1
|
Montemarano A, Fox LD, Alkhaleel FA, Ostman AE, Sohail H, Pandey S, Murdaugh LB, Fox ME. A Drd1-cre mouse line with nucleus accumbens gene dysregulation exhibits blunted fentanyl seeking. Neuropsychopharmacology 2025:10.1038/s41386-025-02116-0. [PMID: 40316698 DOI: 10.1038/s41386-025-02116-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/04/2025] [Accepted: 04/22/2025] [Indexed: 05/04/2025]
Abstract
The synthetic opioid fentanyl remains abundant in the illicit drug supply, contributing to tens of thousands of overdose deaths every year. Despite this, the neurobiological effects of fentanyl use remain largely understudied. The nucleus accumbens (NAc) is a central locus promoting persistent drug use and relapse, largely dependent on activity of dopamine D1 receptors. NAc D1 receptor-expressing medium spiny neurons (D1-MSNs) undergo molecular and physiological neuroadaptations in response to chronic fentanyl that may promote relapse. Here, we obtained Drd1-cre120Mxu mice to investigate D1-dependent mechanisms of fentanyl relapse. We serendipitously discovered this mouse line has reduced fentanyl seeking, despite similar intravenous fentanyl self-administration, similar sucrose self-administration and seeking, and greater fentanyl-induced locomotion compared to wildtype counterparts. We found drug-naïve Drd1-cre120Mxu mice have elevated D1 receptor expression in NAc and increased sensitivity to the D1 receptor agonist SKF-38393. After fentanyl self-administration, Drd1-cre120Mxu mice exhibit divergent expression of MSN markers, opioid receptors, glutamate receptor subunits, and TrkB which may underly their blunted fentanyl seeking. Finally, we show fentanyl-related behavior is unaltered by chemogenetic manipulation of NAc core D1-MSNs in Drd1-cre120Mxu mice. Conversely, chemogenetic stimulation of ventral mesencephalon-projecting NAc core MSNs (putative D1-MSNs) in wildtype mice recapitulated the blunted fentanyl seeking of Drd1-cre120Mxu mice, supporting a role for aberrant D1-MSN signaling in this behavior. Together, our data uncover alterations in NAc gene expression and function with implications for susceptibility and resistance to developing fentanyl use disorder.
Collapse
Affiliation(s)
- Annalisa Montemarano
- Department of Anesthesiology and Perioperative Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Logan D Fox
- Department of Anesthesiology and Perioperative Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Farrah A Alkhaleel
- Department of Anesthesiology and Perioperative Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Alexandria E Ostman
- Department of Anesthesiology and Perioperative Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Hajra Sohail
- Department of Anesthesiology and Perioperative Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Samiksha Pandey
- Department of Anesthesiology and Perioperative Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Laura B Murdaugh
- Department of Anesthesiology and Perioperative Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Megan E Fox
- Department of Anesthesiology and Perioperative Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA.
- Department of Neuroscience and Experimental Therapeutics, Pennsylvania State University College of Medicine, Hershey, PA, USA.
| |
Collapse
|
2
|
Montemarano A, Fox LD, Alkhaleel FA, Ostman AE, Sohail H, Pandey S, Fox ME. A Drd1-cre mouse line with nucleus accumbens gene dysregulation exhibits blunted fentanyl seeking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.14.638324. [PMID: 40027693 PMCID: PMC11870424 DOI: 10.1101/2025.02.14.638324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
The synthetic opioid fentanyl remains abundant in the illicit drug supply, contributing to tens of thousands of overdose deaths every year. Despite this, the neurobiological effects of fentanyl use remain largely understudied. The nucleus accumbens (NAc) is a central locus promoting persistent drug use and relapse, largely dependent on activity of dopamine D1 receptors. NAc D1 receptor-expressing medium spiny neurons (D1-MSNs) undergo molecular and physiological adaptations that contribute to negative affect during fentanyl abstinence, but whether these neuroadaptations also promote fentanyl relapse is unclear. Here, we obtained Drd1-cre 120Mxu mice to investigate D1-dependent mechanisms of fentanyl relapse. We serendipitously discovered this mouse line is resistant to fentanyl seeking, despite similar intravenous fentanyl self-administration, and greater fentanyl-induced locomotion, compared to wildtype counterparts. In drug naïve mice, we found Drd1-cre 120Mxu mice have elevated D1 receptor expression in NAc, alongside increased expression of MSN marker genes Chrm4 and Penk . We show Drd1-cre 120Mxu mice have increased sensitivity to the D1 receptor agonist SKF-38393, and exhibit divergent expression of MSN markers, opioid receptors, glutamate receptor subunits, and TrkB after fentanyl self-administration that may underly blunted fentanyl seeking. Finally, we show fentanyl-related behavior is unaltered by chemogenetic manipulation of D1-MSNs in Drd1-cre 120Mxu mice. Conversely, chemogenetic stimulation of putative D1-MSNs in wildtype mice recapitulated the blunted fentanyl seeking of Drd1-cre 120Mxu mice, supporting a role for aberrant D1-MSN signaling in this behavior. Together, our data uncover alterations in NAc gene expression and function with implications for susceptibility and resistance to developing fentanyl use disorder.
Collapse
|
3
|
Kim HJ, Lee S, Kim GH, Sung K, Yoo T, Pyo JH, Jo HJ, Lee S, Lee HY, Jung JH, Lee KJ, Kim JH. GluN2B-mediated regulation of silent synapses for receptor specification and addiction memory. Exp Mol Med 2025; 57:436-449. [PMID: 39930130 PMCID: PMC11873126 DOI: 10.1038/s12276-025-01399-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 03/04/2025] Open
Abstract
Psychostimulants, including cocaine, elicit stereotyped, addictive behaviors. The reemergence of silent synapses containing only NMDA-type glutamate receptors is a critical mediator of addiction memory and seeking behaviors. Despite the predominant abundance of GluN2B-containing NMDA-type glutamate receptors in silent synapses, their operational mechanisms are not fully understood. Here, using conditional depletion/deletion of GluN2B in D1-expressing accumbal medium spiny neurons, we examined the synaptic and behavioral actions that silent synapses incur after repeated exposure to cocaine. GluN2B ablation reduces the proportion of silent synapses, but some of them can persist by substitution with GluN2C, which drives the aberrantly facilitated synaptic incorporation of calcium-impermeable AMPA-type glutamate receptors (AMPARs). The resulting precocious maturation of silent synapses impairs addiction memory but increases locomotor activity, both of which can be normalized by the blockade of calcium-impermeable AMPAR trafficking. Collectively, GluN2B supports the competence of cocaine-induced silent synapses to specify the subunit composition of AMPARs and thereby the expression of addiction memory and related behaviors.
Collapse
Affiliation(s)
- Hyun Jin Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
- Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Sangjun Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Gyu Hyun Kim
- Neural Circuits Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kibong Sung
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Taesik Yoo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Jung Hyun Pyo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Hee-Jung Jo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Sanghyeon Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyun-Young Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Jung Hoon Jung
- College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Kea Joo Lee
- Neural Circuits Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Joung-Hun Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea.
| |
Collapse
|
4
|
Negishi K, Navarro VI, Montes LP, Arzate LS, Guerra Ruiz JM, Sotelo D, Toccoli AR, Khan AM. Elaborating the connections of a closed-loop forebrain circuit in the rat: Circumscribed evidence for novel topography within a cortico-striato-pallidal triple descending projection, with thalamic feedback, to the anterior lateral hypothalamic area. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.18.633747. [PMID: 39868339 PMCID: PMC11761604 DOI: 10.1101/2025.01.18.633747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Motivated behaviors are regulated by distributed forebrain networks. Traditional approaches have often focused on individual brain regions and connections that do not capture the topographic organization of forebrain connectivity. We performed co-injections of anterograde and retrograde tract tracers in rats to provide novel high-spatial resolution evidence of topographic connections that elaborate a previously identified closed-loop forebrain circuit implicated in affective and motivational processes. The nodes of this circuit include select regions of the medial prefrontal cortex (defined here more specifically as the cingulate region, CNG), a dorsomedial portion of the nucleus accumbens (ACBdm), a portion of the medial substantia innominata (SIm), and the anterior lateral hypothalamic area (LHAa). The circuit also reportedly receives a feedback loop from the anterior region of the paraventricular thalamic nucleus (PVTa). In this draft report, we provide detailed circumscribed evidence supporting these regions as interconnected nodes, and provide several novel findings concerning the topographic organization of their projections. First, we identified the ACBdm based on its unique connectivity. Anterograde labeling from anterior paraventricular thalamic nucleus (PVTa) and retrograde labeling from medial substantia innominata (SIm) and lateral hypothalamic area (LHA) were restricted to the dorsomedial ACB (ACBdm). Strikingly, this labeling formed a longitudinal column extending along virtually the entire anteroposterior axis of ACBdm. Subsequent analysis revealed a convergence of ACBdm axon terminals and retrogradely labeled neurons from LHA within the anterior SIm. Furthermore, we identified cortical CNG regions related to this circuit. These regions contained retrograde labeling from both ACBdm and LHA, and anterograde labeling from PVTa. These cortical subdomains included regions previously implicated in the circuit but for which detailed organization has been unknown: (1) a region between the posterior prelimbic and infralimbic areas; (2) posterior part of basolateral and basomedial amygdalar nuclei, and (3) anterior pole of ventral subiculum. Our circumscribed findings, which await additional samples and analysis, support the existence of a topographically organized closed-loop circuit and identify two additional novel features: (1) direct evidence for an elaborate core rostrocaudal topography for a cortico-striato-pallidal motif comprising a triple descending projection to the LHA via direct, indirect, and "hyperdirect" pathways, and (2) a thalamic feedback system with specific projections to each cortical and striatal node of the circuit. We discuss the implications of this newly elaborated circuit for understanding the neural basis of motivational processes.
Collapse
Affiliation(s)
- Kenichiro Negishi
- UTEP Systems Neuroscience Laboratory, The University of Texas at El Paso, El Paso, TX 79968
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968
- PhD Program in Bioscience, The University of Texas at El Paso, El Paso, TX 79968
- HHMI PERSIST Brain Mapping & Connectomics Undergraduate Teaching Laboratory, The University of Texas at El Paso, El Paso, TX 79968
- Present address: Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD
| | - Vanessa I. Navarro
- UTEP Systems Neuroscience Laboratory, The University of Texas at El Paso, El Paso, TX 79968
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968
- PhD Program in Bioscience, The University of Texas at El Paso, El Paso, TX 79968
- UTEP RISE Program, The University of Texas at El Paso, El Paso, TX 79968
- HHMI PERSIST Brain Mapping & Connectomics Undergraduate Teaching Laboratory, The University of Texas at El Paso, El Paso, TX 79968
| | - Laura P. Montes
- UTEP Systems Neuroscience Laboratory, The University of Texas at El Paso, El Paso, TX 79968
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968
- PhD Program in Bioscience, The University of Texas at El Paso, El Paso, TX 79968
| | - Lidice Soto Arzate
- UTEP Systems Neuroscience Laboratory, The University of Texas at El Paso, El Paso, TX 79968
- UTEP LSAMP PRELS Program, The University of Texas at El Paso, El Paso, TX 79968
- HHMI PERSIST Brain Mapping & Connectomics Undergraduate Teaching Laboratory, The University of Texas at El Paso, El Paso, TX 79968
| | - Josdell M. Guerra Ruiz
- UTEP Systems Neuroscience Laboratory, The University of Texas at El Paso, El Paso, TX 79968
- HHMI PERSIST Brain Mapping & Connectomics Undergraduate Teaching Laboratory, The University of Texas at El Paso, El Paso, TX 79968
| | - Diana Sotelo
- UTEP Systems Neuroscience Laboratory, The University of Texas at El Paso, El Paso, TX 79968
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968
- UTEP RISE Program, The University of Texas at El Paso, El Paso, TX 79968
- HHMI PERSIST Brain Mapping & Connectomics Undergraduate Teaching Laboratory, The University of Texas at El Paso, El Paso, TX 79968
| | - Alejandro R. Toccoli
- UTEP Systems Neuroscience Laboratory, The University of Texas at El Paso, El Paso, TX 79968
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968
- UTEP RISE Program, The University of Texas at El Paso, El Paso, TX 79968
- HHMI PERSIST Brain Mapping & Connectomics Undergraduate Teaching Laboratory, The University of Texas at El Paso, El Paso, TX 79968
| | - Arshad M. Khan
- UTEP Systems Neuroscience Laboratory, The University of Texas at El Paso, El Paso, TX 79968
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968
- PhD Program in Bioscience, The University of Texas at El Paso, El Paso, TX 79968
- UTEP RISE Program, The University of Texas at El Paso, El Paso, TX 79968
- UTEP LSAMP PRELS Program, The University of Texas at El Paso, El Paso, TX 79968
- HHMI PERSIST Brain Mapping & Connectomics Undergraduate Teaching Laboratory, The University of Texas at El Paso, El Paso, TX 79968
| |
Collapse
|
5
|
Enriquez-Traba J, Arenivar M, Yarur-Castillo HE, Noh C, Flores RJ, Weil T, Roy S, Usdin TB, LaGamma CT, Wang H, Tsai VS, Kerspern D, Moritz AE, Sibley DR, Lutas A, Moratalla R, Freyberg Z, Tejeda HA. Dissociable control of motivation and reinforcement by distinct ventral striatal dopamine receptors. Nat Neurosci 2025; 28:105-121. [PMID: 39653808 PMCID: PMC12065418 DOI: 10.1038/s41593-024-01819-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 10/22/2024] [Indexed: 12/18/2024]
Abstract
Dopamine (DA) release in striatal circuits, including the nucleus accumbens medial shell (mNAcSh), tracks separable features of reward like motivation and reinforcement. However, the cellular and circuit mechanisms by which DA receptors transform DA release into distinct constructs of reward remain unclear. Here we show that DA D3 receptor (D3R) signaling in the mNAcSh drives motivated behavior in mice by regulating local microcircuits. Furthermore, D3Rs coexpress with DA D1 receptors, which regulate reinforcement, but not motivation. Paralleling dissociable roles in reward function, we report nonoverlapping physiological actions of D3R and DA D1 receptor signaling in mNAcSh neurons. Our results establish a fundamental framework wherein DA signaling within the same nucleus accumbens cell type is physiologically compartmentalized via actions on distinct DA receptors. This structural and functional organization provides neurons in a limbic circuit with the unique ability to orchestrate dissociable aspects of reward-related behaviors relevant to the etiology of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Juan Enriquez-Traba
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, MD, USA
- Department of Biochemistry, Universidad Autonoma de Madrid, Madrid, Spain
- Department of Functional and Systems Neurobiology, Instituto Cajal-CSIC, Madrid, Spain
| | - Miguel Arenivar
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, MD, USA
| | - Hector E Yarur-Castillo
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, MD, USA
| | - Chloe Noh
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, MD, USA
| | - Rodolfo J Flores
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, MD, USA
| | - Tenley Weil
- Section on Light and Circadian Rhythms, National Institute of Mental Health, Bethesda, MD, USA
| | - Snehashis Roy
- Systems Neuroscience Imaging Resource, National Institute of Mental Health, Bethesda, MD, USA
| | - Ted B Usdin
- Systems Neuroscience Imaging Resource, National Institute of Mental Health, Bethesda, MD, USA
| | - Christina T LaGamma
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, MD, USA
| | - Huikun Wang
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, MD, USA
| | - Valerie S Tsai
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, MD, USA
| | - Damien Kerspern
- Neuromodulation and Motivation Section, Diabetes, Endocrinology, & Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Amy E Moritz
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - David R Sibley
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Andrew Lutas
- Neuromodulation and Motivation Section, Diabetes, Endocrinology, & Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Rosario Moratalla
- Department of Functional and Systems Neurobiology, Instituto Cajal-CSIC, Madrid, Spain
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Hugo A Tejeda
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, MD, USA.
| |
Collapse
|
6
|
Zhou J, Luo D, An Y, Gao Y, Zhang J, Chen Y. Olfactory dysfunction decreased local field potential in the reward system and increased EtOH consumption in mice. Neurochem Int 2024; 180:105875. [PMID: 39393425 DOI: 10.1016/j.neuint.2024.105875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/24/2024] [Accepted: 10/01/2024] [Indexed: 10/13/2024]
Abstract
The relationship between olfactory dysfunction and alcohol intake is unobvious. Chronic alcohol intake results in reduced olfactory acuity and olfactory discrimination and addiction in humans. However, alcohol is a beverage with distinctive odors, which usually works as a cue to induce addictive memories and craving behavior. Whether olfactory impairment increase or decrease alcohol consumption remains an important but unclear issue. In this study, we measured ethanol (EtOH) consumption in the two-bottle choice EtOH drinking test, two bottle choice EtOH/sucrose drinking test and the drinking in the dark (DID) test during the olfactory loss. We also recorded local field potentials (LFPs) from the brain reward system, the ventral tegmental area (VTA), nucleus accumbens (NAc), and piriform cortex (Pir) one and four weeks after the induction of olfactory epithelium lesions using zinc sulfate (ZnSO4) in mice. The results showed that the EtOH consumption and preference were increased during the period of olfactory dysfunction. 1 week after the olfactory injury, LFP powers in the reward system at low- and high-gamma bands decreased significantly, coherence between the Pir and the reward system was also decrease. 4 weeks after the ZnSO4 treatment, LFP powers were reversed, but the coherence between VTA and NAc was decreased, indicating lasting effects post-recovery. This study demonstrates that olfactory dysfunction increased EtOH consumption in mice, which was accompanied by decreased LFP power and coherence in the reward system, which suggest that olfactory deficits changed activities in the reward system and could alter reward-seeking behaviors, which provide insights into the neurobiology of alcohol addiction.
Collapse
Affiliation(s)
- Jianhong Zhou
- Department of Basic Medicine, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, 650550, China; Institute of Medical Biology, Chinese Academy of Medical Sciences, Kunming, Yunnan, 650550, China
| | - Di Luo
- Department of Basic Medicine, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, 650550, China
| | - Yingjie An
- Department of Basic Medicine, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, 650550, China
| | - Yuan Gao
- Department of Basic Medicine, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, 650550, China; Department of Neurology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, 650034, China
| | - Jichuan Zhang
- Department of Basic Medicine, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, 650550, China
| | - Yanmei Chen
- Department of Basic Medicine, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, 650550, China.
| |
Collapse
|
7
|
Costa PA, Everett NA, Turner AJ, Umpierrez LS, Baracz SJ, Cornish JL. Adolescent alcohol binge drinking and withdrawal: behavioural, brain GFAP-positive astrocytes and acute methamphetamine effects in adult female rats. Psychopharmacology (Berl) 2024; 241:1539-1554. [PMID: 38705893 PMCID: PMC11269403 DOI: 10.1007/s00213-024-06580-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/25/2024] [Indexed: 05/07/2024]
Abstract
RATIONALE Alcopop beverages are generally the first alcoholic beverage that young females drink which contain high levels of sugar and alcohol. The over-consumption of these drinks may encourage alcohol co-administration with methamphetamine (METH) impacting on drinking behaviour and glial function. AIMS The aims of this study were to evaluate the effect of adolescent binge alcohol exposure on consumption level, anxiety-like behaviour, cross-sensitization with METH and on astrocyte expression in reward related brain regions. METHODS Adolescent female Sprague-Dawley rats had daily 1-hour oral alcohol consumption of alcopop (ALCP; with sucrose) or ethanol-only (ETOH; without sucrose), transitioned from 5 to 15% (v/v) ethanol content for 34 days. Water and sucrose groups act as controls. During alcohol withdrawal, rats were tested for anxiety on the elevated plus maze (EPM) and locomotor activity following saline or METH (1 mg/kg i.p) treatment. Brains were then collected to assess astrocyte immunofluorescence for glial fibrillary acidic protein (GFAP) in reward-related brain regions. RESULTS Rats pretreated with 5% ALCP consumed significantly more volume and ethanol intake when compared to 5% EtOH rats. Both ALCP and EtOH groups had a higher preference ratio for 5% than 15% alcohol solutions and ALCP rats had greater ethanol intake at 15% than EtOH rats. Alcohol withdrawal showed no significant differences between groups on anxiety, METH cross-sensitization effects or GFAP intensity in the regions studied. CONCLUSIONS Overall, the addition of sucrose to alcoholic solutions encouraged female rats to consume larger volumes and greater ethanol intake compared to ethanol-only solutions, yet did not have long lasting effects on behaviour and astrocytes.
Collapse
Affiliation(s)
- Priscila A Costa
- Behavioural Neuropharmacology Laboratory, School of Psychological Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia.
| | - Nicholas A Everett
- Behavioural Neuropharmacology Laboratory, School of Psychological Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia
| | - Anita J Turner
- Behavioural Neuropharmacology Laboratory, School of Psychological Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia
| | - Laísa S Umpierrez
- Behavioural Neuropharmacology Laboratory, School of Psychological Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia
| | - Sarah J Baracz
- Behavioural Neuropharmacology Laboratory, School of Psychological Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia
| | - Jennifer L Cornish
- Behavioural Neuropharmacology Laboratory, School of Psychological Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia.
| |
Collapse
|
8
|
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
|
9
|
Cristina Bianchi P, Palombo P, Antonagi Engi S, Eduardo Carneiro de Oliveira P, Emily Boaventura Tavares G, Anjos-Santos A, Suemi Yokoyama T, da Silva Planeta C, Cardoso Cruz F, Molini Leão R. Involvement of Pre-limbic Cortex-Nucleus accumbens projections in Context-Induced alcohol seeking. Brain Res 2024; 1841:149086. [PMID: 38876319 DOI: 10.1016/j.brainres.2024.149086] [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: 02/26/2024] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 06/16/2024]
Abstract
Alcohol use disorder (AUD) remains a critical public health issue worldwide, characterized by high relapse rates often triggered by contextual cues. This research investigates the neural mechanisms behind context-induced reinstatement of alcohol-seeking behavior, focusing on the nucleus accumbens and its interactions with the prelimbic cortex, employing Male Long-Evans rats in an ABA renewal model. In our experimental setup, rats were trained to self-administer 10 % ethanol in Context A, followed by extinction of lever pressing in the presence of discrete cues in Context B. The context-induced reinstatement of ethanol-seeking was then assessed by re-exposing rats to Context A or B under extinction conditions, aiming to simulate the environmental cues' influence on relapse behaviors. Three experiments were conducted: Experiment 1 utilized Fos-immunohistochemistry to examine neuronal activation in the nucleus accumbens; Experiment 2 applied the baclofen + muscimol inactivation technique to probe the functional importance of the nucleus accumbens core; Experiment 3 used Fos-immunofluorescence along with Retrobeads injection to investigate activation of neurons projecting from the prelimbic cortex to the nucleus accumbens core. Our findings revealed significant increases in Fos-immunoreactive nuclei within the nucleus accumbens core and shell during the reinstatement phase in Context A, underscoring the environment's potent effect on ethanol-seeking behavior. Additionally, inactivation of the nucleus accumbens core markedly reduced reinstatement, and there was a notable activation of neurons from the prelimbic cortex to the nucleus accumbens core in the ethanol-associated context. These results highlight the critical role of the nucleus accumbens core and its corticostriatal projections in the neural circuitry underlying context-driven ethanol seeking.
Collapse
Affiliation(s)
- Paula Cristina Bianchi
- Laboratory of Behavioral Neuroscience, Paulista Medicine School, Universidade Federal de São Paulo-UNIFESP, São Paulo, SP, Brazil
| | - Paola Palombo
- Laboratory of Behavioral Neuroscience, Paulista Medicine School, Universidade Federal de São Paulo-UNIFESP, São Paulo, SP, Brazil
| | - Sheila Antonagi Engi
- Laboratory of Behavioral Neuroscience, Paulista Medicine School, Universidade Federal de São Paulo-UNIFESP, São Paulo, SP, Brazil
| | | | | | - Alexia Anjos-Santos
- Laboratory of Behavioral Neuroscience, Paulista Medicine School, Universidade Federal de São Paulo-UNIFESP, São Paulo, SP, Brazil
| | - Thais Suemi Yokoyama
- Laboratory of Behavioral Neuroscience, Paulista Medicine School, Universidade Federal de São Paulo-UNIFESP, São Paulo, SP, Brazil
| | - Cleopatra da Silva Planeta
- Laboratory of Neuropsychopharmacology, School of Pharmaceutical Sciences, São Paulo State University, UNESP, Araraquara, SP, Brazil
| | - Fabio Cardoso Cruz
- Laboratory of Behavioral Neuroscience, Paulista Medicine School, Universidade Federal de São Paulo-UNIFESP, São Paulo, SP, Brazil
| | - Rodrigo Molini Leão
- Laboratory of Pharmacology, Biomedical Sciences Institute, Department of Pharmacology, Federal University of Uberlândia, Uberlândia, MG, Brazil; Graduate Program in Genetics and Biochemistry, Institute of Biotechnology, Federal University of Uberlândia/MG, Brazil.
| |
Collapse
|
10
|
Harada M, Capdevila LS, Wilhelm M, Burdakov D, Patriarchi T. Stimulation of VTA dopamine inputs to LH upregulates orexin neuronal activity in a DRD2-dependent manner. eLife 2024; 12:RP90158. [PMID: 38567902 PMCID: PMC10990487 DOI: 10.7554/elife.90158] [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] [Indexed: 04/05/2024] Open
Abstract
Dopamine and orexins (hypocretins) play important roles in regulating reward-seeking behaviors. It is known that hypothalamic orexinergic neurons project to dopamine neurons in the ventral tegmental area (VTA), where they can stimulate dopaminergic neuronal activity. Although there are reciprocal connections between dopaminergic and orexinergic systems, whether and how dopamine regulates the activity of orexin neurons is currently not known. Here we implemented an opto-Pavlovian task in which mice learn to associate a sensory cue with optogenetic dopamine neuron stimulation to investigate the relationship between dopamine release and orexin neuron activity in the lateral hypothalamus (LH). We found that dopamine release can be evoked in LH upon optogenetic stimulation of VTA dopamine neurons and is also naturally evoked by cue presentation after opto-Pavlovian learning. Furthermore, orexin neuron activity could also be upregulated by local stimulation of dopaminergic terminals in the LH in a way that is partially dependent on dopamine D2 receptors (DRD2). Our results reveal previously unknown orexinergic coding of reward expectation and unveil an orexin-regulatory axis mediated by local dopamine inputs in the LH.
Collapse
Affiliation(s)
- Masaya Harada
- Institute of Pharmacology and Toxicology, University of ZürichZürichSwitzerland
| | | | - Maria Wilhelm
- Institute of Pharmacology and Toxicology, University of ZürichZürichSwitzerland
| | - Denis Burdakov
- Neuroscience Center Zürich, University and ETH ZürichZürichSwitzerland
- Department of Health Sciences and Technology, ETH ZürichZürichSwitzerland
| | - Tommaso Patriarchi
- Institute of Pharmacology and Toxicology, University of ZürichZürichSwitzerland
- Neuroscience Center Zürich, University and ETH ZürichZürichSwitzerland
| |
Collapse
|
11
|
Enriquez-Traba J, Yarur-Castillo HE, Flores RJ, Weil T, Roy S, Usdin TB, LaGamma CT, Arenivar M, Wang H, Tsai VS, Moritz AE, Sibley DR, Moratalla R, Freyberg ZZ, Tejeda HA. Dissociable control of motivation and reinforcement by distinct ventral striatal dopamine receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.27.546539. [PMID: 37425766 PMCID: PMC10327105 DOI: 10.1101/2023.06.27.546539] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Dopamine release in striatal circuits, including the nucleus accumbens (NAc), tracks separable features of reward such as motivation and reinforcement. However, the cellular and circuit mechanisms by which dopamine receptors transform dopamine release into distinct constructs of reward remain unclear. Here, we show that dopamine D3 receptor (D3R) signaling in the NAc drives motivated behavior by regulating local NAc microcircuits. Furthermore, D3Rs co-express with dopamine D1 receptors (D1Rs), which regulate reinforcement, but not motivation. Paralleling dissociable roles in reward function, we report non-overlapping physiological actions of D3R and D1R signaling in NAc neurons. Our results establish a novel cellular framework wherein dopamine signaling within the same NAc cell type is physiologically compartmentalized via actions on distinct dopamine receptors. This structural and functional organization provides neurons in a limbic circuit with the unique ability to orchestrate dissociable aspects of reward-related behaviors that are relevant to the etiology of neuropsychiatric disorders.
Collapse
|
12
|
Claypool SM, Reiner DJ, Behdin S, Orihuel J, Batista A, Caldwell KE, Chow JJ, Bossert JM, Rubio FJ, Hope BT, Shaham Y. Role of Piriform Cortex and Its Afferent Projections in Relapse to Fentanyl Seeking after Food Choice-Induced Voluntary Abstinence. J Neurosci 2023; 43:2597-2614. [PMID: 36898838 PMCID: PMC10082459 DOI: 10.1523/jneurosci.0034-23.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/15/2023] [Accepted: 02/20/2023] [Indexed: 03/12/2023] Open
Abstract
We previously demonstrated a role of piriform cortex (Pir) in relapse to fentanyl seeking after food choice-induced voluntary abstinence. Here, we used this model to further study the role of Pir and its afferent projections in fentanyl relapse. We trained male and female rats to self-administer palatable food pellets for 6 d (6 h/day) and fentanyl (2.5 µg/kg/infusion, i.v.) for 12 d (6 h/day). We assessed relapse to fentanyl seeking after 12 voluntary abstinence sessions, achieved through a discrete choice procedure between fentanyl and palatable food (20 trials/session). We determined projection-specific activation of Pir afferents during fentanyl relapse with Fos plus the retrograde tracer cholera toxin B (injected into Pir). Fentanyl relapse was associated with increased Fos expression in anterior insular cortex (AI) and prelimbic cortex (PL) neurons projecting to Pir. We next used an anatomical disconnection procedure to determine the causal role of these two projections (AI→Pir and PL→Pir) in fentanyl relapse. Contralateral but not ipsilateral disconnection of AI→Pir projections decreased fentanyl relapse but not reacquisition of fentanyl self-administration. In contrast, contralateral but not ipsilateral disconnection of PL→Pir projections modestly decreased reacquisition but not relapse. Fluorescence-activated cell sorting and quantitative PCR data showed molecular changes within Pir Fos-expressing neurons associated with fentanyl relapse. Finally, we found minimal or no sex differences in fentanyl self-administration, fentanyl versus food choice, and fentanyl relapse. Our results indicate that AI→Pir and PL→Pir projections play dissociable roles in nonreinforced relapse to fentanyl seeking versus reacquisition of fentanyl self-administration after food choice-induced voluntary abstinence.SIGNIFICANCE STATEMENT We previously showed a role of Pir in fentanyl relapse after food choice-induced voluntary abstinence in rats, a procedure mimicking human abstinence or a significant reduction in drug self-administration because of the availability of alternative nondrug rewards. Here, we aimed to further characterize the role of Pir in fentanyl relapse by investigating the role of Pir afferent projections and analyzing molecular changes in relapse-activated Pir neurons. We identified dissociable roles of two Pir afferent projections (AI→Pir and PL→Pir) in relapse to fentanyl seeking versus reacquisition of fentanyl self-administration after voluntary abstinence. We also characterized molecular changes within Pir Fos-expressing neurons associated with fentanyl relapse.
Collapse
Affiliation(s)
- Sarah M Claypool
- Behavioral Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| | - David J Reiner
- Behavioral Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| | - Sana Behdin
- Behavioral Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| | - Javier Orihuel
- Behavioral Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| | - Ashley Batista
- Behavioral Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| | - Kiera E Caldwell
- Behavioral Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| | - Jonathan J Chow
- Behavioral Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| | - Jennifer M Bossert
- Behavioral Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| | - F Javier Rubio
- Behavioral Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| | - Bruce T Hope
- Behavioral Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| | - Yavin Shaham
- Behavioral Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| |
Collapse
|
13
|
Fox ME, Wulff AB, Franco D, Choi EY, Calarco CA, Engeln M, Turner MD, Chandra R, Rhodes VM, Thompson SM, Ament SA, Lobo MK. Adaptations in Nucleus Accumbens Neuron Subtypes Mediate Negative Affective Behaviors in Fentanyl Abstinence. Biol Psychiatry 2023; 93:489-501. [PMID: 36435669 PMCID: PMC9931633 DOI: 10.1016/j.biopsych.2022.08.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/25/2022] [Accepted: 08/24/2022] [Indexed: 01/18/2023]
Abstract
BACKGROUND Opioid discontinuation generates a withdrawal syndrome marked by increased negative affect. Increased symptoms of anxiety and dysphoria during opioid discontinuation are significant barriers to achieving long-term abstinence in opioid-dependent individuals. While adaptations in the nucleus accumbens are implicated in opioid abstinence syndrome, the precise neural mechanisms are poorly understood. Additionally, our current knowledge is limited to changes following natural and semisynthetic opioids, despite recent increases in synthetic opioid use and overdose. METHODS We used a combination of cell subtype-specific viral labeling and electrophysiology in male and female mice to investigate structural and functional plasticity in nucleus accumbens medium spiny neuron (MSN) subtypes after fentanyl abstinence. We characterized molecular adaptations after fentanyl abstinence with subtype-specific RNA sequencing and weighted gene co-expression network analysis. We used viral-mediated gene transfer to manipulate the molecular signature of fentanyl abstinence in D1-MSNs. RESULTS Here, we show that fentanyl abstinence increases anxiety-like behavior, decreases social interaction, and engenders MSN subtype-specific plasticity in both sexes. D1-MSNs, but not D2-MSNs, exhibit dendritic atrophy and an increase in excitatory drive. We identified a cluster of coexpressed dendritic morphology genes downregulated selectively in D1-MSNs that are transcriptionally coregulated by E2F1. E2f1 expression in D1-MSNs protects against loss of dendritic complexity, altered physiology, and negative affect-like behaviors caused by fentanyl abstinence. CONCLUSIONS Our findings indicate that fentanyl abstinence causes unique structural, functional, and molecular changes in nucleus accumbens D1-MSNs that can be targeted to alleviate negative affective symptoms during abstinence.
Collapse
Affiliation(s)
- Megan E Fox
- Departments of Anesthesiology and Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania; Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland.
| | - Andreas B Wulff
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Daniela Franco
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Eric Y Choi
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Cali A Calarco
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Michel Engeln
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Makeda D Turner
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ramesh Chandra
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Victoria M Rhodes
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Scott M Thompson
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland; Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Seth A Ament
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland; Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Mary Kay Lobo
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland; Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland.
| |
Collapse
|
14
|
Valyear MD, LeCocq MR, Brown A, Villaruel FR, Segal D, Chaudhri N. Learning processes in relapse to alcohol use: lessons from animal models. Psychopharmacology (Berl) 2023; 240:393-416. [PMID: 36264342 DOI: 10.1007/s00213-022-06254-x] [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: 05/04/2022] [Accepted: 10/02/2022] [Indexed: 11/29/2022]
Abstract
RATIONALE Alcohol use is reliably preceded by discrete and contextual stimuli which, through diverse learning processes, acquire the capacity to promote alcohol use and relapse to alcohol use. OBJECTIVE We review contemporary extinction, renewal, reinstatement, occasion setting, and sex differences research within a conditioning framework of relapse to alcohol use to inform the development of behavioural and pharmacological therapies. KEY FINDINGS Diverse learning processes and corresponding neurobiological substrates contribute to relapse to alcohol use. Results from animal models indicate that cortical, thalamic, accumbal, hypothalamic, mesolimbic, glutamatergic, opioidergic, and dopaminergic circuitries contribute to alcohol relapse through separable learning processes. Behavioural therapies could be improved by increasing the endurance and generalizability of extinction learning and should incorporate whether discrete cues and contexts influence behaviour through direct excitatory conditioning or occasion setting mechanisms. The types of learning processes that most effectively influence responding for alcohol differ in female and male rats. CONCLUSION Sophisticated conditioning experiments suggest that diverse learning processes are mediated by distinct neural circuits and contribute to relapse to alcohol use. These experiments also suggest that gender-specific behavioural and pharmacological interventions are a way towards efficacious therapies to prevent relapse to alcohol use.
Collapse
Affiliation(s)
- Milan D Valyear
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montréal, QC, Canada. .,Department of Psychology, McGill University, 1205 Ave. Dr. Penfield, Room N8/5, Montréal, QC, H3A 1B1, Canada.
| | - Mandy R LeCocq
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montréal, QC, Canada
| | - Alexa Brown
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montréal, QC, Canada
| | - Franz R Villaruel
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montréal, QC, Canada
| | - Diana Segal
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montréal, QC, Canada
| | - Nadia Chaudhri
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montréal, QC, Canada
| |
Collapse
|
15
|
Brown A, Villaruel FR, Chaudhri N. Neural correlates of recall and extinction in a rat model of appetitive Pavlovian conditioning. Behav Brain Res 2023; 440:114248. [PMID: 36496079 DOI: 10.1016/j.bbr.2022.114248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/30/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022]
Abstract
Extinction is a fundamental form of inhibitory learning that is important for adapting to changing environmental contingencies. While numerous studies have investigated the neural correlates of extinction using Pavlovian fear conditioning and appetitive operant reward-seeking procedures, less is known about the neural circuitry mediating the extinction of appetitive Pavlovian responding. Here, we aimed to generate an extensive brain activation map of extinction learning in a rat model of appetitive Pavlovian conditioning. Male Long-Evans rats were trained to associate a conditioned stimulus (CS; 20 s white noise) with the delivery of a 10% sucrose unconditioned stimulus (US; 0.3 ml/CS) to a fluid port. Control groups also received CS presentations, but sucrose was delivered either during the inter-trial interval or in the home-cage. After conditioning, 1 or 6 extinction sessions were conducted in which the CS was presented but sucrose was withheld. We performed Fos immunohistochemistry and network connectivity analyses on a set of cortical, striatal, thalamic, and amygdalar brain regions. Neural activity in the prelimbic cortex, ventral orbitofrontal cortex, nucleus accumbens core, and paraventricular nucleus of the thalamus was greater during recall relative to extinction. Conversely, prolonged extinction following 6 sessions induced increased neural activity in the infralimbic cortex, medial orbitofrontal cortex, and nucleus accumbens shell compared to home-cage controls. All these structures were similarly recruited during recall on the first extinction session. These findings provide novel evidence for the contribution of brain areas and neural networks that are differentially involved in the recall versus extinction of appetitive Pavlovian conditioned responding.
Collapse
Affiliation(s)
- Alexa Brown
- Center for Studies in Behavioural Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada.
| | - Franz R Villaruel
- Center for Studies in Behavioural Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada
| | - Nadia Chaudhri
- Center for Studies in Behavioural Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada
| |
Collapse
|
16
|
Murphy S, Collis Glynn M, Dixon TN, Grill HJ, McNally GP, Ong ZY. Nucleus of the solitary tract A2 neurons control feeding behaviors via projections to the paraventricular hypothalamus. Neuropsychopharmacology 2023; 48:351-361. [PMID: 36114285 PMCID: PMC9751294 DOI: 10.1038/s41386-022-01448-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/08/2022] [Accepted: 08/29/2022] [Indexed: 12/26/2022]
Abstract
Hindbrain NTS neurons are highly attuned to internal physiological and external environmental factors that contribute to the control of food intake but the relevant neural phenotypes and pathways remain elusive. Here, we investigated the role of NTS A2 neurons and their projections in the control of feeding behaviors. In male TH Cre rats, we first confirmed selective targeting of NTS A2 neurons and showed that chemogenetic stimulation of these neurons significantly suppressed dark cycle food intake, deprivation re-feed and high fat diet intake. Despite reducing intake, activation of NTS A2 neurons had no effect on food approach, anxiety-like behaviors, locomotor activity, blood glucose levels nor did it induce nausea/malaise, thus revealing a selective role for these neurons in the consummatory aspect of food intake control. Pathway-specific mapping and manipulation of NTS A2 neurons showed that these effects were mediated by NTS A2 neurons projecting to the paraventricular nucleus of the hypothalamus (PVH) because chemogenetic activation of these projections, but not projections to bed nucleus of the stria terminalis (BNST), reduced food intake. Cell-type specific analyses demonstrated that activation of NTS A2 neurons recruited both PVH oxytocin (OT)- and corticotropin-releasing factor (CRF)-expressing neurons, and plasma analyses showed increased plasma corticosterone following NTS A2 stimulation. While we also showed that chemogenetic inhibition of NTS A2 neurons attenuated the intake inhibitory effects of CCK, the specificity of transgene expression was low. Together, these findings showed that NTS A2 neurons are sufficient to control the consummatory aspects of feeding, regardless of energy status or food palatability and identified their projections to PVH, but not BNST, in food intake control.
Collapse
Affiliation(s)
- Stephanie Murphy
- School of Psychology, University of New South Wales, UNSW, Sydney, NSW, 2052, Australia
| | - Metika Collis Glynn
- School of Psychology, University of New South Wales, UNSW, Sydney, NSW, 2052, Australia
| | - Tiarani N Dixon
- School of Psychology, University of New South Wales, UNSW, Sydney, NSW, 2052, Australia
| | - Harvey J Grill
- Department of Psychology, Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Gavan P McNally
- School of Psychology, University of New South Wales, UNSW, Sydney, NSW, 2052, Australia
| | - Zhi Yi Ong
- School of Psychology, University of New South Wales, UNSW, Sydney, NSW, 2052, Australia.
| |
Collapse
|
17
|
Hammerslag LR, Humburg BA, Malone SG, Beckmann JS, Saatman KE, Grinevich V, Bardo MT. Peer-induced cocaine seeking in rats: Comparison to nonsocial stimuli and role of paraventricular hypothalamic oxytocin neurons. Addict Biol 2022; 27:e13217. [PMID: 36001434 PMCID: PMC9413367 DOI: 10.1111/adb.13217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 07/13/2022] [Accepted: 07/18/2022] [Indexed: 11/27/2022]
Abstract
The purpose of this study was to determine if social vs nonsocial cues (peer vs light/tone) can serve as discriminative stimuli to reinstate cocaine seeking. In addition, to assess a potential mechanism, an oxytocin (OT) promoter-linked hM3Dq DREADD was infused into the paraventricular nucleus of the hypothalamus to determine whether peer-induced cocaine seeking is decreased by activation of OT neurons. Male rats underwent twice-daily self-administration sessions, once with cocaine in the presence of one peer (S+) and once with saline in the presence of a different peer (S-). Another experiment used similar procedures, except the discriminative stimuli were nonsocial (constant vs flashing light/tone), with one stimulus paired with cocaine (S+) and the other paired with saline (S-). A third experiment injected male and female rats with OTp-hM3Dq DREADD or control virus into PVN and tested them for peer-induced reinstatement of cocaine seeking following clozapine (0.1 mg/kg). Although acquisition of cocaine self-administration was similar in rats trained with either peer or light/tone discriminative stimuli, the latency to first response was reduced by the peer S+, but not by the light/tone S+. In addition, the effect of the conditioned stimulus was overshadowed by the peer S+ but not by the light/tone S+. Clozapine blocked the effect of the peer S+ in rats receiving the OTp-hM3Dq DREADD virus, but not in rats receiving the control virus. These results demonstrate that a social peer can serve as potent trigger for drug seeking and that OT in PVN modulates peer-induced reinstatement of cocaine seeking.
Collapse
Affiliation(s)
| | - Bree A. Humburg
- Department of Psychology, University of Kentucky, USA, 40536
| | | | | | - Kathryn E. Saatman
- Department of Physiology, and Spinal Cord and Brain Injury Research Center, University of Kentucky, USA, 40536
| | - Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | |
Collapse
|
18
|
Jin S, Maddern XJ, Campbell EJ, Lawrence AJ. Examining ventral subiculum and basolateral amygdala projections to the nucleus accumbens shell: Differential expression of VGLuT1, VGLuT2 and VGaT in the rat. Neurosci Lett 2022; 788:136858. [PMID: 36038028 DOI: 10.1016/j.neulet.2022.136858] [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: 07/15/2022] [Revised: 08/18/2022] [Accepted: 08/23/2022] [Indexed: 12/01/2022]
Abstract
Projections to the striatum are well-identified. For example, in the ventral striatum, two major inputs to the medial nucleus accumbens shell include the ventral subiculum and basolateral amygdala. However, the chemical phenotype(s) of these projection neurons remain unclear. In this study, we examined amygdalostriatal and corticostriatal connectivity in rats using injections of the retrograde tracer cholera toxin b into the nucleus accumbens shell. To determine the neurotransmitter identity of projection neurons, we combined retrograde tracing with RNAscope in-situ hybridization, using mRNA probes against vesicular transporters associated with glutamatergic (VGluT1 - Slc17a7, VGluT2 - Slc17a6) or GABAergic (VGaT - Slc32a1) neurotransmission. Confocal imaging was used to examine vesicular transporter mRNA expression in the ventral subiculum and basolateral amygdala inputs to the nucleus accumbens shell. Both projections contained mostly VGluT1-expressing neurons. Interestingly, almost a quarter of ventral subiculum to nucleus accumbens shell projections co-expressed VGluT1 and VGluT2 compared to a relatively small number (∼3%) that were co-expressed in basolateral amygdala to nucleus accumbens shell afferents. However, almost a quarter of basolateral amygdala to nucleus accumbens shell projections were VGaT-positive. These findings highlight the diverse proportions of glutamatergic and GABAergic afferents in two major projections to the nucleus accumbens shell and raise important questions for functional studies.
Collapse
Affiliation(s)
- Shubo Jin
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3010, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia
| | - Xavier J Maddern
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3010, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia
| | - Erin J Campbell
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3010, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia.
| | - Andrew J Lawrence
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3010, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia.
| |
Collapse
|
19
|
Zhao ZD, Han X, Chen R, Liu Y, Bhattacherjee A, Chen W, Zhang Y. A molecularly defined D1 medium spiny neuron subtype negatively regulates cocaine addiction. SCIENCE ADVANCES 2022; 8:eabn3552. [PMID: 35960793 PMCID: PMC9374336 DOI: 10.1126/sciadv.abn3552] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 06/30/2022] [Indexed: 05/21/2023]
Abstract
The striatum plays a critical role in regulating addiction-related behaviors. The conventional dichotomy model suggests that striatal D1/D2 medium spiny neurons (MSNs) positively/negatively regulate addiction-related behaviors. However, this model does not account for the neuronal heterogeneity and functional diversity of the striatum, and whether MSN subtypes beyond the pan-D1/D2 populations play distinct roles in drug addiction remains unknown. We characterized the role of a tachykinin 2-expressing D1 MSN subtype (Tac2+), present in both rodent and primate striatum, using cocaine addiction mouse models. We found that acute cocaine administration reduces Tac2 neuronal activity, and cocaine conditioning alters neuronal response related to cocaine reward contextual associations. In addition, activation/inhibition of Tac2+ neurons attenuates/promotes cocaine-induced conditioned place preference and cocaine intravenous self-administration. Furthermore, stimulation of the NAc-to-lateral hypothalamic projection of Tac2+ neurons suppresses cocaine reward behavior. Our study reveals an unconventional negative regulatory function of D1 MSNs in drug addiction that operates in a subtype- and projection-specific manner.
Collapse
Affiliation(s)
- Zheng-dong Zhao
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Xiao Han
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Renchao Chen
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Yiqiong Liu
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Aritra Bhattacherjee
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Wenqiang Chen
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Yi Zhang
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, WAB-149G, 200 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
20
|
Xie C, Power J, Prasad AA. Bidirectional Optogenetic Modulation of the Subthalamic Nucleus in a Rodent Model of Parkinson's Disease. Front Neurosci 2022; 16:848821. [PMID: 35655750 PMCID: PMC9152094 DOI: 10.3389/fnins.2022.848821] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/31/2022] [Indexed: 11/17/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by a range of motor symptoms. Treatments are focused on dopamine replacement therapy or deep brain stimulation (DBS). The subthalamic nucleus (STN) is a common target for DBS treatment of PD. However, the function of the STN in normal conditions and pathology is poorly understood. Here, we show in rats that optogenetic modulation of STN neuronal activity exerts bidirectional control of motor function, where inhibition of the STN increases movement and STN activation decreases movement. We also examined the effect of bidirectional optogenetic manipulation STN neuronal activity under dopamine depleted condition using the bilateral rodent 6-hydroxydopamine (6-OHDA) model of Parkinson's disease. Optogenetic inhibition of the STN in the absence of dopamine had no impact on motor control yet STN excitation led to pronounced abnormal involuntary movement. Administration of levodopa rescued the abnormal involuntary movements induced by STN excitation. Although dopamine and STN dysfunction are well established in PD pathology, here we demonstrate simultaneous STN over activity and loss of dopamine lead to motor deficits. Moreover, we show the dysfunction of the STN is dependent on dopamine. This study provides evidence that the loss of dopamine and the over activity of the STN are key features of PD motor deficits. These results provide insight into the STN pathology in PD and therapeutic mechanism of targeting the STN for the treatment for PD.
Collapse
Affiliation(s)
- Caroline Xie
- School of Psychology, University of New South Wales, Sydney, NSW, Australia
| | - John Power
- Department of Physiology and Translational Neuroscience Facility, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Asheeta A. Prasad
- School of Psychology, University of New South Wales, Sydney, NSW, Australia
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
21
|
Deehan GA. The enduring behavioral and neurobiological effects of a flavor cue paired with alcohol drinking during adolescence on the incentive properties of the flavor cue in adulthood in female alcohol-preferring (P) rats. Drug Alcohol Depend 2022; 232:109289. [PMID: 35051698 DOI: 10.1016/j.drugalcdep.2022.109289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/07/2021] [Accepted: 12/27/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Alcohol use disorders (AUDs) affect 15 million people nationwide, 4% of which are adolescents (ages 12-17) and adolescents who binge drink significantly increase their likelihood of suffering from an AUD in adulthood. Research shows that cues (i.e. flavors) paired with alcohol (EtOH) produce significant cue-induced alcohol craving and contribute to relapse in adolescent and adult populations. However, there is a lack of research focused on how cues that accompany EtOH drinking during adolescence, affect EtOH craving later in life. The current study sought to examine the sex- and developmental-dependent effects of adolescent exposure to flavor cues associated with EtOH on operant-lick behavior and cue-induced dopamine (DA) levels within the nucleus accumbens shell (AcbSh; reward structure) in adulthood. METHODS Adolescent alcohol-preferring (P) rats were randomly assigned to one of 4 groups and received 24 hr. access to three bottles on their home cage: Paired: 0.1% blueberry flavor extract (BB) + 15% v/v EtOH and 2 water bottles; Unpaired: 0.1% BB, 15% v/v EtOH, and water; 15% EtOH alone, and 2 water bottles; BB alone and 2 water bottles. Home cage fluid consumption was measured for 2-weeks. On the third week bottles were removed and all animals underwent 9 days of operant training using an operant sipper paradigm. This consisted of two sipper spouts connected to the computer by a lickometer, which registered tongue contacts with the sipper tube (Paired: BB+EtOH or water; Unpaired BB or EtOH; EtOH alone: EtOH or water; BB alone: BB or water). When the fixed ratio (FR) requirement for number of licks/tongue contacts was met, a liquid delivery solenoid dispensed 0.05 ml of fluid into the sipper tube. Following the final operant session all rats remained in their home-cage for approximately 40 days until adulthood at which point they were returned to the operant chambers and tested for appetitive and consummatory behavior in response to the flavor cue (all rats: BB or water; NO EtOH). Two weeks after the final operant session all rats underwent microdialysis testing to examine cue-induced DA levels in the AcbSh. RESULTS Data indicated that animals in the paired group exhibited a significantly greater level of licking at the BB sipper and a significantly greater level of DA release in response to the flavor cue compared to the other groups. CONCLUSIONS Overall, the data suggest that cues paired with EtOH during adolescence may produce persistent changes to the behavioral and neurobiological mechanisms that contribute to an increased risk of developing an AUD later in life.
Collapse
Affiliation(s)
- Gerald A Deehan
- Department of Psychology, East Tennessee State University, Johnson City, Tennessee, USA.
| |
Collapse
|
22
|
Hunger dampens a nucleus accumbens circuit to drive persistent food seeking. Curr Biol 2022; 32:1689-1702.e4. [DOI: 10.1016/j.cub.2022.02.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/11/2022] [Accepted: 02/09/2022] [Indexed: 02/07/2023]
|
23
|
Chen R, Blosser TR, Djekidel MN, Hao J, Bhattacherjee A, Chen W, Tuesta LM, Zhuang X, Zhang Y. Decoding molecular and cellular heterogeneity of mouse nucleus accumbens. Nat Neurosci 2021; 24:1757-1771. [PMID: 34663959 PMCID: PMC8639815 DOI: 10.1038/s41593-021-00938-x] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 09/07/2021] [Indexed: 11/09/2022]
Abstract
The nucleus accumbens (NAc) plays an important role in regulating multiple behaviors, and its dysfunction has been linked to many neural disorders. However, the molecular, cellular and anatomic heterogeneity underlying its functional diversity remains incompletely understood. In this study, we generated a cell census of the mouse NAc using single-cell RNA sequencing and multiplexed error-robust fluorescence in situ hybridization, revealing a high level of cell heterogeneity in this brain region. Here we show that the transcriptional and spatial diversity of neuron subtypes underlie the NAc's anatomic and functional heterogeneity. These findings explain how the seemingly simple neuronal composition of the NAc achieves its highly heterogenous structure and diverse functions. Collectively, our study generates a spatially resolved cell taxonomy for understanding the structure and function of the NAc, which demonstrates the importance of combining molecular and spatial information in revealing the fundamental features of the nervous system.
Collapse
Affiliation(s)
- Renchao Chen
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Timothy R Blosser
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Department of Physics,, Harvard University, Cambridge, MA, USA
| | - Mohamed N Djekidel
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Junjie Hao
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Department of Physics,, Harvard University, Cambridge, MA, USA
| | - Aritra Bhattacherjee
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Wenqiang Chen
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Luis M Tuesta
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Xiaowei Zhuang
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA.
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.
- Department of Physics,, Harvard University, Cambridge, MA, USA.
| | - Yi Zhang
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA.
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA.
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Boston, MA, USA.
| |
Collapse
|
24
|
Egervari G, Siciliano CA, Whiteley EL, Ron D. Alcohol and the brain: from genes to circuits. Trends Neurosci 2021; 44:1004-1015. [PMID: 34702580 PMCID: PMC8616825 DOI: 10.1016/j.tins.2021.09.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/08/2021] [Accepted: 09/30/2021] [Indexed: 01/27/2023]
Abstract
Alcohol use produces wide-ranging and diverse effects on the central nervous system. It influences intracellular signaling mechanisms, leading to changes in gene expression, chromatin remodeling, and translation. As a result of these molecular alterations, alcohol affects the activity of neuronal circuits. Together, these mechanisms produce long-lasting cellular adaptations in the brain that in turn can drive the development and maintenance of alcohol use disorder (AUD). We provide an update on alcohol research, focusing on multiple levels of alcohol-induced adaptations, from intracellular changes to changes in neural circuits. A better understanding of how alcohol affects these diverse and interlinked mechanisms may lead to the identification of novel therapeutic targets and to the development of much-needed novel and efficacious treatment options.
Collapse
Affiliation(s)
- Gabor Egervari
- Department of Cell and Developmental Biology, Epigenetics Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Cody A Siciliano
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37203, USA.
| | - Ellanor L Whiteley
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dorit Ron
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
25
|
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: 24] [Impact Index Per Article: 6.0] [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
|
26
|
Giannotti G, Gong S, Fayette N, Heinsbroek JA, Orfila JE, Herson PS, Ford CP, Peters J. Extinction blunts paraventricular thalamic contributions to heroin relapse. Cell Rep 2021; 36:109605. [PMID: 34433067 PMCID: PMC8418780 DOI: 10.1016/j.celrep.2021.109605] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/25/2021] [Accepted: 08/03/2021] [Indexed: 12/31/2022] Open
Abstract
Here, we use optogenetics and chemogenetics to investigate the contribution of the paraventricular thalamus (PVT) to nucleus accumbens (NAc) pathway in aversion and heroin relapse in two different heroin self-administration models in rats. In one model, rats undergo forced abstinence in the home cage prior to relapse testing, and in the other, they undergo extinction training, a procedure that is likened to cognitive behavioral therapy. We find that the PVT→NAc pathway is both sufficient and necessary to drive aversion and heroin seeking after abstinence, but not extinction. The ability of extinction to reduce this pathway's contribution to heroin relapse is accompanied by a loss of synaptic plasticity in PVT inputs onto a specific subset of NAc neurons. Thus, extinction may exert therapeutic reductions in opioid seeking by altering synaptic plasticity within the PVT→NAc pathway, resulting in reduced aversion during opioid withdrawal as well as reduced relapse propensity.
Collapse
Affiliation(s)
- Giuseppe Giannotti
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sheng Gong
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Nicholas Fayette
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jasper A Heinsbroek
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - James E Orfila
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Paco S Herson
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Christopher P Ford
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jamie Peters
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
27
|
Yu X, Chen S, Shan Q. Depression in the Direct Pathway of the Dorsomedial Striatum Permits the Formation of Habitual Action. Cereb Cortex 2021; 31:3551-3564. [PMID: 33774666 DOI: 10.1093/cercor/bhab031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 01/16/2021] [Accepted: 01/26/2021] [Indexed: 02/05/2023] Open
Abstract
In order to achieve optimal outcomes in an ever-changing environment, humans and animals generally manage their action control via either goal-directed action or habitual action. These two action strategies are thought to be encoded in distinct parallel circuits in the dorsal striatum, specifically, the posterior dorsomedial striatum (DMS) and the dorsolateral striatum (DLS), respectively. The striatum is primarily composed of two subtypes of medium spiny neurons (MSNs): the direct-pathway striatonigral and the indirect-pathway striatopallidal MSNs. MSN-subtype-specific synaptic plasticity in the DMS and the DLS has been revealed to underlie goal-directed action and habitual action, respectively. However, whether any MSN-subtype-specific synaptic plasticity in the DMS is associated with habitual action, and if so, whether the synaptic plasticity affects the formation of habitual action, are not known. This study demonstrates that postsynaptic depression in the excitatory synapses of the direct-pathway striatonigral MSNs in the DMS is formed after habit learning. Moreover, chemogenetically rescuing this depression compromises the acquisition, but not the expression, of habitual action. These findings reveal that an MSN-subtype-specific synaptic plasticity in the DMS affects habitual action and suggest that plasticity in the DMS as well as in the DLS contributes to the formation of habitual action.
Collapse
Affiliation(s)
- Xiaoxuan Yu
- Laboratory for Synaptic Plasticity, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Shijie Chen
- Laboratory for Synaptic Plasticity, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Qiang Shan
- Laboratory for Synaptic Plasticity, Shantou University Medical College, Shantou, Guangdong 515041, China
| |
Collapse
|
28
|
Walker LC, Huckstep KL, Chen NA, Hand LJ, Lindsley CW, Langmead CJ, Lawrence AJ. Muscarinic M 4 and M 5 receptors in the ventral subiculum differentially modulate alcohol seeking versus consumption in male alcohol-preferring rats. Br J Pharmacol 2021; 178:3730-3746. [PMID: 33942300 DOI: 10.1111/bph.15513] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/11/2021] [Accepted: 04/20/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Muscarinic acetylcholine receptors mediate alcohol consumption and seeking in rats. While M4 and M5 receptors have recently been implicated to mediate these behaviours in the striatum, their role in other brain regions remain unknown. The ventral tegmental area (VTA) and ventral subiculum (vSub) both densely express M4 and M5 receptors and modulate alcohol-seeking, via their projections to the nucleus accumbens shell (AcbSh). EXPERIMENTAL APPROACH In Indiana alcohol-preferring (iP) male rats, we examined Chrm4 (M4 ) and Chrm5 (M5 ) expression in the VTA and vSub following long-term alcohol consumption and abstinence using RT-qPCR. Using a combination of retrograde tracing and RNAscope, we examined the localisation of Chrm4 and Chrm5 on vSub cells that project to the AcbSh. Using selective allosteric modulators, we examined the functional role of M4 and M5 receptors within the vSub in alcohol consumption, context-induced alcohol-seeking, locomotor activity, and food/water consumption. KEY RESULTS Long-term alcohol and abstinence dysregulated the expression of genes for muscarinic receptors in the vSub, not in the VTA. Chrm4 was down-regulated following long-term alcohol and abstinence, while Chrm5 was up-regulated following long-term alcohol consumption. Consistent with these data, a positive allosteric modulator (VU0467154) of intra-vSub M4 receptors reduced context-induced alcohol-seeking, but not motivation for alcohol self-administration, while M5 receptor negative allosteric modulator (ML375) reduced initial motivation for alcohol self-administration, but not context-induced alcohol-seeking. CONCLUSION AND IMPLICATIONS Collectively, our data highlight alcohol-induced cholinergic dysregulation in the vSub and distinct roles for M4 and M5 receptor allosteric modulators to reduce alcohol consumption or seeking.
Collapse
Affiliation(s)
- Leigh C Walker
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Kate L Huckstep
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Nicola A Chen
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Lexi J Hand
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Craig W Lindsley
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Christopher J Langmead
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Andrew J Lawrence
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
29
|
Nall RW, Heinsbroek JA, Nentwig TB, Kalivas PW, Bobadilla AC. Circuit selectivity in drug versus natural reward seeking behaviors. J Neurochem 2021; 157:1450-1472. [PMID: 33420731 PMCID: PMC8178159 DOI: 10.1111/jnc.15297] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/16/2020] [Accepted: 01/03/2021] [Indexed: 12/23/2022]
Abstract
Substance use disorder (SUD) is characterized, in part by behavior biased toward drug use and away from natural sources of reward (e.g., social interaction, food, sex). The neurobiological underpinnings of SUDs reveal distinct brain regions where neuronal activity is necessary for the manifestation of SUD-characteristic behaviors. Studies that specifically examine how these regions are involved in behaviors motivated by drug versus natural reward allow determinations of which regions are necessary for regulating seeking of both reward types, and appraisals of novel SUD therapies for off-target effects on behaviors motivated by natural reward. Here, we evaluate studies directly comparing regulatory roles for specific brain regions in drug versus natural reward. While it is clear that many regions drive behaviors motivated by all reward types, based on the literature reviewed we propose a set of interconnected regions that become necessary for behaviors motivated by drug, but not natural rewards. The circuitry is selectively necessary for drug seeking includes an Action/Reward subcircuit, comprising nucleus accumbens, ventral pallidum, and ventral tegmental area, a Prefrontal subcircuit comprising prelimbic, infralimbic, and insular cortices, a Stress subcircuit comprising the central nucleus of the amygdala and the bed nucleus of the stria terminalis, and a Diencephalon circuit including lateral hypothalamus. Evidence was mixed for nucleus accumbens shell, insular cortex, and ventral pallidum. Studies for all other brain nuclei reviewed supported a necessary role in regulating both drug and natural reward seeking. Finally, we discuss emerging strategies to further disambiguate the necessity of brain regions in drug- versus natural reward-associated behaviors.
Collapse
Affiliation(s)
- Rusty W. Nall
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Jasper A. Heinsbroek
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Todd B. Nentwig
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Peter W. Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
- These authors share senior authorship
| | - Ana-Clara Bobadilla
- School of Pharmacy, University of Wyoming, Laramie, WY, USA
- These authors share senior authorship
| |
Collapse
|
30
|
Farrell MR, Esteban JSD, Faget L, Floresco SB, Hnasko TS, Mahler SV. Ventral Pallidum GABA Neurons Mediate Motivation Underlying Risky Choice. J Neurosci 2021; 41:4500-4513. [PMID: 33837052 PMCID: PMC8152612 DOI: 10.1523/jneurosci.2039-20.2021] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 02/01/2021] [Accepted: 02/12/2021] [Indexed: 12/29/2022] Open
Abstract
Pursuing rewards while avoiding danger is an essential function of any nervous system. Here, we examine a new mechanism helping rats negotiate the balance between risk and reward when making high-stakes decisions. Specifically, we focus on GABA neurons within an emerging mesolimbic circuit nexus: the ventral pallidum (VP). These neurons play a distinct role from other VP neurons in simple motivated behaviors in mice, but their role in more complex motivated behaviors is unknown. Here, we interrogate the behavioral functions of VPGABA neurons in male and female transgenic GAD1:Cre rats (and WT littermates), using a reversible chemogenetic inhibition approach. Using a behavioral assay of risky decision-making, and of the food-seeking and shock-avoidance components of this task, we show that engaging inhibitory Gi/o signaling specifically in VPGABA neurons suppresses motivation to pursue highly salient palatable foods, and possibly also motivation to avoid being shocked. In contrast, inhibiting these neurons did not affect seeking of low-value food, free consumption of palatable food, or unconditioned affective responses to shock. Accordingly, when rats considered whether to pursue food despite potential for shock in a risky decision-making task, inhibiting VPGABA neurons caused them to more readily select a small but safe reward over a large but dangerous one, an effect not seen in the absence of shock threat. Together, results indicate that VPGABA neurons are critical for high-stakes adaptive responding that is necessary for survival, but which may also malfunction in psychiatric disorders.SIGNIFICANCE STATEMENT In a dynamic world, it is essential to implement appropriate behaviors under circumstances involving rewards, threats, or both. Here, we demonstrate a crucial role for VPGABA neurons in high-stakes motivated behavior of several types. We show that this VPGABA role in motivation impacts decision-making, as inhibiting these neurons yields a conservative, risk-averse strategy not seen when the task is performed without threat of shock. These new roles for VPGABA neurons in behavior may inform future strategies for treating addiction, and other disorders of maladaptive decision-making.
Collapse
Affiliation(s)
- Mitchell R Farrell
- Department of Neurobiology & Behavior, University of California, Irvine, California 92697
| | | | - Lauren Faget
- Department of Neurosciences, University of California, San Diego, California 92093
| | - Stan B Floresco
- Department of Psychology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Thomas S Hnasko
- Department of Neurosciences, University of California, San Diego, California 92093
- VASDHS Research Service, San Diego, California 92161
| | - Stephen V Mahler
- Department of Neurobiology & Behavior, University of California, Irvine, California 92697
| |
Collapse
|
31
|
Bouton ME, Maren S, McNally GP. BEHAVIORAL AND NEUROBIOLOGICAL MECHANISMS OF PAVLOVIAN AND INSTRUMENTAL EXTINCTION LEARNING. Physiol Rev 2021; 101:611-681. [PMID: 32970967 PMCID: PMC8428921 DOI: 10.1152/physrev.00016.2020] [Citation(s) in RCA: 202] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
This article reviews the behavioral neuroscience of extinction, the phenomenon in which a behavior that has been acquired through Pavlovian or instrumental (operant) learning decreases in strength when the outcome that reinforced it is removed. Behavioral research indicates that neither Pavlovian nor operant extinction depends substantially on erasure of the original learning but instead depends on new inhibitory learning that is primarily expressed in the context in which it is learned, as exemplified by the renewal effect. Although the nature of the inhibition may differ in Pavlovian and operant extinction, in either case the decline in responding may depend on both generalization decrement and the correction of prediction error. At the neural level, Pavlovian extinction requires a tripartite neural circuit involving the amygdala, prefrontal cortex, and hippocampus. Synaptic plasticity in the amygdala is essential for extinction learning, and prefrontal cortical inhibition of amygdala neurons encoding fear memories is involved in extinction retrieval. Hippocampal-prefrontal circuits mediate fear relapse phenomena, including renewal. Instrumental extinction involves distinct ensembles in corticostriatal, striatopallidal, and striatohypothalamic circuits as well as their thalamic returns for inhibitory (extinction) and excitatory (renewal and other relapse phenomena) control over operant responding. The field has made significant progress in recent decades, although a fully integrated biobehavioral understanding still awaits.
Collapse
Affiliation(s)
- Mark E Bouton
- Department of Psychological Science, University of Vermont, Burlington, Vermont
| | - Stephen Maren
- Department of Psychological and Brain Sciences and Institute for Neuroscience, Texas A&M University, College Station, Texas
| | - Gavan P McNally
- School of Psychology, University of New South Wales, Sydney, Australia
| |
Collapse
|
32
|
When to Stop Eating: An Auxiliary Brake on Food Consumption from the Nucleus Accumbens. J Neurosci 2021; 41:1847-1849. [PMID: 33658336 DOI: 10.1523/jneurosci.1666-20.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/24/2020] [Accepted: 12/28/2020] [Indexed: 12/17/2022] Open
|
33
|
Buck SA, Torregrossa MM, Logan RW, Freyberg Z. Roles of dopamine and glutamate co-release in the nucleus accumbens in mediating the actions of drugs of abuse. FEBS J 2021; 288:1462-1474. [PMID: 32702182 PMCID: PMC7854787 DOI: 10.1111/febs.15496] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/30/2020] [Accepted: 07/21/2020] [Indexed: 12/20/2022]
Abstract
Projections of ventral tegmental area dopamine (DA) neurons to the medial shell of the nucleus accumbens have been increasingly implicated as integral to the behavioral and physiological changes involved in the development of substance use disorders (SUDs). Recently, many of these nucleus accumbens-projecting DA neurons were found to also release the neurotransmitter glutamate. This glutamate co-release from DA neurons is critical in mediating the effect of drugs of abuse on addiction-related behaviors. Potential mechanisms underlying the role(s) of dopamine/glutamate co-release in contributing to SUDs are unclear. Nevertheless, an important clue may relate to glutamate's ability to potentiate loading of DA into synaptic vesicles within terminals in the nucleus accumbens in response to drug-induced elevations in neuronal activity, enabling a more robust release of DA after stimulation. Here, we summarize how drugs of abuse, particularly cocaine, opioids, and alcohol, alter DA release in the nucleus accumbens medial shell, examine the potential role of DA/glutamate co-release in mediating these effects, and discuss future directions for further investigating these mechanisms.
Collapse
Affiliation(s)
- Silas A. Buck
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mary M. Torregrossa
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ryan W. Logan
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, Bar Harbor, ME, USA
| | - Zachary Freyberg
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
34
|
Motivational competition and the paraventricular thalamus. Neurosci Biobehav Rev 2021; 125:193-207. [PMID: 33609570 DOI: 10.1016/j.neubiorev.2021.02.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 07/16/2020] [Accepted: 02/13/2021] [Indexed: 11/22/2022]
Abstract
Although significant progress has been made in understanding the behavioral and brain mechanisms for motivational systems, much less is known about competition between motivational states or motivational conflict (e.g., approach - avoidance conflict). Despite being produced under diverse conditions, behavior during motivational competition has two signatures: bistability and metastability. These signatures reveal the operation of positive feedback mechanisms in behavioral selection. Different neuronal architectures can instantiate this selection to achieve bistability and metastability in behavior, but each relies on circuit-level inhibition to achieve rapid and stable selection between competing tendencies. Paraventricular thalamus (PVT) is identified as critical to this circuit level inhibition, resolving motivational competition via its extensive projections to local inhibitory networks in the ventral striatum and extended amygdala, enabling adaptive responding under motivational conflict.
Collapse
|
35
|
Liu Y, McNally GP. Dopamine and relapse to drug seeking. J Neurochem 2021; 157:1572-1584. [PMID: 33486769 DOI: 10.1111/jnc.15309] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/04/2021] [Accepted: 01/13/2021] [Indexed: 12/29/2022]
Abstract
The actions of dopamine are essential to relapse to drug seeking but we still lack a precise understanding of how dopamine achieves these effects. Here we review recent advances from animal models in understanding how dopamine controls relapse to drug seeking. These advances have been enabled by important developments in understanding the basic neurochemical, molecular, anatomical, physiological and functional properties of the major dopamine pathways in the mammalian brain. The literature shows that although different forms of relapse to seeking different drugs of abuse each depend on dopamine, there are distinct dopamine mechanisms for relapse. Different circuit-level mechanisms, different populations of dopamine neurons and different activity profiles within these dopamine neurons, are important for driving different forms of relapse. This diversity highlights the need to better understand when, where and how dopamine contributes to relapse behaviours.
Collapse
Affiliation(s)
- Yu Liu
- School of Psychology, UNSW Sydney, Sydney, NSW, Australia
| | | |
Collapse
|
36
|
Schall TA, Wright WJ, Dong Y. Nucleus accumbens fast-spiking interneurons in motivational and addictive behaviors. Mol Psychiatry 2021; 26:234-246. [PMID: 32071384 PMCID: PMC7431371 DOI: 10.1038/s41380-020-0683-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/02/2020] [Accepted: 02/07/2020] [Indexed: 02/07/2023]
Abstract
The development of drug addiction is associated with functional adaptations within the reward circuitry, within which the nucleus accumbens (NAc) is anatomically positioned as an interface between motivational salience and behavioral output. The functional output of NAc is profoundly altered after exposure to drugs of abuse, and some of the functional changes continue to evolve during drug abstinence, contributing to numerous emotional and motivational alterations related drug taking, seeking, and relapse. As in most brain regions, the functional output of NAc is critically dependent on the dynamic interaction between excitation and inhibition. One of the most prominent sources of inhibition within the NAc arises from fast-spiking interneurons (FSIs). Each NAc FSI innervates hundreds of principal neurons, and orchestrates population activity through its powerful and sustained feedforward inhibition. While the role of NAc FSIs in the context of drug addiction remains poorly understood, emerging evidence suggests that FSIs and FSI-mediated local circuits are key targets for drugs of abuse to tilt the functional output of NAc toward a motivational state favoring drug seeking and relapse. In this review, we discuss recent findings and our conceptualization about NAc FSI-mediated regulation of motivated and cocaine-induced behaviors. We hope that the conceptual framework proposed in this review may provide a useful guidance for ongoing and future studies to determine how FSIs influence the function of NAc and related reward circuits, ultimately leading to addictive behaviors.
Collapse
Affiliation(s)
- Terra A Schall
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - William J Wright
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Yan Dong
- Departments of Neuroscience and Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
37
|
Cooperative synaptic and intrinsic plasticity in a disynaptic limbic circuit drive stress-induced anhedonia and passive coping in mice. Mol Psychiatry 2021; 26:1860-1879. [PMID: 32161361 PMCID: PMC7735389 DOI: 10.1038/s41380-020-0686-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 01/19/2020] [Accepted: 02/10/2020] [Indexed: 12/17/2022]
Abstract
Stress promotes negative affective states, which include anhedonia and passive coping. While these features are in part mediated by neuroadaptations in brain reward circuitry, a comprehensive framework of how stress-induced negative affect may be encoded within key nodes of this circuit is lacking. Here, we show in a mouse model for stress-induced anhedonia and passive coping that these phenomena are associated with increased synaptic strength of ventral hippocampus (VH) excitatory synapses onto D1 medium spiny neurons (D1-MSNs) in the nucleus accumbens medial shell (NAcmSh), and with lateral hypothalamus (LH)-projecting D1-MSN hyperexcitability mediated by decreased inwardly rectifying potassium channel (IRK) function. Stress-induced negative affective states are prevented by depotentiation of VH to NAcmSh synapses, restoring Kir2.1 function in D1R-MSNs, or disrupting co-participation of these synaptic and intrinsic adaptations in D1-MSNs. In conclusion, our data provide strong evidence for a disynaptic pathway controlling maladaptive emotional behavior.
Collapse
|
38
|
Wright WJ, Dong Y. Psychostimulant-Induced Adaptations in Nucleus Accumbens Glutamatergic Transmission. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a039255. [PMID: 31964644 DOI: 10.1101/cshperspect.a039255] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Carrying different aspects of emotional and motivational signals, glutamatergic synaptic projections from multiple limbic and paralimbic brain regions converge to the nucleus accumbens (NAc), in which these arousing signals are processed and prioritized for behavioral output. In animal models of drug addiction, some key drug-induced alterations at NAc glutamatergic synapses underlie important cellular and circuit mechanisms that promote subsequent drug taking, seeking, and relapse. With the focus of cocaine, we review changes at NAc glutamatergic synapses that occur after different drug procedures and abstinence durations, and the behavioral impact of these changes.
Collapse
Affiliation(s)
- William J Wright
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA
| |
Collapse
|
39
|
Campbell EJ, Hill MK, Maddern XJ, Jin S, Pang TY, Lawrence AJ. Orexin-1 receptor signaling within the lateral hypothalamus, but not bed nucleus of the stria terminalis, mediates context-induced relapse to alcohol seeking. J Psychopharmacol 2020; 34:1261-1270. [PMID: 33063594 DOI: 10.1177/0269881120959638] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The lateral hypothalamic orexin (hypocretin) system has a well-established role in the motivation for reward. This has particular relevance to substance use disorders since orexin-1 receptors play a critical role in alcohol-seeking behavior, acting at multiple nodes in relapse-associated networks. AIMS This study aimed to further our understanding of the role of orexin-1 receptor signaling within the lateral hypothalamus and bed nucleus of the stria terminalis, specifically in context-induced relapse to alcohol-seeking following punishment-imposed abstinence. METHODS We trained inbred male alcohol-preferring rats to self-administer alcohol in one environment or context (Context A) and subsequently punished their alcohol-reinforced lever presses in a different environment (Context B) using contingent foot shock punishment. Finally, we tested rats for relapse-like behavior in either context following systemic, intra-lateral hypothalamus or intra-bed nucleus of the stria terminalis orexin-1 receptor antagonism with SB-334867. RESULTS/OUTCOMES We found that systemic orexin-1 receptor antagonism significantly reduced alcohol-seeking in both contexts. Intra-lateral hypothalamus orexin-1 receptor antagonism significantly reduced alcohol-seeking in Context A whereas intra-bed nucleus of the stria terminalis orexin-1 receptor antagonism had no effect on alcohol-seeking behavior. CONCLUSIONS/INTERPRETATION Our results suggest a role for the orexin-1 receptor system in context-induced relapse to alcohol-seeking. Specifically, intra-lateral hypothalamus orexin microcircuits contribute to alcohol-seeking.
Collapse
Affiliation(s)
- Erin J Campbell
- Florey Institute of Neuroscience and Mental Health and Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Mitchell Kri Hill
- Florey Institute of Neuroscience and Mental Health and Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Xavier J Maddern
- Florey Institute of Neuroscience and Mental Health and Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Shubo Jin
- Florey Institute of Neuroscience and Mental Health and Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Terence Y Pang
- Florey Institute of Neuroscience and Mental Health and Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Andrew J Lawrence
- Florey Institute of Neuroscience and Mental Health and Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3052, Australia
| |
Collapse
|
40
|
Wang X, Tian Z, Ma J, Feng Z, Ou Y, Zhou M, Peng J, Lv Y, Gao G, Qi S. NPY alterations induced by chronic morphine exposure affect the maintenance and reinstatement of morphine conditioned place preference. Neuropharmacology 2020; 181:108350. [PMID: 33027625 DOI: 10.1016/j.neuropharm.2020.108350] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/28/2020] [Accepted: 10/02/2020] [Indexed: 01/23/2023]
Abstract
Opioid addiction is a brain disease that severely harms society and personal health. Although the tremendous numbers of patients worldwide and emerged negative events, effective treatments for opioid addiction are still lacking. Neuropeptide Y (NPY) is one of the main orexigenic peptides that play vital roles in food intake and energy metabolism. However, increasing evidence indicates that NPY may have great potential in mediating reward effects and drug dependence. In the present study, we assessed the expression changes of NPY in the nucleus accumbens at different timepoints following morphine conditioned place preference (CPP) and investigated the functional importance of potential NPY changes. Our results showed that NPY expression significantly decreased in the nucleus accumbens shell (AcbSh) immediately after chronic morphine exposure. Subsequently, it increased rapidly at first and then gradually returned to normal levels. Further data indicated that these NPY changes were involved in morphine reward memory, demonstrated by a reduction in the extinction period after blocking of the Y5 receptor by L-152,804 in the AcbSh and a prolonged duration of the extinction period following the application of NPY. More importantly, the additional results revealed that L-152,804 also remarkably suppressed the reinstatement of morphine CPP. Together, our results indicate that a complicated plasticity of the NPY pathway in AcbSh occurs following morphine CPP, and this plasticity plays an important role in modulating morphine reward memory. These findings may enhance our understanding of the role of the NPY system in opioid addiction and indicate a promising target for opioid addiction treatment.
Collapse
Affiliation(s)
- Xingqin Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhen Tian
- Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China; Department of Pharmacy, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, Guangzhou, China
| | - Jie Ma
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Zhanpeng Feng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yichao Ou
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Mingfeng Zhou
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Junjie Peng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yunfei Lv
- Department of Anesthesiology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Guodong Gao
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China.
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
41
|
Piantadosi PT, Yeates DCM, Floresco SB. Prefrontal cortical and nucleus accumbens contributions to discriminative conditioned suppression of reward-seeking. ACTA ACUST UNITED AC 2020; 27:429-440. [PMID: 32934096 PMCID: PMC7497111 DOI: 10.1101/lm.051912.120] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/02/2020] [Indexed: 12/18/2022]
Abstract
Fear can potently inhibit ongoing behavior, including reward-seeking, yet the neural circuits that underlie such suppression remain to be clarified. Prior studies have demonstrated that distinct subregions of the rodent medial prefrontal cortex (mPFC) differentially affect fear behavior, whereby fear expression is promoted by the more dorsal prelimbic cortex (PL) and inhibited by the more ventral infralimbic cortex (IL). These mPFC regions project to subregions of the nucleus accumbens, the core (NAcC) and shell (NAcS), that differentially contribute to reward-seeking as well as affective processes that may be relevant to fear expression. Here, we investigated how these mPFC and NAc subregions contribute to discriminative fear conditioning, assessed by conditioned suppression of reward-seeking. Bilateral inactivation of the NAcS or PL reduced the expression of conditioned suppression to a shock-associated CS+, whereas NAcC inactivation reduced reward-seeking without affecting suppression. IL inactivation caused a general reduction in conditioned suppression following discriminative conditioning, but not when using a single-stimulus design. Pharmacological disconnection of the PL → NAcS pathway revealed that this projection mediates conditioned suppression. These data add to a growing literature implicating discrete cortico-striatal pathways in the suppression of reward-seeking in response to aversive stimuli. Dysfunction within related structures may contribute to aberrant patterns of behavior in psychiatric illnesses including substance use disorders.
Collapse
Affiliation(s)
- Patrick T Piantadosi
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Dylan C M Yeates
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Stan B Floresco
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| |
Collapse
|
42
|
Liu Y, Jean-Richard-Dit-Bressel P, Yau JOY, Willing A, Prasad AA, Power JM, Killcross S, Clifford CWG, McNally GP. The Mesolimbic Dopamine Activity Signatures of Relapse to Alcohol-Seeking. J Neurosci 2020; 40:6409-6427. [PMID: 32669355 PMCID: PMC7424877 DOI: 10.1523/jneurosci.0724-20.2020] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 06/08/2020] [Accepted: 07/06/2020] [Indexed: 12/17/2022] Open
Abstract
The mesolimbic dopamine system comprises distinct compartments supporting different functions in learning and motivation. Less well understood is how complex addiction-related behaviors emerge from activity patterns across these compartments. Here we show how different forms of relapse to alcohol-seeking in male rats are assembled from activity across the VTA and the nucleus accumbens. First, we used chemogenetic approaches to show a causal role for VTA TH neurons in two forms of relapse to alcohol-seeking: renewal (context-induced reinstatement) and reacquisition. Then, using gCaMP fiber photometry of VTA TH neurons, we identified medial and lateral VTA TH neuron activity profiles during self-administration, renewal, and reacquisition. Next, we used optogenetic inhibition of VTA TH neurons to show distinct causal roles for VTA subregions in distinct forms of relapse. We then used dLight fiber photometry to measure dopamine binding across the ventral striatum (medial accumbens shell, accumbens core, lateral accumbens shell) and showed complex and heterogeneous profiles of dopamine binding during self-administration and relapse. Finally, we used representational similarity analysis to identify mesolimbic dopamine signatures of self-administration, extinction, and relapse. Our results show that signatures of relapse can be identified from heterogeneous activity profiles across the mesolimbic dopamine system and that these signatures are unique for different forms of relapse.SIGNIFICANCE STATEMENT It is axiomatic that the actions of dopamine are critical to drug addiction. Yet how relapse to drug-seeking is assembled from activity across the mesolimbic dopamine system is poorly understood. Here we show how relapse to alcohol-seeking relates to activity in specific VTA and accumbens compartments, how these change for different forms of relapse, and how relapse-associated activity relates to activity during self-administration and extinction. We report the mesolimbic dopamine activity signatures for relapse and show that these signatures are unique for different forms of relapse.
Collapse
Affiliation(s)
- Yu Liu
- School of Psychology, University of New South Wales Sydney, Sydney, New South Wales 2052, Australia
| | | | - Joanna Oi-Yue Yau
- School of Psychology, University of New South Wales Sydney, Sydney, New South Wales 2052, Australia
| | - Alexandra Willing
- School of Psychology, University of New South Wales Sydney, Sydney, New South Wales 2052, Australia
| | - Asheeta A Prasad
- School of Psychology, University of New South Wales Sydney, Sydney, New South Wales 2052, Australia
| | - John M Power
- Department of Physiology and Translational Neuroscience Facility, School of Medical Sciences, University of New South Wales Sydney, Sydney, New South Wales 2052, Australia
| | - Simon Killcross
- School of Psychology, University of New South Wales Sydney, Sydney, New South Wales 2052, Australia
| | - Colin W G Clifford
- School of Psychology, University of New South Wales Sydney, Sydney, New South Wales 2052, Australia
| | - Gavan P McNally
- School of Psychology, University of New South Wales Sydney, Sydney, New South Wales 2052, Australia
| |
Collapse
|
43
|
Keyes PC, Adams EL, Chen Z, Bi L, Nachtrab G, Wang VJ, Tessier-Lavigne M, Zhu Y, Chen X. Orchestrating Opiate-Associated Memories in Thalamic Circuits. Neuron 2020; 107:1113-1123.e4. [PMID: 32679036 DOI: 10.1016/j.neuron.2020.06.028] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 05/28/2020] [Accepted: 06/23/2020] [Indexed: 02/06/2023]
Abstract
Disrupting memories that associate environmental cues with drug experiences holds promise for treating addiction, yet accessing the distributed neural network that stores such memories is challenging. Here, we show that the paraventricular nucleus of the thalamus (PVT) orchestrates the acquisition and maintenance of opiate-associated memories via projections to the central nucleus of the amygdala (CeA) and nucleus accumbens (NAc). PVT→CeA activity associates morphine reward to the environment, whereas transient inhibition of the PVT→NAc pathway during retrieval causes enduring protection against opiate-primed relapse. Using brain-wide activity mapping, we revealed distributed network activities that are altered in non-relapsing mice, which enabled us to find that activating the downstream NAc→lateral hypothalamus (LH) pathway also prevents relapse. These findings establish the PVT as a key node in the opiate-associated memory network and demonstrate the potential of targeting the PVT→NAc→LH pathway for treating opioid addiction.
Collapse
Affiliation(s)
- Piper C Keyes
- Neurosciences Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Eliza L Adams
- Neurosciences Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Zijun Chen
- Shenzhen Key Laboratory of Drug Addiction, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Linlin Bi
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Gregory Nachtrab
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Vickie J Wang
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | | | - Yingjie Zhu
- Shenzhen Key Laboratory of Drug Addiction, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China; Department of Biology, Stanford University, Stanford, CA 94305, USA.
| | - Xiaoke Chen
- Neurosciences Graduate Program, Stanford University, Stanford, CA 94305, USA; Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
44
|
Prasad AA, McNally GP. The ventral pallidum and relapse in alcohol seeking. Br J Pharmacol 2020; 177:3855-3864. [PMID: 32557550 DOI: 10.1111/bph.15160] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/29/2020] [Accepted: 06/05/2020] [Indexed: 12/13/2022] Open
Abstract
Alcohol-use disorders are chronically relapsing conditions characterized by cycles of use, abstinence and relapse. The ventral pallidum (VP) is a key node in the neural circuits controlling relapse to alcohol seeking and a key target of pharmacotherapies for relapse prevention. There has been a significant increase in our understanding of the molecular, anatomical, pharmacological and functional properties of the ventral pallidum, laying foundations for a new understanding of its role in relapse to alcohol seeking and motivation. Here we review these advances, placing special emphasis on how advances in understanding in the cellular and circuit architectures of ventral pallidum contributes to the relapse to alcohol seeking. We show how this knowledge improves mechanistic understanding of current relapse prevention pharmacotherapies, how it may be used to tailor these against different forms of relapse and how it may help provide insights into the mental health problems frequently co-morbid with alcohol-use disorders.
Collapse
|
45
|
Thoeni S, Loureiro M, O'Connor EC, Lüscher C. Depression of Accumbal to Lateral Hypothalamic Synapses Gates Overeating. Neuron 2020; 107:158-172.e4. [PMID: 32333845 PMCID: PMC7616964 DOI: 10.1016/j.neuron.2020.03.029] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 02/21/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023]
Abstract
Overeating typically follows periods of energy deficit, but it is also sustained by highly palatable foods, even without metabolic demand. Dopamine D1 receptor-expressing medium spiny neurons (D1-MSNs) of the nucleus accumbens shell (NAcSh) project to the lateral hypothalamus (LH) to authorize feeding when inhibited. Whether plasticity at these synapses can affect food intake is unknown. Here, ex vivo electrophysiology recordings reveal that D1-MSN-to-LH inhibitory transmission is depressed in circumstances in which overeating is promoted. Endocannabinoid signaling is identified as the induction mechanism, since inhibitory plasticity and concomitant overeating were blocked or induced by CB1R antagonism or agonism, respectively. D1-MSN-to-LH projectors were largely non-overlapping with D1-MSNs targeting ventral pallidum or ventral midbrain, providing an anatomical basis for distinct circuit plasticity mechanisms. Our study reveals a critical role for plasticity at D1-MSN-to-LH synapses in adaptive feeding control, which may underlie persistent overeating of unhealthy foods, a major risk factor for developing obesity.
Collapse
Affiliation(s)
- Sarah Thoeni
- Department of Basic Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Michaël Loureiro
- Department of Basic Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Eoin C O'Connor
- Department of Basic Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Christian Lüscher
- Department of Basic Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Clinic of Neurology, Department of Clinical Neurosciences, Geneva University Hospital, Geneva, Switzerland.
| |
Collapse
|
46
|
Halladay LR, Kocharian A, Piantadosi PT, Authement ME, Lieberman AG, Spitz NA, Coden K, Glover LR, Costa VD, Alvarez VA, Holmes A. Prefrontal Regulation of Punished Ethanol Self-administration. Biol Psychiatry 2020; 87:967-978. [PMID: 31937415 PMCID: PMC7217757 DOI: 10.1016/j.biopsych.2019.10.030] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 10/08/2019] [Accepted: 10/25/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND A clinical hallmark of alcohol use disorder is persistent drinking despite potential adverse consequences. The ventromedial prefrontal cortex (vmPFC) and dorsomedial prefrontal cortex (dmPFC) are positioned to exert top-down control over subcortical regions, such as the nucleus accumbens shell (NAcS) and basolateral amygdala, which encode positive and negative valence of ethanol (EtOH)-related stimuli. Prior rodent studies have implicated these regions in regulation of punished EtOH self-administration (EtOH-SA). METHODS We conducted in vivo electrophysiological recordings in mouse vmPFC and dmPFC to obtain neuronal correlates of footshock-punished EtOH-SA. Ex vivo recordings were performed in NAcS D1 receptor-expressing medium spiny neurons receiving vmPFC input to examine punishment-related plasticity in this pathway. Optogenetic photosilencing was employed to assess the functional contribution of the vmPFC, dmPFC, vmPFC projections to NAcS, or vmPFC projections to basolateral amygdala, to punished EtOH-SA. RESULTS Punishment reduced EtOH lever pressing and elicited aborted presses (lever approach followed by rapid retraction). Neurons in the vmPFC and dmPFC exhibited phasic firing to EtOH lever presses and aborts, but only in the vmPFC was there a population-level shift in coding from lever presses to aborts with punishment. Closed-loop vmPFC, but not dmPFC, photosilencing on a postpunishment probe test negated the reduction in EtOH lever presses but not in aborts. Punishment was associated with altered plasticity at vmPFC inputs to D1 receptor-expressing medium spiny neurons in the NAcS. Photosilencing vmPFC projections to the NAcS, but not to the basolateral amygdala, partially reversed suppression of EtOH lever presses on probe testing. CONCLUSIONS These findings demonstrate a key role for the vmPFC in regulating EtOH-SA after punishment, with implications for understanding the neural basis of compulsive drinking in alcohol use disorder.
Collapse
Affiliation(s)
- Lindsay R Halladay
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Department of Psychology, Santa Clara University, Santa Clara, California.
| | - Adrina Kocharian
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Patrick T Piantadosi
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Center on Compulsive Behaviors, Intramural Research Program, National Institutes of Health, Bethesda, Maryland
| | - Michael E Authement
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Center on Compulsive Behaviors, Intramural Research Program, National Institutes of Health, Bethesda, Maryland
| | - Abby G Lieberman
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Nathen A Spitz
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Kendall Coden
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Lucas R Glover
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Vincent D Costa
- Department of Behavioral Neuroscience, Oregon Health Sciences University, Portland, Oregon
| | - Veronica A Alvarez
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Center on Compulsive Behaviors, Intramural Research Program, National Institutes of Health, Bethesda, Maryland
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
47
|
Medial Nucleus Accumbens Projections to the Ventral Tegmental Area Control Food Consumption. J Neurosci 2020; 40:4727-4738. [PMID: 32354856 DOI: 10.1523/jneurosci.3054-18.2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 04/15/2020] [Accepted: 04/22/2020] [Indexed: 12/12/2022] Open
Abstract
Decades of research have shown that the NAc is a critical region influencing addiction, mood, and food consumption through its effects on reinforcement learning, motivation, and hedonic experience. Pharmacological studies have demonstrated that inhibition of the NAc shell induces voracious feeding, leading to the hypothesis that the inhibitory projections that emerge from the NAc normally act to restrict feeding. While much of this work has focused on projections to the lateral hypothalamus, the role of NAc projections to the VTA in the control food intake has been largely unexplored. Using a retrograde viral labeling technique and real-time monitoring of neural activity with fiber photometry, we find that medial NAc shell projections to the VTA (mNAc→VTA) are inhibited during food-seeking and food consumption in male mice. We also demonstrate that this circuit bidirectionally controls feeding: optogenetic activation of NAc projections to the VTA inhibits food-seeking and food intake (in both sexes), while optogenetic inhibition of this circuit potentiates food-seeking behavior. Additionally, we show that activity of the NAc to VTA pathway is necessary for adaptive inhibition of food intake in response to external cues. These data provide new insight into NAc control over feeding in mice, and contribute to an emerging literature elucidating the role of inhibitory midbrain feedback within the mesolimbic circuit.SIGNIFICANCE STATEMENT The medial NAc has long been known to control consummatory behavior, with particular focus on accumbens projections to the lateral hypothalamus. Conversely, NAc projections to the VTA have mainly been studied in the context of drug reward. We show that NAc projections to the VTA bidirectionally control food intake, consistent with a permissive role in feeding. Additionally, we show that this circuit is normally inactivated during consumption and food-seeking. Together, these findings elucidate how mesolimbic circuits control food consumption.
Collapse
|
48
|
Prasad AA, Xie C, Chaichim C, Nguyen JH, McClusky HE, Killcross S, Power JM, McNally GP. Complementary Roles for Ventral Pallidum Cell Types and Their Projections in Relapse. J Neurosci 2020; 40:880-893. [PMID: 31818977 PMCID: PMC6975293 DOI: 10.1523/jneurosci.0262-19.2019] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 11/29/2019] [Accepted: 12/04/2019] [Indexed: 02/06/2023] Open
Abstract
The ventral pallidum (VP) is a key node in the neural circuits controlling relapse to drug seeking. How this role relates to different VP cell types and their projections is poorly understood. Using male rats, we show how different forms of relapse to alcohol-seeking are assembled from VP cell types and their projections to lateral hypothalamus (LH) and ventral tegmental area (VTA). Using RNAScope in situ hybridization to characterize activity of different VP cell types during relapse to alcohol-seeking provoked by renewal (context-induced reinstatement), we found that VP Gad1 and parvalbumin (PV), but not vGlut2, neurons show relapse-associated changes in c-Fos expression. Next, we used retrograde tracing, chemogenetic, and electrophysiological approaches to study the roles of VPGad1 and VPPV neurons in relapse. We show that VPGad1 neurons contribute to contextual control over relapse (renewal), but not to relapse during reacquisition, via projections to LH, where they converge with ventral striatal inputs onto LHGad1 neurons. This convergence of striatopallidal inputs at the level of individual LHGad1 neurons may be critical to balancing propensity for relapse versus abstinence. In contrast, VPPV neurons contribute to relapse during both renewal and reacquisition via projections to VTA. These findings identify a double dissociation in the roles for different VP cell types and their projections in relapse. VPGad1 neurons control relapse during renewal via projections to LH. VPPV neurons control relapse during both renewal and reacquisition via projections to VTA. Targeting these different pathways may provide tailored interventions for different forms of relapse.SIGNIFICANCE STATEMENT Relapse to drug or reward seeking after a period of extinction or abstinence remains a key impediment to successful treatment. The ventral pallidum, located in the ventral basal ganglia, has long been recognized as an obligatory node in a 'final common pathway' for relapse. Yet how this role relates to the considerable VP cellular and circuit heterogeneity is not well understood. We studied the cellular and circuit architecture for VP in relapse control. We show that different forms of relapse have complementary VP cellular and circuit architectures, raising the possibility that targeting these different neural architectures may provide tailored interventions for different forms of relapse.
Collapse
Affiliation(s)
| | | | - Chanchanok Chaichim
- Department of Physiology and Translational Neuroscience Facility, School of Medical Sciences, University of New South Wales Sydney (UNSW), Sydney, New South Wales 2052, Australia
| | | | | | | | - John M Power
- Department of Physiology and Translational Neuroscience Facility, School of Medical Sciences, University of New South Wales Sydney (UNSW), Sydney, New South Wales 2052, Australia
| | | |
Collapse
|
49
|
Perry CJ, Lawrence AJ. An imperfect model is still useful. Addiction 2020; 115:14-16. [PMID: 31496025 DOI: 10.1111/add.14790] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 08/16/2019] [Indexed: 11/29/2022]
Affiliation(s)
- Christina J Perry
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Andrew J Lawrence
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
50
|
Visualizing Infralimbic Control over Incubation of Cocaine Craving. Neuron 2019; 103:1-3. [PMID: 31271753 DOI: 10.1016/j.neuron.2019.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The infralimbic prefrontal cortex serves complex roles in controlling drug-seeking behavior. In this issue of Neuron, Cameron et al. (2019) provide new insights into engagement and function of the infralimbic cortex → nucleus accumbens corticostriatal pathway in the incubation of cocaine craving.
Collapse
|