventral pallidum to obesity, and finally examine the similarities and differences between drug addiction and obesity in the context of these two structures. The understanding of how pathology in brain regions involved in reward seeking and consumption may drive obesity and how mechanistically similar obesity and addiction are, is only now starting to be revealed. We hope that future research will advance knowledge in the field and open new avenues to studying and treating obesity.
Collapse
|
42
|
Gong S, Fayette N, Heinsbroek JA, Ford CP. Cocaine shifts dopamine D2 receptor sensitivity to gate conditioned behaviors. Neuron 2021; 109:3421-3435.e5. [PMID: 34506723 PMCID: PMC8571051 DOI: 10.1016/j.neuron.2021.08.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 07/16/2021] [Accepted: 08/11/2021] [Indexed: 12/21/2022]
Abstract
Cocaine addiction is a chronic, relapsing disorder characterized by maladaptation in the brain mesolimbic and nigrostriatal dopamine system. Although changes in the properties of D2-receptor-expressing medium spiny neurons (D2-MSNs) and connected striatal circuits following cocaine treatment are known, the contributions of altered D2-receptor (D2R) function in mediating the rewarding properties of cocaine remain unclear. Here, we describe how a 7-day exposure to cocaine alters dopamine signaling by selectively reducing the sensitivity, but not the expression, of nucleus accumbens D2-MSN D2Rs via an alteration in the relative expression and coupling of G protein subunits. This cocaine-induced reduction of D2R sensitivity facilitated the development of the rewarding effects of cocaine as blocking the reduction in G protein expression was sufficient to prevent cocaine-induced behavioral adaptations. These findings identify an initial maladaptive change in sensitivity by which mesolimbic dopamine signals are encoded by D2Rs following cocaine exposure.
Collapse
Affiliation(s)
- Sheng Gong
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Nicholas Fayette
- Department of Anesthesiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jasper A Heinsbroek
- Department of Anesthesiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Christopher P Ford
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
43
|
Andrianarivelo A, Saint-Jour E, Pousinha P, Fernandez SP, Petitbon A, De Smedt-Peyrusse V, Heck N, Ortiz V, Allichon MC, Kappès V, Betuing S, Walle R, Zhu Y, Joséphine C, Bemelmans AP, Turecki G, Mechawar N, Javitch JA, Caboche J, Trifilieff P, Barik J, Vanhoutte P. Disrupting D1-NMDA or D2-NMDA receptor heteromerization prevents cocaine's rewarding effects but preserves natural reward processing. SCIENCE ADVANCES 2021; 7:eabg5970. [PMID: 34669474 PMCID: PMC8528421 DOI: 10.1126/sciadv.abg5970] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Addictive drugs increase dopamine in the nucleus accumbens (NAc), where it persistently shapes excitatory glutamate transmission and hijacks natural reward processing. Here, we provide evidence, from mice to humans, that an underlying mechanism relies on drug-evoked heteromerization of glutamate N-methyl-d-aspartate receptors (NMDAR) with dopamine receptor 1 (D1R) or 2 (D2R). Using temporally controlled inhibition of D1R-NMDAR heteromerization, we unraveled their selective implication in early phases of cocaine-mediated synaptic, morphological, and behavioral responses. In contrast, preventing D2R-NMDAR heteromerization blocked the persistence of these adaptations. Interfering with these heteromers spared natural reward processing. Notably, we established that D2R-NMDAR complexes exist in human samples and showed that, despite a decreased D2R protein expression in the NAc, individuals with psychostimulant use disorder display a higher proportion of D2R forming heteromers with NMDAR. These findings contribute to a better understanding of molecular mechanisms underlying addiction and uncover D2R-NMDAR heteromers as targets with potential therapeutic value.
Collapse
Affiliation(s)
- Andry Andrianarivelo
- CNRS, UMR 8246, Neuroscience Paris Seine, F-75005 Paris, France
- INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, F-75005 Paris, France
- Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, F-75005 Paris, France
| | - Estefani Saint-Jour
- CNRS, UMR 8246, Neuroscience Paris Seine, F-75005 Paris, France
- INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, F-75005 Paris, France
- Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, F-75005 Paris, France
| | - Paula Pousinha
- Université Côte d’Azur, Nice, France
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR7275, Valbonne, France
| | - Sebastian P. Fernandez
- Université Côte d’Azur, Nice, France
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR7275, Valbonne, France
| | - Anna Petitbon
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, 33000 Bordeaux, France
| | | | - Nicolas Heck
- CNRS, UMR 8246, Neuroscience Paris Seine, F-75005 Paris, France
- INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, F-75005 Paris, France
- Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, F-75005 Paris, France
| | - Vanesa Ortiz
- Université Côte d’Azur, Nice, France
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR7275, Valbonne, France
| | - Marie-Charlotte Allichon
- CNRS, UMR 8246, Neuroscience Paris Seine, F-75005 Paris, France
- INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, F-75005 Paris, France
- Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, F-75005 Paris, France
| | - Vincent Kappès
- CNRS, UMR 8246, Neuroscience Paris Seine, F-75005 Paris, France
- INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, F-75005 Paris, France
- Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, F-75005 Paris, France
| | - Sandrine Betuing
- CNRS, UMR 8246, Neuroscience Paris Seine, F-75005 Paris, France
- INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, F-75005 Paris, France
- Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, F-75005 Paris, France
| | - Roman Walle
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, 33000 Bordeaux, France
| | - Ying Zhu
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
| | - Charlène Joséphine
- Commissariat à l’Énergie Atomique et aux Énergies Alternatives (CEA), Département de la Recherche Fondamentale, Institut de biologie François Jacob, MIRCen, and CNRS UMR 9199, Université Paris-Sud, Université Paris-Saclay, Neurodegenerative Diseases Laboratory, Fontenay-aux-Roses, France
| | - Alexis-Pierre Bemelmans
- Commissariat à l’Énergie Atomique et aux Énergies Alternatives (CEA), Département de la Recherche Fondamentale, Institut de biologie François Jacob, MIRCen, and CNRS UMR 9199, Université Paris-Sud, Université Paris-Saclay, Neurodegenerative Diseases Laboratory, Fontenay-aux-Roses, France
| | - Gustavo Turecki
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Naguib Mechawar
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Jonathan A. Javitch
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
- Department of Pharmacology, Columbia University, New York, NY 10032, USA
| | - Jocelyne Caboche
- CNRS, UMR 8246, Neuroscience Paris Seine, F-75005 Paris, France
- INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, F-75005 Paris, France
- Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, F-75005 Paris, France
| | - Pierre Trifilieff
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, 33000 Bordeaux, France
| | - Jacques Barik
- Université Côte d’Azur, Nice, France
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR7275, Valbonne, France
| | - Peter Vanhoutte
- CNRS, UMR 8246, Neuroscience Paris Seine, F-75005 Paris, France
- INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, F-75005 Paris, France
- Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, F-75005 Paris, France
- Corresponding author.
| |
Collapse
|
44
|
Kupchik YM, Prasad AA. Ventral pallidum cellular and pathway specificity in drug seeking. Neurosci Biobehav Rev 2021; 131:373-386. [PMID: 34562544 DOI: 10.1016/j.neubiorev.2021.09.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/03/2021] [Accepted: 09/05/2021] [Indexed: 01/12/2023]
Abstract
The ventral pallidum (VP) is central to the reinforcing effects across a variety of drugs and relapse to drug seeking. Emerging studies from animal models of reinstatement reveal a complex neurobiology of the VP that contributes to different aspects of relapse to drug seeking. This review builds on classical understanding of the VP as part of the final common pathway of relapse but also discusses the properties of the VP as an independent structure. These include VP neural anatomical subregions, cellular heterogeneity, circuitry, neurotransmitters and peptides. Collectively, this review provides a current understanding of the VP from molecular to circuit level architecture that contributes to both the appetitive and aversive symptoms of drug addiction. We show the complex neurobiology of the VP in drug seeking, emphasizing its critical role in addiction, and review strategic approaches that target the VP to reduce relapse rates.
Collapse
Affiliation(s)
- Yonatan M Kupchik
- Faculty of Medicine, The Hebrew University of Jerusalem, Ein Kerem. P.O. Box 12271, Jerusalem, 9112102, Israel
| | - Asheeta A Prasad
- School of Psychology, UNSW Sydney, NSW, 2052, Australia; Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia.
| |
Collapse
|
45
|
Neuroplasticity and Multilevel System of Connections Determine the Integrative Role of Nucleus Accumbens in the Brain Reward System. Int J Mol Sci 2021; 22:ijms22189806. [PMID: 34575969 PMCID: PMC8471564 DOI: 10.3390/ijms22189806] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 12/27/2022] Open
Abstract
A growing body of evidence suggests that nucleus accumbens (NAc) plays a significant role not only in the physiological processes associated with reward and satisfaction but also in many diseases of the central nervous system. Summary of the current state of knowledge on the morphological and functional basis of such a diverse function of this structure may be a good starting point for further basic and clinical research. The NAc is a part of the brain reward system (BRS) characterized by multilevel organization, extensive connections, and several neurotransmitter systems. The unique role of NAc in the BRS is a result of: (1) hierarchical connections with the other brain areas, (2) a well-developed morphological and functional plasticity regulating short- and long-term synaptic potentiation and signalling pathways, (3) cooperation among several neurotransmitter systems, and (4) a supportive role of neuroglia involved in both physiological and pathological processes. Understanding the complex function of NAc is possible by combining the results of morphological studies with molecular, genetic, and behavioral data. In this review, we present the current views on the NAc function in physiological conditions, emphasizing the role of its connections, neuroplasticity processes, and neurotransmitter systems.
Collapse
|
46
|
Lafferty CK, Christinck TD, Britt JP. All-optical approaches to studying psychiatric disease. Methods 2021; 203:46-55. [PMID: 34314828 DOI: 10.1016/j.ymeth.2021.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 11/17/2022] Open
Abstract
Improvements in all-optical means of monitoring and manipulating neural activity have generated new ways of studying psychiatric disease. The combination of calcium imaging techniques with optogenetics to concurrently record and manipulate neural activity has been used to create new disease models that link distinct circuit abnormalities to specific disease dimensions. These approaches represent a new path towards the development of more effective treatments, as they allow researchers to identify circuit manipulations that normalize pathological network activity. In this review we highlight the utility of all-optical approaches to generate new psychiatric disease models where the specific circuit abnormalities associated with disease symptomology can be assessed in vivo and in response to manipulations designed to normalize disease states. We then outline the principles underlying all-optical interrogations of neural circuits and discuss practical considerations for experimental design.
Collapse
Affiliation(s)
- Christopher K Lafferty
- Department of Psychology, McGill University, Montreal, QC, Canada; Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, QC, Canada
| | - Thomas D Christinck
- Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, QC, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Jonathan P Britt
- Department of Psychology, McGill University, Montreal, QC, Canada; Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, QC, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada.
| |
Collapse
|
47
|
Reactivating a positive feedback loop VTA-BLA-NAc circuit associated with positive experience ameliorates the attenuated reward sensitivity induced by chronic stress. Neurobiol Stress 2021; 15:100370. [PMID: 34381852 PMCID: PMC8334743 DOI: 10.1016/j.ynstr.2021.100370] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 12/24/2022] Open
Abstract
Both genetic predisposition and life events, particularly life stress, are thought to increase the risk for depression. Reward sensitivity appears to be attenuated in major depressive disorder (MDD), suggesting deficits in reward processing in these patients. We identified the VTA-BLA-NAc circuit as being activated by sex reward, and the VTA neurons that respond to sex reward are mostly dopaminergic. Acute or chronic reactivation of this circuit ameliorates the reward insensitivity induced by chronic restraint stress. Our histological and electrophysiological results show that the VTA neuron subpopulation responding to restraint stress, predominantly GABAergic neurons, inhibits the responsiveness of VTA dopaminergic neurons to reward stimuli, which is probably the mechanism by which stress modulates the reward processing neural circuits and subsequently disrupts reward-related behaviours. Furthermore, we found that the VTA-BLA-NAc circuit is a positive feedback loop. Blocking the projections from the BLA to the NAc associated with sex reward increases the excitability of VTA GABAergic neurons and decreases the excitability of VTA dopaminergic neurons, while activating this pathway decreases the excitability of VTA GABAergic neurons and increases the excitability of VTA dopaminergic neurons, which may be the cellular mechanism by which the VTA-BLA-NAc circuit associated with sex reward ameliorates the attenuated reward sensitivity induced by chronic stress.
Collapse
|
48
|
DRD3-dependent plasticity within the VP drives subcircuit activity critical for cocaine seeking. Neuron 2021; 109:2043-2044. [PMID: 34237276 DOI: 10.1016/j.neuron.2021.06.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The ventral pallidum (VP) is a key hub within the reward system that mediates drug-seeking behaviors. However, molecular and cellular adaptations within the VP following drug use are not fully elucidated. In this issue of Neuron, Pribiag et al. (2021) demonstrate how cocaine induces circuit-specific changes within the VP via dopamine-receptor-D3-dependent processes to promote cocaine seeking.
Collapse
|
49
|
Pribiag H, Shin S, Wang EHJ, Sun F, Datta P, Okamoto A, Guss H, Jain A, Wang XY, De Freitas B, Honma P, Pate S, Lilascharoen V, Li Y, Lim BK. Ventral pallidum DRD3 potentiates a pallido-habenular circuit driving accumbal dopamine release and cocaine seeking. Neuron 2021; 109:2165-2182.e10. [PMID: 34048697 PMCID: PMC9013317 DOI: 10.1016/j.neuron.2021.05.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 04/01/2021] [Accepted: 05/04/2021] [Indexed: 01/18/2023]
Abstract
Drugs of abuse induce persistent remodeling of reward circuit function, a process thought to underlie the emergence of drug craving and relapse to drug use. However, how circuit-specific, drug-induced molecular and cellular plasticity can have distributed effects on the mesolimbic dopamine reward system to facilitate relapse to drug use is not fully elucidated. Here, we demonstrate that dopamine receptor D3 (DRD3)-dependent plasticity in the ventral pallidum (VP) drives potentiation of dopamine release in the nucleus accumbens during relapse to cocaine seeking after abstinence. We show that two distinct VP DRD3+ neuronal populations projecting to either the lateral habenula (LHb) or the ventral tegmental area (VTA) display different patterns of activity during drug seeking following abstinence from cocaine self-administration and that selective suppression of elevated activity or DRD3 signaling in the LHb-projecting population reduces drug seeking. Together, our results uncover how circuit-specific DRD3-mediated plasticity contributes to the process of drug relapse.
Collapse
Affiliation(s)
- Horia Pribiag
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sora Shin
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA 24016, USA; Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA 24061, USA
| | - Eric Hou-Jen Wang
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Fangmiao Sun
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, 100871 10 Beijing, China; PKU-IDG/McGovern Institute for Brain Research, 100871 Beijing, China
| | - Paul Datta
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Alexander Okamoto
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Hayden Guss
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Akanksha Jain
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Xiao-Yun Wang
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Bruna De Freitas
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Patrick Honma
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Stefan Pate
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Varoth Lilascharoen
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Biological Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, 100871 10 Beijing, China; PKU-IDG/McGovern Institute for Brain Research, 100871 Beijing, China
| | - Byung Kook Lim
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA; Biological Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
50
|
Li YD, Luo YJ, Xu W, Ge J, Cherasse Y, Wang YQ, Lazarus M, Qu WM, Huang ZL. Ventral pallidal GABAergic neurons control wakefulness associated with motivation through the ventral tegmental pathway. Mol Psychiatry 2021; 26:2912-2928. [PMID: 33057171 PMCID: PMC8505244 DOI: 10.1038/s41380-020-00906-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 09/13/2020] [Accepted: 10/01/2020] [Indexed: 01/08/2023]
Abstract
The ventral pallidum (VP) regulates motivation, drug addiction, and several behaviors that rely on heightened arousal. However, the role and underlying neural circuits of the VP in the control of wakefulness remain poorly understood. In the present study, we sought to elucidate the specific role of VP GABAergic neurons in controlling sleep-wake behaviors in mice. Fiber photometry revealed that the population activity of VP GABAergic neurons was increased during physiological transitions from non-rapid eye movement (non-REM, NREM) sleep to either wakefulness or REM sleep. Moreover, chemogenetic and optogenetic manipulations were leveraged to investigate a potential causal role of VP GABAergic neurons in initiating and/or maintaining arousal. In vivo optogenetic stimulation of VP GABAergic neurons innervating the ventral tegmental area (VTA) strongly promoted arousal via disinhibition of VTA dopaminergic neurons. Functional in vitro mapping revealed that VP GABAergic neurons, in principle, inhibited VTA GABAergic neurons but also inhibited VTA dopaminergic neurons. In addition, optogenetic stimulation of terminals of VP GABAergic neurons revealed that they promoted arousal by innervating the lateral hypothalamus, but not the mediodorsal thalamus or lateral habenula. The increased wakefulness chemogenetically evoked by VP GABAergic neuronal activation was completely abolished by pretreatment with dopaminergic D1 and D2/D3 receptor antagonists. Furthermore, activation of VP GABAergic neurons increased exploration time in both the open-field and light-dark box tests but did not modulate depression-like behaviors or food intake. Finally, chemogenetic inhibition of VP GABAergic neurons decreased arousal. Taken together, our findings indicate that VP GABAergic neurons are essential for arousal related to motivation.
Collapse
Affiliation(s)
- Ya-Dong Li
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yan-Jia Luo
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Wei Xu
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Jing Ge
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yoan Cherasse
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Yi-Qun Wang
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Wei-Min Qu
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|