1
|
Schultz W. A dopamine mechanism for reward maximization. Proc Natl Acad Sci U S A 2024; 121:e2316658121. [PMID: 38717856 PMCID: PMC11098095 DOI: 10.1073/pnas.2316658121] [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: 05/18/2024] Open
Abstract
Individual survival and evolutionary selection require biological organisms to maximize reward. Economic choice theories define the necessary and sufficient conditions, and neuronal signals of decision variables provide mechanistic explanations. Reinforcement learning (RL) formalisms use predictions, actions, and policies to maximize reward. Midbrain dopamine neurons code reward prediction errors (RPE) of subjective reward value suitable for RL. Electrical and optogenetic self-stimulation experiments demonstrate that monkeys and rodents repeat behaviors that result in dopamine excitation. Dopamine excitations reflect positive RPEs that increase reward predictions via RL; against increasing predictions, obtaining similar dopamine RPE signals again requires better rewards than before. The positive RPEs drive predictions higher again and thus advance a recursive reward-RPE-prediction iteration toward better and better rewards. Agents also avoid dopamine inhibitions that lower reward prediction via RL, which allows smaller rewards than before to elicit positive dopamine RPE signals and resume the iteration toward better rewards. In this way, dopamine RPE signals serve a causal mechanism that attracts agents via RL to the best rewards. The mechanism improves daily life and benefits evolutionary selection but may also induce restlessness and greed.
Collapse
Affiliation(s)
- Wolfram Schultz
- Department of Physiology, Development and Neuroscience, University of Cambridge, CambridgeCB2 3DY, United Kingdom
| |
Collapse
|
2
|
Donlon J, Kumari P, Varghese SP, Bai M, Florentin OD, Frost ED, Banks J, Vadlapatla N, Kam O, Shad MU, Rahman S, Abulseoud OA, Stone TW, Koola MM. Integrative Pharmacology in the Treatment of Substance Use Disorders. J Dual Diagn 2024; 20:132-177. [PMID: 38117676 DOI: 10.1080/15504263.2023.2293854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
The detrimental physical, mental, and socioeconomic effects of substance use disorders (SUDs) have been apparent to the medical community for decades. However, it has become increasingly urgent in recent years to develop novel pharmacotherapies to treat SUDs. Currently, practitioners typically rely on monotherapy. Monotherapy has been shown to be superior to no treatment at all for most substance classes. However, many randomized controlled trials (RCTs) have revealed that monotherapy leads to poorer outcomes when compared with combination treatment in all specialties of medicine. The results of RCTs suggest that monotherapy frequently fails since multiple dysregulated pathways, enzymes, neurotransmitters, and receptors are involved in the pathophysiology of SUDs. As such, research is urgently needed to determine how various neurobiological mechanisms can be targeted by novel combination treatments to create increasingly specific yet exceedingly comprehensive approaches to SUD treatment. This article aims to review the neurobiology that integrates many pathophysiologic mechanisms and discuss integrative pharmacology developments that may ultimately improve clinical outcomes for patients with SUDs. Many neurobiological mechanisms are known to be involved in SUDs including dopaminergic, nicotinic, N-methyl-D-aspartate (NMDA), and kynurenic acid (KYNA) mechanisms. Emerging evidence indicates that KYNA, a tryptophan metabolite, modulates all these major pathophysiologic mechanisms. Therefore, achieving KYNA homeostasis by harmonizing integrative pathophysiology and pharmacology could prove to be a better therapeutic approach for SUDs. We propose KYNA-NMDA-α7nAChRcentric pathophysiology, the "conductor of the orchestra," as a novel approach to treat many SUDs concurrently. KYNA-NMDA-α7nAChR pathophysiology may be the "command center" of neuropsychiatry. To date, extant RCTs have shown equivocal findings across comparison conditions, possibly because investigators targeted single pathophysiologic mechanisms, hit wrong targets in underlying pathophysiologic mechanisms, and tested inadequate monotherapy treatment. We provide examples of potential combination treatments that simultaneously target multiple pathophysiologic mechanisms in addition to KYNA. Kynurenine pathway metabolism demonstrates the greatest potential as a target for neuropsychiatric diseases. The investigational medications with the most evidence include memantine, galantamine, and N-acetylcysteine. Future RCTs are warranted with novel combination treatments for SUDs. Multicenter RCTs with integrative pharmacology offer a promising, potentially fruitful avenue to develop novel therapeutics for the treatment of SUDs.
Collapse
Affiliation(s)
- Jack Donlon
- Cooper Medical School of Rowan University, Camden, New Jersey, USA
| | - Pooja Kumari
- Community Living Trent Highlands, Peterborough, Canada
| | - Sajoy P Varghese
- Addiction Recovery Treatment Services, Veterans Affairs Northern California Health Care System, University of California, Davis, Sacramento, California, USA
| | - Michael Bai
- Columbia University, New York, New York, USA
| | - Ori David Florentin
- Department of Psychiatry, Westchester Medical Center, Valhalla, New York, USA
| | - Emma D Frost
- Department of Neurology, Cooper University Health Care, Camden, New Jersey, USA
| | - John Banks
- Talkiatry Mental Health Clinic, New York, New York, USA
| | - Niyathi Vadlapatla
- Thomas Jefferson High School for Science and Technology, Alexandria, Virginia, USA
| | - Olivia Kam
- Stony Brook University Renaissance School of Medicine, Stony Brook, New York, USA
| | - Mujeeb U Shad
- Department of Psychiatry, University of Nevada Las Vegas, Las Vegas, Nevada, USA
| | - Shafiqur Rahman
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, South Dakota, USA
| | - Osama A Abulseoud
- Department of Psychiatry and Psychology, Alix School of Medicine at Mayo Clinic, Phoenix, Arizona, USA
| | - Trevor W Stone
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, UK
| | - Maju Mathew Koola
- Department of Psychiatry and Behavioral Health, Cooper University Health Care, Cooper Medical School of Rowan University, Camden, New Jersey, USA
| |
Collapse
|
3
|
Seiler JL, Zhuang X, Nelson AB, Lerner TN. Dopamine across timescales and cell types: Relevance for phenotypes in Parkinson's disease progression. Exp Neurol 2024; 374:114693. [PMID: 38242300 DOI: 10.1016/j.expneurol.2024.114693] [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: 09/12/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 01/21/2024]
Abstract
Dopamine neurons in the substantia nigra pars compacta (SNc) synthesize and release dopamine, a critical neurotransmitter for movement and learning. SNc dopamine neurons degenerate in Parkinson's Disease (PD), causing a host of motor and non-motor symptoms. Here, we review recent conceptual advances in our basic understanding of the dopamine system - including our rapidly advancing knowledge of dopamine neuron heterogeneity - with special attention to their importance for understanding PD. In PD patients, dopamine neuron degeneration progresses from lateral SNc to medial SNc, suggesting clinically relevant heterogeneity in dopamine neurons. With technical advances in dopamine system interrogation, we can understand the relevance of this heterogeneity for PD progression and harness it to develop new treatments.
Collapse
Affiliation(s)
- Jillian L Seiler
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Xiaowen Zhuang
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA; Weill Institute for Neuroscience, University of California San Francisco, San Francisco, CA, USA; Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Alexandra B Nelson
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA; Weill Institute for Neuroscience, University of California San Francisco, San Francisco, CA, USA; Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Talia N Lerner
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Northwestern University Interdepartmental Neuroscience Program (NUIN), Evanston, IL, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
4
|
Kuś J, Saramowicz K, Czerniawska M, Wiese W, Siwecka N, Rozpędek-Kamińska W, Kucharska-Lusina A, Strzelecki D, Majsterek I. Molecular Mechanisms Underlying NMDARs Dysfunction and Their Role in ADHD Pathogenesis. Int J Mol Sci 2023; 24:12983. [PMID: 37629164 PMCID: PMC10454781 DOI: 10.3390/ijms241612983] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is one of the most common neurodevelopmental disorders, although the aetiology of ADHD is not yet understood. One proposed theory for developing ADHD is N-methyl-D-aspartate receptors (NMDARs) dysfunction. NMDARs are involved in regulating synaptic plasticity and memory function in the brain. Abnormal expression or polymorphism of some genes associated with ADHD results in NMDAR dysfunction. Correspondingly, NMDAR malfunction in animal models results in ADHD-like symptoms, such as impulsivity and hyperactivity. Currently, there are no drugs for ADHD that specifically target NMDARs. However, NMDAR-stabilizing drugs have shown promise in improving ADHD symptoms with fewer side effects than the currently most widely used psychostimulant in ADHD treatment, methylphenidate. In this review, we outline the molecular and genetic basis of NMDAR malfunction and how it affects the course of ADHD. We also present new therapeutic options related to treating ADHD by targeting NMDAR.
Collapse
Affiliation(s)
- Justyna Kuś
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Kamil Saramowicz
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Maria Czerniawska
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Wojciech Wiese
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Natalia Siwecka
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Wioletta Rozpędek-Kamińska
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Aleksandra Kucharska-Lusina
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Dominik Strzelecki
- Department of Affective and Psychotic Disorders, Medical University of Lodz, Czechoslowacka 8/10, 92-216 Lodz, Poland;
| | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| |
Collapse
|
5
|
Fusaroli M, Giunchi V, Battini V, Gringeri M, Rimondini R, Menchetti M, Radice S, Pozzi M, Nobile M, Clementi E, De Ponti F, Carnovale C, Raschi E, Poluzzi E. Exploring the underlying mechanisms of drug-induced impulse control disorders: a pharmacovigilance-pharmacodynamic study. Psychiatry Clin Neurosci 2023; 77:160-167. [PMID: 36436204 DOI: 10.1111/pcn.13511] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 11/07/2022] [Accepted: 11/18/2022] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Impulse control disorders (e.g. pathological gambling, hypersexuality) may develop as adverse reactions to drugs. Pathogenetic hypotheses have mainly focused on D3-receptor agonism, and switching to alternatives with different pharmacologic mechanisms represents a common management strategy. Nonetheless, treatment failure is common and gaining pathophysiological insights is needed. AIM We aimed to identify targets potentially contributing to pathologic impulsivity. METHOD We performed a pharmacovigilance-pharmacodynamic study on dopamine agonists and antipsychotics using the Food and Drug Administration Adverse Event Reporting System (January 2004-December 2021). We estimated disproportionate reporting using the Bayesian information component. Using online public databases (IUPHAR, ChEMBL, PDSP, DrugBank), we calculated drug occupancies. To identify the targets potentially contributing to impulsivity, we fitted univariate regression models interpolating information components and occupancies within dopamine agonists and antipsychotics. Sensitivity analyses were performed to check for the robustness of the results. RESULTS Among 19 887 reports of impulsivity, 5898 recorded an antipsychotic, and 3100 a dopamine agonist. The more robust signals concerned aripiprazole (N = 3091; median information component [95% confidence interval] = 4.51[4.45-4.55]) and brexpiprazole (229; 4.00[3.78-4.16]) for antipsychotics, pergolide (105; 5.82[5.50-6.06]) and pramipexole (2009; 5.43[5.36-5.48]) for dopamine agonists. Robust, significant positive associations between drug occupancy and impulsivity reporting were found for D3 within dopamine agonists (beta = 1.52; P-value = 0.047) and 5-HT1a within antipsychotics (1.92, 0.029). CONCLUSION Our results supported the role of D3-receptor agonism in inducing impulsivity in dopamine receptor agonists and identified a potential role of 5-HT1a receptor agonism in antipsychotics. Investigating these receptors may drive towards a better management of drug-induced impulsivity.
Collapse
Affiliation(s)
- Michele Fusaroli
- Pharmacology Unit, Department of Medical and Surgical Sciences (DIMEC), Università di Bologna, Bologna, Italy
| | - Valentina Giunchi
- Pharmacology Unit, Department of Medical and Surgical Sciences (DIMEC), Università di Bologna, Bologna, Italy
| | - Vera Battini
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences (DIBIC), ASST Fatebenefratelli-Sacco University Hospital, Università degli Studi di Milano, Milan, Italy
| | - Michele Gringeri
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences (DIBIC), ASST Fatebenefratelli-Sacco University Hospital, Università degli Studi di Milano, Milan, Italy
| | - Roberto Rimondini
- Pharmacology Unit, Department of Medical and Surgical Sciences (DIMEC), Università di Bologna, Bologna, Italy
| | - Marco Menchetti
- Unit of Psychiatry, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna
| | - Sonia Radice
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences (DIBIC), ASST Fatebenefratelli-Sacco University Hospital, Università degli Studi di Milano, Milan, Italy
| | - Marco Pozzi
- Scientific Institute IRCCS Eugenio Medea, Bosisio Parini (LC), Italy
| | - Maria Nobile
- Scientific Institute IRCCS Eugenio Medea, Bosisio Parini (LC), Italy
| | - Emilio Clementi
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences (DIBIC), ASST Fatebenefratelli-Sacco University Hospital, Università degli Studi di Milano, Milan, Italy.,Scientific Institute IRCCS Eugenio Medea, Bosisio Parini (LC), Italy
| | - Fabrizio De Ponti
- Pharmacology Unit, Department of Medical and Surgical Sciences (DIMEC), Università di Bologna, Bologna, Italy
| | - Carla Carnovale
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences (DIBIC), ASST Fatebenefratelli-Sacco University Hospital, Università degli Studi di Milano, Milan, Italy
| | - Emanuel Raschi
- Pharmacology Unit, Department of Medical and Surgical Sciences (DIMEC), Università di Bologna, Bologna, Italy
| | - Elisabetta Poluzzi
- Pharmacology Unit, Department of Medical and Surgical Sciences (DIMEC), Università di Bologna, Bologna, Italy
| |
Collapse
|
6
|
Cui Z, Guo Z, Wei L, Zou X, Zhu Z, Liu Y, Wang J, Chen L, Wang D, Ke Z. Altered pain sensitivity in 5×familial Alzheimer disease mice is associated with dendritic spine loss in anterior cingulate cortex pyramidal neurons. Pain 2022; 163:2138-2153. [PMID: 35384934 PMCID: PMC9578529 DOI: 10.1097/j.pain.0000000000002648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 11/26/2022]
Abstract
ABSTRACT Chronic pain is highly prevalent. Individuals with cognitive disorders such as Alzheimer disease are a susceptible population in which pain is frequently difficult to diagnosis. It is still unclear whether the pathological changes in patients with Alzheimer disease will affect pain processing. Here, we leverage animal behavior, neural activity recording, optogenetics, chemogenetics, and Alzheimer disease modeling to examine the contribution of the anterior cingulate cortex (ACC) neurons to pain response. The 5× familial Alzheimer disease mice show alleviated mechanical allodynia which can be regained by the genetic activation of ACC excitatory neurons. Furthermore, the lower peak neuronal excitation, delayed response initiation, as well as the dendritic spine reduction of ACC pyramidal neurons in 5×familial Alzheimer disease mice can be mimicked by Rac1 or actin polymerization inhibitor in wild-type (WT) mice. These findings indicate that abnormal of pain sensitivity in Alzheimer disease modeling mice is closely related to the variation of neuronal activity and dendritic spine loss in ACC pyramidal neurons, suggesting the crucial role of dendritic spine density in pain processing.
Collapse
Affiliation(s)
- Zhengyu Cui
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Internal Medicine of Traditional Chinese Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Zhongzhao Guo
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Luyao Wei
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiang Zou
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zilu Zhu
- Department of Physiology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuchen Liu
- Department of Physiology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Wang
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liang Chen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Deheng Wang
- Department of Physiology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zunji Ke
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
7
|
van Elzelingen W, Warnaar P, Matos J, Bastet W, Jonkman R, Smulders D, Goedhoop J, Denys D, Arbab T, Willuhn I. Striatal dopamine signals are region specific and temporally stable across action-sequence habit formation. Curr Biol 2022; 32:1163-1174.e6. [PMID: 35134325 PMCID: PMC8926842 DOI: 10.1016/j.cub.2021.12.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 11/03/2021] [Accepted: 12/09/2021] [Indexed: 12/24/2022]
Abstract
Habits are automatic, inflexible behaviors that develop slowly with repeated performance. Striatal dopamine signaling instantiates this habit-formation process, presumably region specifically and via ventral-to-dorsal and medial-to-lateral signal shifts. Here, we quantify dopamine release in regions implicated in these presumed shifts (ventromedial striatum [VMS], dorsomedial striatum [DMS], and dorsolateral striatum [DLS]) in rats performing an action-sequence task and characterize habit development throughout a 10-week training. Surprisingly, all regions exhibited stable dopamine dynamics throughout habit development. VMS and DLS signals did not differ between habitual and non-habitual animals, but DMS dopamine release increased during action-sequence initiation and decreased during action-sequence completion in habitual rats, whereas non-habitual rats showed opposite effects. Consistently, optogenetic stimulation of DMS dopamine release accelerated habit formation. Thus, we demonstrate that dopamine signals do not shift regionally during habit formation and that dopamine in DMS, but not VMS or DLS, determines habit bias, attributing "habit functions" to a region previously associated exclusively with non-habitual behavior.
Collapse
Affiliation(s)
- Wouter van Elzelingen
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands; Department of Psychiatry, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 5, 1105 AZ Amsterdam, the Netherlands
| | - Pascal Warnaar
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands; Department of Psychiatry, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 5, 1105 AZ Amsterdam, the Netherlands
| | - João Matos
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands; Department of Psychiatry, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 5, 1105 AZ Amsterdam, the Netherlands
| | - Wieneke Bastet
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands; Department of Psychiatry, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 5, 1105 AZ Amsterdam, the Netherlands
| | - Roos Jonkman
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands; Department of Psychiatry, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 5, 1105 AZ Amsterdam, the Netherlands
| | - Dyonne Smulders
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands; Department of Psychiatry, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 5, 1105 AZ Amsterdam, the Netherlands
| | - Jessica Goedhoop
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands; Department of Psychiatry, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 5, 1105 AZ Amsterdam, the Netherlands
| | - Damiaan Denys
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands; Department of Psychiatry, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 5, 1105 AZ Amsterdam, the Netherlands
| | - Tara Arbab
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands; Department of Psychiatry, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 5, 1105 AZ Amsterdam, the Netherlands
| | - Ingo Willuhn
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands; Department of Psychiatry, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 5, 1105 AZ Amsterdam, the Netherlands.
| |
Collapse
|
8
|
Wu C, Zheng W, Jia X, Li Y, Shen F, Haghparast A, Liang J, Sui N, Zhang J. Adolescent chronic unpredictable stress causes a bias in goal‐directed behavior and distinctively changes the expression of NMDA and dopamine receptors in the dorsomedial and dorsolateral striatum in male rats. Dev Psychobiol 2022; 64:e22235. [DOI: 10.1002/dev.22235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/10/2021] [Accepted: 12/05/2021] [Indexed: 11/08/2022]
Affiliation(s)
- Chao Wu
- CAS Key Laboratory of Mental Health Institute of Psychology Beijing China
- Department of Psychology University of Chinese Academy of Sciences Beijing China
| | - Wei Zheng
- CAS Key Laboratory of Mental Health Institute of Psychology Beijing China
- Department of Psychology University of Chinese Academy of Sciences Beijing China
| | - Xiaohua Jia
- Key Laboratory of Molecular Imaging of Chinese Academy of Sciences Institute of Automation Chinese Academy of Sciences Beijing China
| | - Yonghui Li
- CAS Key Laboratory of Mental Health Institute of Psychology Beijing China
- Department of Psychology University of Chinese Academy of Sciences Beijing China
| | - Fang Shen
- CAS Key Laboratory of Mental Health Institute of Psychology Beijing China
- Department of Psychology University of Chinese Academy of Sciences Beijing China
| | - Abbas Haghparast
- Neuroscience Research Center School of Medicine Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Jing Liang
- CAS Key Laboratory of Mental Health Institute of Psychology Beijing China
- Department of Psychology University of Chinese Academy of Sciences Beijing China
| | - Nan Sui
- CAS Key Laboratory of Mental Health Institute of Psychology Beijing China
- Department of Psychology University of Chinese Academy of Sciences Beijing China
| | - Jian‐Jun Zhang
- CAS Key Laboratory of Mental Health Institute of Psychology Beijing China
- Department of Psychology University of Chinese Academy of Sciences Beijing China
| |
Collapse
|
9
|
Hollon NG, Williams EW, Howard CD, Li H, Traut TI, Jin X. Nigrostriatal dopamine signals sequence-specific action-outcome prediction errors. Curr Biol 2021; 31:5350-5363.e5. [PMID: 34637751 DOI: 10.1016/j.cub.2021.09.040] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 08/31/2021] [Accepted: 09/15/2021] [Indexed: 01/08/2023]
Abstract
Dopamine has been suggested to encode cue-reward prediction errors during Pavlovian conditioning, signaling discrepancies between actual versus expected reward predicted by the cues.1-5 While this theory has been widely applied to reinforcement learning concerning instrumental actions, whether dopamine represents action-outcome prediction errors and how it controls sequential behavior remain largely unknown. The vast majority of previous studies examining dopamine responses primarily have used discrete reward-predictive stimuli,1-15 whether Pavlovian conditioned stimuli for which no action is required to earn reward or explicit discriminative stimuli that essentially instruct an animal how and when to respond for reward. Here, by training mice to perform optogenetic intracranial self-stimulation, we examined how self-initiated goal-directed behavior influences nigrostriatal dopamine transmission during single and sequential instrumental actions, in behavioral contexts with minimal overt changes in the animal's external environment. We found that dopamine release evoked by direct optogenetic stimulation was dramatically reduced when delivered as the consequence of the animal's own action, relative to non-contingent passive stimulation. This dopamine suppression generalized to food rewards was specific to the reinforced action, was temporally restricted to counteract the expected outcome, and exhibited sequence-selectivity consistent with hierarchical control of sequential behavior. These findings demonstrate that nigrostriatal dopamine signals sequence-specific prediction errors in action-outcome associations, with fundamental implications for reinforcement learning and instrumental behavior in health and disease.
Collapse
Affiliation(s)
- Nick G Hollon
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Elora W Williams
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Christopher D Howard
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Hao Li
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Tavish I Traut
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Xin Jin
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Center for Motor Control and Disease, Key Laboratory of Brain Functional Genomics, East China Normal University, Shanghai 200062, China; NYU-ECNU Institute of Brain and Cognitive Science, New York University Shanghai, Shanghai 200062, China.
| |
Collapse
|
10
|
Wang Y, Zhang H, Wang J, Yu M, Zhang Q, Yan S, You D, Shi L, Zhang L, Wang L, Wu H, Cao X. Aconiti lateralis Radix Praeparata inhibits Alzheimer's disease by regulating the complex regulation network with the core of GRIN1 and MAPK1. PHARMACEUTICAL BIOLOGY 2021; 59:311-320. [PMID: 33784489 PMCID: PMC8018400 DOI: 10.1080/13880209.2021.1900879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/26/2021] [Accepted: 03/04/2021] [Indexed: 06/01/2023]
Abstract
CONTEXT Current medicine for Alzheimer's disease (AD) cannot effectively reverse or block nerve injury. Traditional Chinese Medicine practice and research imply Aconiti lateralis Radix Praeparata (Fuzi) may meet this goal. OBJECTIVE Analysing the anti-AD effect of Fuzi and its potential molecular mechanism. MATERIALS AND METHODS AD model cells were treated with Fuzi in 0-300 mg/mL for 24 h in 37 °C. The cell viability (CV) and length of cell projections (LCP) for each group were observed, analysed, and standardised using control as a baseline (CVs and LCPs). The Fuzi and AD relevant genes were identified basing on databases, and the molecular mechanism of Fuzi anti-AD was predicted by network analysis. RESULTS Experiment results showed that Fuzi in 0.4 mg/mL boosted LCP (LCPs = 1.2533, p ≤ 0.05), and in 1.6-100 mg/mL increased CV (CVs from 1.1673 to 1.3321, p ≤ 0.05). Bioinformatics analysis found 17 Fuzi target genes (relevant scores ≥ 20), showing strong AD relevant signals (RMS_p ≤ 0.05, related scores ≥ 5), enriched in the pathways regulating axon growth, synaptic plasticity, cell survival, proliferation, apoptosis, and death (p ≤ 0.05). Especially, GRIN1 and MAPK1 interacted with APP protein and located in the key point of the "Alzheimer's disease" pathway. DISCUSSION AND CONCLUSIONS These results suggest that Fuzi may have therapeutic and prevention potential in AD, and GRIN1 and MAPK1 may be the core of the pathways of the Fuzi anti-AD process. Fuzi should be studied more extensively, especially for the prevention of AD.
Collapse
Affiliation(s)
- Yutao Wang
- Department of Laboratory Animal Science, Kunming Medical University, Kunming, China
- Basic Medical College, Kunming Medical University, Kunming, China
| | - Huixiang Zhang
- Institute of Neuroscience, Basic Medical College, Kunming Medical University, Kunming, China
| | - Jing Wang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Ming Yu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Bioengineering Centre, Kunming Medical University, Kunming, P.R. China
| | - Qianqian Zhang
- Basic Medical College, Kunming Medical University, Kunming, China
| | - Shan Yan
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Bioengineering Centre, Kunming Medical University, Kunming, P.R. China
| | - Dingyun You
- School of Public Health, Kunming Medical University, Kunming, China
| | - Lanlan Shi
- Basic Medical College, Kunming Medical University, Kunming, China
| | - Lihuan Zhang
- Department of Laboratory Animal Science, Kunming Medical University, Kunming, China
| | - Limei Wang
- Department of Laboratory Animal Science, Kunming Medical University, Kunming, China
| | - Hongxiang Wu
- Faculty of Rehabilitation Medicine, Kunming Medical University, Kunming, China
| | - Xue Cao
- Department of Laboratory Animal Science, Kunming Medical University, Kunming, China
| |
Collapse
|
11
|
Smaga I, Wydra K, Suder A, Sanak M, Caffino L, Fumagalli F, Filip M. Enhancement of the GluN2B subunit of glutamatergic NMDA receptors in rat brain areas after cocaine abstinence. J Psychopharmacol 2021; 35:1226-1239. [PMID: 34587833 DOI: 10.1177/02698811211048283] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Cocaine use disorder is associated with compulsive drug-seeking and drug-taking, whereas relapse may be induced by several factors, including stress, drug-related places, people, and cues. Recent observations strongly support the involvement of the N-methyl-D-aspartate (NMDA) receptors in cocaine use disorders and abstinence, whereas withdrawal in different environments may affect the intensification of relapse. METHODS The aim of this study was to examine the GluN2B subunit expression and its association with the postsynaptic density protein 95 (PSD95) in several brain structures in rats with a history of cocaine self-administration and housed either in an enriched environment or in an isolated condition. Furthermore, a selective antagonist of the GluN2B subunit-CP 101,606 (10 and 20 mg/kg) administered during exposure to cocaine or a drug-associated conditional stimulus (a cue) was used to evaluate seeking behavior in rats. RESULTS In rats previously self-administering cocaine, we observed an increase in the GluN2B expression in the total homogenate from the dorsal hippocampus under both enriched environment and isolation. Cocaine abstinence under isolation conditions increased the GluN2B and GluN2B/PSD95 complex levels in the PSD fraction of the prelimbic cortex in rats previously self-administering cocaine. Administration of CP 101,606 attenuated cue-induced cocaine-seeking behavior only in isolation-housed rats. CONCLUSION In summary, in this study we showed region-specific changes in both the expression of GluN2B subunit and NMDA receptor trafficking during cocaine abstinence under different housing conditions. Furthermore, we showed that the pharmacological blockade of the GluN2B subunit may be useful in attenuating cocaine-seeking behavior.
Collapse
Affiliation(s)
- Irena Smaga
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Karolina Wydra
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Agata Suder
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Marek Sanak
- Department of Internal Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Małgorzata Filip
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| |
Collapse
|
12
|
Varul J, Eskla KL, Piirsalu M, Innos J, Philips MA, Visnapuu T, Plaas M, Vasar E. Dopamine System, NMDA Receptor and EGF Family Expressions in Brain Structures of Bl6 and 129Sv Strains Displaying Different Behavioral Adaptation. Brain Sci 2021; 11:brainsci11060725. [PMID: 34072341 PMCID: PMC8227283 DOI: 10.3390/brainsci11060725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 12/14/2022] Open
Abstract
C57BL/6NTac (Bl6) and 129S6/SvEvTac (129Sv) mice display different coping strategies in stressful conditions. Our aim was to evaluate biomarkers related to different adaptation strategies in the brain of male 129Sv and Bl6 mice. We focused on signaling pathways related to the dopamine (DA) system, N-methyl-D-aspartate (NMDA) receptor and epidermal growth factor (EGF) family, shown as the key players in behavioral adaptation. Mice from Bl6 and 129Sv lines were divided into either home cage controls (HCC group) or exposed to repeated motility testing and treated with saline for 11 days (RMT group). Distinct stress responses were reflected in severe body weight loss in 129Sv and the increased exploratory behavior in Bl6 mice. Besides that, amphetamine caused significantly stronger motor stimulation in Bl6. Together with the results from gene expression (particularly Maob), this study supports higher baseline activity of DA system in Bl6. Interestingly, the adaptation is reflected with opposite changes of DA markers in dorsal and ventral striatum. In forebrain, stress increased the gene expressions of Egf-Erbb1 and Nrg1/Nrg2-Erbb4 pathways more clearly in 129Sv, whereas the corresponding proteins were significantly elevated in Bl6. We suggest that not only inhibited activity of the DA system, but also reduced activity of EGF family and NMDA receptor signaling underlies higher susceptibility to stress in 129Sv. Altogether, this study underlines the better suitability of 129Sv for modelling neuropsychiatric disorders than Bl6.
Collapse
Affiliation(s)
- Jane Varul
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (K.-L.E.); (M.P.); (J.I.); (M.-A.P.); (T.V.); (M.P.); (E.V.)
- Center of Excellence for Genomics and Translational Medicine, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia
- Correspondence:
| | - Kattri-Liis Eskla
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (K.-L.E.); (M.P.); (J.I.); (M.-A.P.); (T.V.); (M.P.); (E.V.)
- Center of Excellence for Genomics and Translational Medicine, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia
| | - Maria Piirsalu
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (K.-L.E.); (M.P.); (J.I.); (M.-A.P.); (T.V.); (M.P.); (E.V.)
- Center of Excellence for Genomics and Translational Medicine, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia
| | - Jürgen Innos
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (K.-L.E.); (M.P.); (J.I.); (M.-A.P.); (T.V.); (M.P.); (E.V.)
- Center of Excellence for Genomics and Translational Medicine, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia
| | - Mari-Anne Philips
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (K.-L.E.); (M.P.); (J.I.); (M.-A.P.); (T.V.); (M.P.); (E.V.)
- Center of Excellence for Genomics and Translational Medicine, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia
| | - Tanel Visnapuu
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (K.-L.E.); (M.P.); (J.I.); (M.-A.P.); (T.V.); (M.P.); (E.V.)
- Center of Excellence for Genomics and Translational Medicine, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia
| | - Mario Plaas
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (K.-L.E.); (M.P.); (J.I.); (M.-A.P.); (T.V.); (M.P.); (E.V.)
- Center of Excellence for Genomics and Translational Medicine, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia
- Laboratory Animal Center, Institute of Biomedicine and Translational Medicine, University of Tartu, 14B Ravila Street, 50411 Tartu, Estonia
| | - Eero Vasar
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (K.-L.E.); (M.P.); (J.I.); (M.-A.P.); (T.V.); (M.P.); (E.V.)
- Center of Excellence for Genomics and Translational Medicine, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia
| |
Collapse
|
13
|
Salesse C, Charest J, Doucet-Beaupré H, Castonguay AM, Labrecque S, De Koninck P, Lévesque M. Opposite Control of Excitatory and Inhibitory Synapse Formation by Slitrk2 and Slitrk5 on Dopamine Neurons Modulates Hyperactivity Behavior. Cell Rep 2021; 30:2374-2386.e5. [PMID: 32075770 DOI: 10.1016/j.celrep.2020.01.084] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 12/03/2019] [Accepted: 01/24/2020] [Indexed: 11/26/2022] Open
Abstract
The neurodevelopmental origin of hyperactivity disorder has been suggested to involve the dopaminergic system, but the underlying mechanisms are still unknown. Here, transcription factors Lmx1a and Lmx1b are shown to be essential for midbrain dopaminergic (mDA) neuron excitatory synaptic inputs and dendritic development. Strikingly, conditional knockout (cKO) of Lmx1a/b in postmitotic mDA neurons results in marked hyperactivity. In seeking Lmx1a/b target genes, we identify positively regulated Slitrk2 and negatively regulated Slitrk5. These two synaptic adhesion proteins promote excitatory and inhibitory synapses on mDA neurons, respectively. Knocking down Slitrk2 reproduces some of the Lmx1a/b cKO cellular and behavioral phenotypes, whereas Slitrk5 knockdown has opposite effects. The hyperactivity caused by this imbalance in excitatory/inhibitory synaptic inputs on dopamine neurons is reproduced by chronically inhibiting the ventral tegmental area during development using pharmacogenetics. Our study shows that alterations in developing dopaminergic circuits strongly impact locomotor activity, shedding light on mechanisms causing hyperactivity behaviors.
Collapse
Affiliation(s)
- Charleen Salesse
- CERVO Brain Research Centre, 2601 de la Canardière, Québec, QC G1J 2G3, Canada
| | - Julien Charest
- CERVO Brain Research Centre, 2601 de la Canardière, Québec, QC G1J 2G3, Canada
| | | | | | - Simon Labrecque
- CERVO Brain Research Centre, 2601 de la Canardière, Québec, QC G1J 2G3, Canada
| | - Paul De Koninck
- CERVO Brain Research Centre, 2601 de la Canardière, Québec, QC G1J 2G3, Canada; Department of Biochemistry, Microbiology and Bioinformatics, Faculty of Science and Engineering, Université Laval, Québec, QC G1V 0A6, Canada
| | - Martin Lévesque
- CERVO Brain Research Centre, 2601 de la Canardière, Québec, QC G1J 2G3, Canada; Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada.
| |
Collapse
|
14
|
Goodman J. Place vs. Response Learning: History, Controversy, and Neurobiology. Front Behav Neurosci 2021; 14:598570. [PMID: 33643005 PMCID: PMC7904695 DOI: 10.3389/fnbeh.2020.598570] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 12/21/2020] [Indexed: 01/26/2023] Open
Abstract
The present article provides a historical review of the place and response learning plus-maze tasks with a focus on the behavioral and neurobiological findings. The article begins by reviewing the conflict between Edward C. Tolman's cognitive view and Clark L. Hull's stimulus-response (S-R) view of learning and how the place and response learning plus-maze tasks were designed to resolve this debate. Cognitive learning theorists predicted that place learning would be acquired faster than response learning, indicating the dominance of cognitive learning, whereas S-R learning theorists predicted that response learning would be acquired faster, indicating the dominance of S-R learning. Here, the evidence is reviewed demonstrating that either place or response learning may be dominant in a given learning situation and that the relative dominance of place and response learning depends on various parametric factors (i.e., amount of training, visual aspects of the learning environment, emotional arousal, et cetera). Next, the neurobiology underlying place and response learning is reviewed, providing strong evidence for the existence of multiple memory systems in the mammalian brain. Research has indicated that place learning is principally mediated by the hippocampus, whereas response learning is mediated by the dorsolateral striatum. Other brain regions implicated in place and response learning are also discussed in this section, including the dorsomedial striatum, amygdala, and medial prefrontal cortex. An exhaustive review of the neurotransmitter systems underlying place and response learning is subsequently provided, indicating important roles for glutamate, dopamine, acetylcholine, cannabinoids, and estrogen. Closing remarks are made emphasizing the historical importance of the place and response learning tasks in resolving problems in learning theory, as well as for examining the behavioral and neurobiological mechanisms of multiple memory systems. How the place and response learning tasks may be employed in the future for examining extinction, neural circuits of memory, and human psychopathology is also briefly considered.
Collapse
Affiliation(s)
- Jarid Goodman
- Department of Psychology, Delaware State University, Dover, DE, United States
| |
Collapse
|
15
|
Neurobiology of reward-related learning. Neurosci Biobehav Rev 2021; 124:224-234. [PMID: 33581225 DOI: 10.1016/j.neubiorev.2021.02.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 11/23/2022]
Abstract
A major goal in psychology is to understand how environmental stimuli associated with primary rewards come to function as conditioned stimuli, acquiring the capacity to elicit similar responses to those elicited by primary rewards. Our neurobiological model is predicated on the Hebbian idea that concurrent synaptic activity on the primary reward neural substrate-proposed to be ventral tegmental area (VTA) dopamine (DA) neurons-strengthens the synapses involved. We propose that VTA DA neurons receive both a strong unconditioned stimulus signal (acetylcholine stimulation of DA cells) from the primary reward capable of unconditionally activating DA cells and a weak stimulus signal (glutamate stimulation of DA cells) from the neutral stimulus. Through joint stimulation the weak signal is potentiated and capable of activating the VTA DA cells, eliciting a conditioned response. The learning occurs when this joint stimulation initiates intracellular second-messenger cascades resulting in enhanced glutamate-DA synapses. In this review we present evidence that led us to propose this model and the most recent evidence supporting it.
Collapse
|
16
|
Altered baseline and amphetamine-mediated behavioral profiles in dopamine transporter Cre (DAT-Ires-Cre) mice compared to tyrosine hydroxylase Cre (TH-Cre) mice. Psychopharmacology (Berl) 2020; 237:3553-3568. [PMID: 32778904 PMCID: PMC10120402 DOI: 10.1007/s00213-020-05635-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 07/31/2020] [Indexed: 02/06/2023]
Abstract
RATIONALE Transgenic mouse lines expressing Cre-recombinase under the regulation of either dopamine transporter (DAT) or tyrosine hydroxylase (TH) promoters are commonly used to study the dopamine (DA) system. While use of the TH promoter appears to have less liability to changes in native gene expression, transgene insertion in the DAT locus results in reduced DAT expression and function. This confound is sometimes overlooked in genetically targeted behavioral experiments. OBJECTIVES We sought to evaluate the suitability of DAT-Ires-Cre and TH-Cre transgenic lines for behavioral pharmacology experiments with DA agonists. We hypothesized that DAT-Ires-Cre expression would impact DAT-mediated behaviors, but no impact of TH-Cre expression would be observed. METHODS DAT-Ires-Cre and TH-Cre mice bred on mixed 129S6/C57BL/6 and pure C57BL/6 backgrounds were evaluated for novelty-induced, baseline, and amphetamine (AMPH)-induced locomotion, and for AMPH and D1 agonist (SKF-38393)-induced preservative behaviors. RESULTS DAT-Ires-Cre mice on both mixed 129S6/C57BL/6 and pure C57BL/6 backgrounds displayed increased novelty-induced activity and decreased AMPH-induced locomotion, with mixed results for AMPH-induced stereotypy. TH-Cre mice on both backgrounds showed typical baseline activity and AMPH-induced stereotypy, with a difference in AMPH-induced locomotion observed only on the mixed background. Both transgenic lines displayed unaltered SKF-38393-induced grooming behavior. CONCLUSIONS Our findings indicate that the DAT-Ires-Cre transgenic line may lead to confounds for experiments that are dependent on DAT expression. The TH-Cre transgenic line studied here may be a more useful option, depending on background strain, because of its lack of baseline and drug-induced phenotypes. These data highlight the importance of appropriate controls in studies employing transgenic mice.
Collapse
|
17
|
K369I Tau Mice Demonstrate a Shift Towards Striatal Neuron Burst Firing and Goal-directed Behaviour. Neuroscience 2020; 449:46-62. [PMID: 32949670 DOI: 10.1016/j.neuroscience.2020.09.023] [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: 04/05/2020] [Revised: 08/31/2020] [Accepted: 09/07/2020] [Indexed: 11/22/2022]
Abstract
Pathological forms of the microtubule-associated protein tau are involved in a large group of neurodegenerative diseases named tauopathies, including frontotemporal lobar degeneration (FTLD-tau). K369I mutant tau transgenic mice (K3 mice) recapitulate neural and behavioural symptoms of FTLD, including tau aggregates in the cortex, alterations to nigrostriatum, memory deficits and parkinsonism. The aim of this study was to further characterise the K3 mouse model by examining functional alterations to the striatum. Whole-cell patch-clamp electrophysiology was used to investigate the properties of striatal neurons in K3 mice and wildtype controls. Additionally, striatal-based instrumental learning tasks were conducted to assess goal-directed versus habitual behaviours (i.e., by examining sensitivity to outcome devaluation and progressive ratios). The K3 model demonstrated significant alterations in the discharge properties of striatal neurons relative to wildtype mice, which manifested as a shift in neuronal output towards a burst firing state. K3 mice acquired goal-directed responding faster than control mice and were goal-directed at test unlike wildtype mice, which is likely to indicate reduced capacity to develop habitual behaviour. The observed pattern of behaviour in K3 mice is suggestive of deficits in dorsal lateral striatal function and this was supported by our electrophysiological findings. Thus, both the electrophysiological and behavioural alterations indicate that K3 mice have early deficits in striatal function. This finding adds to the growing literature which indicate that the striatum is impacted in tau-related neuropathies such as FTLD, and further suggests that the K3 model is a unique mouse model for investigating FTLD especially with striatal involvement.
Collapse
|
18
|
Geramita MA, Yttri EA, Ahmari SE. The two‐step task, avoidance, and OCD. J Neurosci Res 2020; 98:1007-1019. [DOI: 10.1002/jnr.24594] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 01/02/2020] [Accepted: 01/30/2020] [Indexed: 01/12/2023]
Affiliation(s)
- Matthew A. Geramita
- Department of Psychiatry University of Pittsburgh Pittsburgh PA USA
- Department of Biological Sciences Carnegie Mellon University Pittsburgh PA USA
- Center for Neural Basis of Cognition University of Pittsburgh Pittsburgh PA USA
| | - Eric A. Yttri
- Department of Biological Sciences Carnegie Mellon University Pittsburgh PA USA
- Center for Neural Basis of Cognition University of Pittsburgh Pittsburgh PA USA
| | - Susanne E. Ahmari
- Department of Psychiatry University of Pittsburgh Pittsburgh PA USA
- Center for Neural Basis of Cognition University of Pittsburgh Pittsburgh PA USA
| |
Collapse
|
19
|
Coddington LT, Dudman JT. Learning from Action: Reconsidering Movement Signaling in Midbrain Dopamine Neuron Activity. Neuron 2020; 104:63-77. [PMID: 31600516 DOI: 10.1016/j.neuron.2019.08.036] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/10/2019] [Accepted: 08/22/2019] [Indexed: 01/07/2023]
Abstract
Animals infer when and where a reward is available from experience with informative sensory stimuli and their own actions. In vertebrates, this is thought to depend upon the release of dopamine from midbrain dopaminergic neurons. Studies of the role of dopamine have focused almost exclusively on their encoding of informative sensory stimuli; however, many dopaminergic neurons are active just prior to movement initiation, even in the absence of sensory stimuli. How should current frameworks for understanding the role of dopamine incorporate these observations? To address this question, we review recent anatomical and functional evidence for action-related dopamine signaling. We conclude by proposing a framework in which dopaminergic neurons encode subjective signals of action initiation to solve an internal credit assignment problem.
Collapse
|
20
|
Smaga I, Sanak M, Filip M. Cocaine-induced Changes in the Expression of NMDA Receptor Subunits. Curr Neuropharmacol 2020; 17:1039-1055. [PMID: 31204625 PMCID: PMC7052821 DOI: 10.2174/1570159x17666190617101726] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 05/31/2019] [Accepted: 06/11/2019] [Indexed: 11/28/2022] Open
Abstract
Cocaine use disorder is manifested by repeated cycles of drug seeking and drug taking. Cocaine exposure causes synaptic transmission in the brain to exhibit persistent changes, which are poorly understood, while the pharmacotherapy of this disease has not been determined. Multiple potential mechanisms have been indicated to be involved in the etiology of co-caine use disorder. The glutamatergic system, especially N-methyl-D-aspartate (NMDA) receptors, may play a role in sever-al physiological processes (synaptic plasticity, learning and memory) and in the pathogenesis of cocaine use disorder. The composition of the NMDA receptor subunits changes after contingent and noncontingent cocaine administration and after drug abstinence in a region-specific and time-dependent manner, as well as depending on the different protocols used for co-caine administration. Changes in the expression of NMDA receptor subunits may underlie the transition from cocaine abuse to dependence, as well as the transition from cocaine dependence to cocaine withdrawal. In this paper, we summarize the cur-rent knowledge regarding neuroadaptations within NMDA receptor subunits and scaffolding proteins observed following voluntary and passive cocaine intake, as well as the effects of NMDA receptor antagonists on cocaine-induced behavioral changes during cocaine seeking and relapse.
Collapse
Affiliation(s)
- Irena Smaga
- Department of Internal Medicine, Jagiellonian University Medical College, Skawińska 8, PL 31-066 Kraków, Poland.,Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna 12, PL 31-343 Kraków, Poland
| | - Marek Sanak
- Department of Internal Medicine, Jagiellonian University Medical College, Skawińska 8, PL 31-066 Kraków, Poland
| | - Małgorzata Filip
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna 12, PL 31-343 Kraków, Poland
| |
Collapse
|
21
|
Zhang W, Yu W, Liu X, Wang Q, Bai X, Cui X, Wang S. Effect of Jian-Pi-Zhi-Dong Decoction on the Amino Acid Neurotransmitters in a Rat Model of Tourette Syndrome and Comorbid Anxiety Disorder. Front Psychiatry 2020; 11:515. [PMID: 32581885 PMCID: PMC7292006 DOI: 10.3389/fpsyt.2020.00515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/19/2020] [Indexed: 01/22/2023] Open
Abstract
Amino acid neurotransmitters have been shown to correlate with Tourette syndrome (TS) and its comorbidities. In this study, we investigated the effects of Jian-Pi-Zhi-Dong Decoction (JPZDD), a formula containing 10 different Chinese medical herbs, on amino acid neurotransmitters in rats. We established a rat model of Tourette syndrome and comorbid anxiety with an iminodipropionitrile injection plus uncertain empty water bottle stimulation for 3 weeks. Then the rats were randomly divided into four groups: control group and model group were gavaged with saline, while the remaining two treatment groups were gavaged with fluoxetine hydrochloride or JPZDD for four consecutive weeks. We recorded the behaviors of the rats with TS and comorbid anxiety by stereotypy recording, open field test, and elevated plus maze. We observed mitochondrial changes with transmission electron microscopy. We measured the content of glutamate (GLU) and γ-aminobutyric acid (GABA) both in the serum and striatum and the expression of their receptors by Western blot and real-time polymerase chain reaction. The study revealed that JPZDD was effective in alleviating the behavioral symptoms of both tic and anxiety in the rat model groups. These results might be associated with the increase in GABA levels and decrease in GLU levels in the serum, as well as an increase in striatal GABA level by the activation of GABA receptors Type A (GABAAR). JPZDD treatment also reversed the mitochondrial dysfunction both in the striatum and cortex in affected animals.
Collapse
Affiliation(s)
- Wen Zhang
- Department of Pediatrics, The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Wenjing Yu
- Department of Pediatrics, The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaofang Liu
- Department of Pediatrics, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qian Wang
- Department of Pediatrics, The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xue Bai
- Department of Pediatrics, The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xia Cui
- Department of Pediatrics, The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Sumei Wang
- Department of Pediatrics, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
22
|
Neural circuits in goal-directed and habitual behavior: Implications for circuit dysfunction in obsessive-compulsive disorder. Neurochem Int 2019; 129:104464. [DOI: 10.1016/j.neuint.2019.104464] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 04/11/2019] [Accepted: 05/09/2019] [Indexed: 01/04/2023]
|
23
|
Delius JD, Delius JAM. Systematic Analysis of Pigeons' Discrimination of Pixelated Stimuli: A Hierarchical Pattern Recognition System Is Not Identifiable. Sci Rep 2019; 9:13929. [PMID: 31558750 PMCID: PMC6763494 DOI: 10.1038/s41598-019-50212-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 09/06/2019] [Indexed: 02/07/2023] Open
Abstract
Pigeons learned to discriminate two different patterns displayed with miniature light-emitting diode arrays. They were then tested with 84 interspersed, non-reinforced degraded pattern pairs. Choices ranged between 100% and 50% for one or other of the patterns. Stimuli consisting of few pixels yielded low choice scores whereas those consisting of many pixels yielded a broad range of scores. Those patterns with a high number of pixels coinciding with those of the rewarded training stimulus were preferred and those with a high number of pixels coinciding with the non-rewarded training pattern were avoided; a discrimination index based on this correlated 0.74 with the pattern choices. Pixels common to both training patterns had a minimal influence. A pixel-by-pixel analysis revealed that eight pixels of one pattern and six pixels of the other pattern played a prominent role in the pigeons’ choices. These pixels were disposed in four and two clusters of neighbouring locations. A summary index calculated on this basis still only yielded a weak 0.73 correlation. The individual pigeons’ data furthermore showed that these clusters were a mere averaging mirage. The pigeons’ performance depends on deep learning in a midbrain-based multimillion synapse neuronal network. Pixelated visual patterns should be helpful when simulating perception of patterns with artificial networks.
Collapse
Affiliation(s)
- Juan D Delius
- Experimental Psychology, University of Konstanz, 78457, Konstanz, Germany.
| | - Julia A M Delius
- Center for Lifespan Psychology, Max Planck Institute for Human Development, 14195, Berlin, Germany
| |
Collapse
|
24
|
Lipton DM, Gonzales BJ, Citri A. Dorsal Striatal Circuits for Habits, Compulsions and Addictions. Front Syst Neurosci 2019; 13:28. [PMID: 31379523 PMCID: PMC6657020 DOI: 10.3389/fnsys.2019.00028] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 06/27/2019] [Indexed: 12/18/2022] Open
Abstract
Here, we review the neural circuit bases of habits, compulsions, and addictions, behaviors which are all characterized by relatively automatic action performance. We discuss relevant studies, primarily from the rodent literature, and describe how major headway has been made in identifying the brain regions and neural cell types whose activity is modulated during the acquisition and performance of these automated behaviors. The dorsal striatum and cortical inputs to this structure have emerged as key players in the wider basal ganglia circuitry encoding behavioral automaticity, and changes in the activity of different neuronal cell-types in these brain regions have been shown to co-occur with the formation of automatic behaviors. We highlight how disordered functioning of these neural circuits can result in neuropsychiatric disorders, such as obsessive-compulsive disorder (OCD) and drug addiction. Finally, we discuss how the next phase of research in the field may benefit from integration of approaches for access to cells based on their genetic makeup, activity, connectivity and precise anatomical location.
Collapse
Affiliation(s)
- David M Lipton
- Edmond and Lily Safra Center for Brain Sciences, Hebrew University of Jerusalem, Jerusalem, Israel.,Zuckerman Postdoctoral Scholar, Jerusalem, Israel
| | - Ben J Gonzales
- Institute of Life Sciences, Edmond J. Safra Campus, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ami Citri
- Edmond and Lily Safra Center for Brain Sciences, Hebrew University of Jerusalem, Jerusalem, Israel.,Institute of Life Sciences, Edmond J. Safra Campus, Hebrew University of Jerusalem, Jerusalem, Israel.,Program in Child and Brain Development, MaRS Centre, Canadian Institute for Advanced Research, Toronto, ON, Canada
| |
Collapse
|
25
|
Destreel G, Seutin V, Engel D. Subsaturation of the N-methyl-D-aspartate receptor glycine site allows the regulation of bursting activity in juvenile rat nigral dopamine neurons. Eur J Neurosci 2019; 50:3454-3471. [PMID: 31206829 DOI: 10.1111/ejn.14491] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/23/2019] [Accepted: 06/10/2019] [Indexed: 01/09/2023]
Abstract
The activation of N-methyl-D-aspartate receptors (NMDARs) in substantia nigra pars compacta (SNc) dopamine (DA) cells is central to generate the bursting activity, a phasic signal linked to DA-related behaviours via the change in postsynaptic DA release. NMDARs are recruited during excitatory synaptic transmission by glutamate release, but the glycine site level of occupancy of these receptors during basal action potential-dependent activity is not known for SNc DA neurons. We explored NMDAR-dependent signals during exogenous applications of co-agonists in midbrain slices from juvenile rats. We found that both glycine and D-serine strengthened the NMDAR-dependent component of excitatory postsynaptic currents (EPSCs) in a concentration-dependent manner. EPSCs were also increased by endogenous glycine via the blockade of the glycine transport. The glycine site of NMDARs contributing to synaptic transmission is therefore subsaturated. The behaviourally relevant burst firing was more sensitive to exogenous D-serine and endogenous glycine than to exogenous glycine. The mechanisms regulating the availability of the co-agonists exert consequently a critical influence on the excitability of DA neurons via NMDARs. The modulation of the phasic firing in DA neurons by ambient NMDAR co-agonists may be important for nigral information processing and downstream motor-related behaviour.
Collapse
Affiliation(s)
- Geoffrey Destreel
- GIGA-Neurosciences, Neurophysiology Group, University of Liege, Liege, Belgium
| | - Vincent Seutin
- GIGA-Neurosciences, Neurophysiology Group, University of Liege, Liege, Belgium
| | - Dominique Engel
- GIGA-Neurosciences, Neurophysiology Group, University of Liege, Liege, Belgium
| |
Collapse
|
26
|
Shearrer GE, Nansel TR, Lipsky LM, Sadler JR, Burger KS. The impact of elevated body mass on brain responses during appetitive prediction error in postpartum women. Physiol Behav 2019; 206:243-251. [PMID: 30986423 DOI: 10.1016/j.physbeh.2019.04.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 04/11/2019] [Accepted: 04/11/2019] [Indexed: 12/01/2022]
Abstract
Repeated exposure to highly palatable foods and elevated weight promote: 1) insensitivity to punishment in striatal regions and, 2) increased willingness to work for food. We hypothesized that BMI would be positively associated with negative prediction error BOLD response in the occipital cortex. Additionally, we postulated that food reinforcement value would be negatively associated with negative prediction error BOLD response in the orbital frontal cortex and amygdala. Postpartum women (n = 47; BMI = 25.5 ± 5.1) were 'trained' to associate specific cues paired to either a highly palatable milkshake or a sub-palatable milkshake. We then violated these cue-taste pairings in 40% of the trials by showing a palatable cue followed by the sub-palatable taste (negative prediction error). Contrary to our hypotheses, during negative prediction error (mismatched cue-taste) versus matched palatable cue-taste, women showed increased BOLD response in the central operculum (pFWE = 0.002; k = 1680; MNI: -57, -7,14) and postcentral gyrus (pFWE = 0.006, k = 1219; MNI: 62, -8,18). When comparing the matched sub-palatable cue-taste to the negative prediction error trials, BOLD response increased in the postcentral gyrus (r = -0.60, pFWE = 0.008), putamen (r = -0.55, pFWE = 0.02), and insula (r = -0.50, pFWE = 0.01). Similarly, viewing the palatable cue vs sub-palatable cue was related to BOLD response in the putamen (pFWE = 0.025, k = 53; MNI: -20, 6, -8) and the insula (pFWE = 0.04, k = 19, MNI:38, -12, -6). Neither BMI at 6-month postpartum nor food reinforcement value was related to BOLD response. The insula and putamen appear to encode for visual food cue processing, and the gustatory and somatosensory cortices appear to encode negative prediction errors. Differential response in the somatosensory cortex to the matched cue-taste pairs to negative prediction error may indicate that a palatable cue may dull aversive qualities in the stimulus.
Collapse
Affiliation(s)
- Grace E Shearrer
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Tonja R Nansel
- Social and Behavioral Sciences Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health & Human Development, Bethesda, MD, United States of America
| | - Leah M Lipsky
- Social and Behavioral Sciences Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health & Human Development, Bethesda, MD, United States of America
| | - Jennifer R Sadler
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Kyle S Burger
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America; Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America.
| |
Collapse
|
27
|
Tovar-Díaz J, Pomrenze MB, Kan R, Pahlavan B, Morikawa H. Cooperative CRF and α1 Adrenergic Signaling in the VTA Promotes NMDA Plasticity and Drives Social Stress Enhancement of Cocaine Conditioning. Cell Rep 2019. [PMID: 29514102 PMCID: PMC5877815 DOI: 10.1016/j.celrep.2018.02.039] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Stressful events rapidly trigger activity-dependent synaptic plasticity, driving the formation of aversive memories. However, it remains unclear how stressful experience affects plasticity mechanisms to regulate appetitive learning, such as intake of addictive drugs. Using rats, we show that corticotropin-releasing factor (CRF) and α1 adrenergic receptor (α1AR) signaling enhance the plasticity of NMDA-receptor-mediated glutamatergic transmission in ventral tegmental area (VTA) dopamine (DA) neurons through distinct effects on inositol 1,4,5-triphosphate (IP3)-dependent Ca2+ signaling. We find that CRF amplifies IP3-Ca2+ signaling induced by stimulation of α1ARs, revealing a cooperative mechanism that promotes glutamatergic plasticity. In line with this, acute social defeat stress engages similar cooperative CRF and α1AR signaling in the VTA to enhance learning of cocaine-paired cues. These data provide evidence that CRF and α1ARs act in concert to regulate IP3-Ca2+ signaling in the VTA and promote learning of drug-associated cues.
Collapse
Affiliation(s)
- Jorge Tovar-Díaz
- Department of Neuroscience, University of Texas at Austin, Austin, TX 78712, USA; Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | - Matthew B Pomrenze
- Department of Neuroscience, University of Texas at Austin, Austin, TX 78712, USA; Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA.
| | - Russell Kan
- Department of Neuroscience, University of Texas at Austin, Austin, TX 78712, USA
| | - Bahram Pahlavan
- Department of Neuroscience, University of Texas at Austin, Austin, TX 78712, USA; Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | - Hitoshi Morikawa
- Department of Neuroscience, University of Texas at Austin, Austin, TX 78712, USA; Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
28
|
Saunders A, Macosko EZ, Wysoker A, Goldman M, Krienen FM, de Rivera H, Bien E, Baum M, Bortolin L, Wang S, Goeva A, Nemesh J, Kamitaki N, Brumbaugh S, Kulp D, McCarroll SA. Molecular Diversity and Specializations among the Cells of the Adult Mouse Brain. Cell 2018; 174:1015-1030.e16. [PMID: 30096299 PMCID: PMC6447408 DOI: 10.1016/j.cell.2018.07.028] [Citation(s) in RCA: 978] [Impact Index Per Article: 163.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/26/2018] [Accepted: 07/20/2018] [Indexed: 02/06/2023]
Abstract
The mammalian brain is composed of diverse, specialized cell populations. To systematically ascertain and learn from these cellular specializations, we used Drop-seq to profile RNA expression in 690,000 individual cells sampled from 9 regions of the adult mouse brain. We identified 565 transcriptionally distinct groups of cells using computational approaches developed to distinguish biological from technical signals. Cross-region analysis of these 565 cell populations revealed features of brain organization, including a gene-expression module for synthesizing axonal and presynaptic components, patterns in the co-deployment of voltage-gated ion channels, functional distinctions among the cells of the vasculature and specialization of glutamatergic neurons across cortical regions. Systematic neuronal classifications for two complex basal ganglia nuclei and the striatum revealed a rare population of spiny projection neurons. This adult mouse brain cell atlas, accessible through interactive online software (DropViz), serves as a reference for development, disease, and evolution.
Collapse
Affiliation(s)
- Arpiar Saunders
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Evan Z Macosko
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA.
| | - Alec Wysoker
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Melissa Goldman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Fenna M Krienen
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Heather de Rivera
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Elizabeth Bien
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Matthew Baum
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Laura Bortolin
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Shuyu Wang
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Aleksandrina Goeva
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - James Nemesh
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Nolan Kamitaki
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sara Brumbaugh
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - David Kulp
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Steven A McCarroll
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
29
|
Cieślak PE, Ahn WY, Bogacz R, Rodriguez Parkitna J. Selective Effects of the Loss of NMDA or mGluR5 Receptors in the Reward System on Adaptive Decision-Making. eNeuro 2018; 5:ENEURO.0331-18.2018. [PMID: 30302389 PMCID: PMC6175304 DOI: 10.1523/eneuro.0331-18.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 05/23/2018] [Accepted: 06/03/2018] [Indexed: 11/30/2022] Open
Abstract
Selecting the most advantageous actions in a changing environment is a central feature of adaptive behavior. The midbrain dopamine (DA) neurons along with the major targets of their projections, including dopaminoceptive neurons in the frontal cortex and basal ganglia, play a key role in this process. Here, we investigate the consequences of a selective genetic disruption of NMDA receptor and metabotropic glutamate receptor 5 (mGluR5) in the DA system on adaptive choice behavior in mice. We tested the effects of the mutation on performance in the probabilistic reinforcement learning and probability-discounting tasks. In case of the probabilistic choice, both the loss of NMDA receptors in dopaminergic neurons or the loss mGluR5 receptors in D1 receptor-expressing dopaminoceptive neurons reduced the probability of selecting the more rewarded alternative and lowered the likelihood of returning to the previously rewarded alternative (win-stay). When observed behavior was fitted to reinforcement learning models, we found that these two mutations were associated with a reduced effect of the expected outcome on choice (i.e., more random choices). None of the mutations affected probability discounting, which indicates that all animals had a normal ability to assess probability. However, in both behavioral tasks animals with targeted loss of NMDA receptors in dopaminergic neurons or mGluR5 receptors in D1 neurons were significantly slower to perform choices. In conclusion, these results show that glutamate receptor-dependent signaling in the DA system is essential for the speed and accuracy of choices, but at the same time probably is not critical for correct estimation of probable outcomes.
Collapse
Affiliation(s)
- Przemysław Eligiusz Cieślak
- Department of Molecular Neuropharmacology, Institute of Pharmacology of the Polish Academy of Sciences, 31-343, Krakow, Poland
| | - Woo-Young Ahn
- Department of Psychology, Seoul National University, Seoul 08826, Korea
| | - Rafał Bogacz
- MRC Brain Networks Dynamics Unit, Nuffield Department of Clinical Neurosciences, Oxford University, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom
| | - Jan Rodriguez Parkitna
- Department of Molecular Neuropharmacology, Institute of Pharmacology of the Polish Academy of Sciences, 31-343, Krakow, Poland
| |
Collapse
|
30
|
Hutchison MA, Gu X, Adrover MF, Lee MR, Hnasko TS, Alvarez VA, Lu W. Genetic inhibition of neurotransmission reveals role of glutamatergic input to dopamine neurons in high-effort behavior. Mol Psychiatry 2018; 23:1213-1225. [PMID: 28194005 PMCID: PMC5555825 DOI: 10.1038/mp.2017.7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/29/2016] [Accepted: 12/28/2016] [Indexed: 02/07/2023]
Abstract
Midbrain dopamine neurons are crucial for many behavioral and cognitive functions. As the major excitatory input, glutamatergic afferents are important for control of the activity and plasticity of dopamine neurons. However, the role of glutamatergic input as a whole onto dopamine neurons remains unclear. Here we developed a mouse line in which glutamatergic inputs onto dopamine neurons are specifically impaired, and utilized this genetic model to directly test the role of glutamatergic inputs in dopamine-related functions. We found that while motor coordination and reward learning were largely unchanged, these animals showed prominent deficits in effort-related behavioral tasks. These results provide genetic evidence that glutamatergic transmission onto dopaminergic neurons underlies incentive motivation, a willingness to exert high levels of effort to obtain reinforcers, and have important implications for understanding the normal function of the midbrain dopamine system.
Collapse
Affiliation(s)
- M A Hutchison
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - X Gu
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - M F Adrover
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - M R Lee
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - T S Hnasko
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - V A Alvarez
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - W Lu
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA,Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 3C 1000, 35 Convent Drive, Bethesda, MD 20892, USA. E-mail:
| |
Collapse
|
31
|
|
32
|
Mahone EM, Puts NA, Edden RAE, Ryan M, Singer HS. GABA and glutamate in children with Tourette syndrome: A 1H MR spectroscopy study at 7T. Psychiatry Res 2018; 273:46-53. [PMID: 29329743 PMCID: PMC5815927 DOI: 10.1016/j.pscychresns.2017.12.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 10/06/2017] [Accepted: 12/30/2017] [Indexed: 02/08/2023]
Abstract
Tourette syndrome (TS) is characterized by presence of chronic, fluctuating motor and phonic tics. The underlying neurobiological basis for these movements is hypothesized to involve cortical-striatal-thalamo-cortical (CSTC) pathways. Two major neurotransmitters within these circuits are γ-aminobutyric acid (GABA) and glutamate. Seventy-five participants (32 with TS, 43 controls) ages 5-12 years completed 1H MRS at 7T. GABA and glutamate were measured in dorsolateral prefrontal cortex (DLPFC), ventromedial prefrontal cortex (VMPFC), premotor cortex (PMC), and striatum, and metabolites quantified using LCModel. Participants also completed neuropsychological assessment emphasizing inhibitory control. Scans were well tolerated by participants. Across ROIs combined, glutamate was significantly higher in the TS group, compared to controls, with no significant group differences in GABA observed. ROI analyses revealed significantly increased PMC glutamate in the TS group. Among children with TS, increased PMC glutamate was associated with improved selective motor inhibition; however, no significant associations were identified between levels of glutamate or GABA and tic severity. The dopaminergic system has long been considered to have a dominant role in TS. Accumulating evidence, however, suggests involvement of other neurotransmitter systems. Data obtained using 1H MRS at 7T supports alteration of glutamate within habitual behavior-related CSTC pathways of children with TS.
Collapse
Affiliation(s)
- E Mark Mahone
- Department of Neuropsychology, Kennedy Krieger Institute, 1750 E. Fairmount Ave., Baltimore, MD 21231, USA; Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, 600 N Wolfe St., Baltimore, MD 21287, USA.
| | - Nicolaas A Puts
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 600 N Wolfe St., Baltimore, MD 21287, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, 707 N Broadway, Baltimore, MD 21205, USA
| | - Richard A E Edden
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 600 N Wolfe St., Baltimore, MD 21287, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, 707 N Broadway, Baltimore, MD 21205, USA
| | - Matthew Ryan
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, 600 N Wolfe St., Baltimore, MD 21287, USA
| | - Harvey S Singer
- Department of Neurology, The Johns Hopkins University School of Medicine, 600 N Wolfe St., Baltimore, MD 21287, USA
| |
Collapse
|
33
|
Gu X, Lu W. Genetic deletion of NMDA receptors suppresses GABAergic synaptic transmission in two distinct types of central neurons. Neurosci Lett 2018; 668:147-153. [PMID: 29355693 DOI: 10.1016/j.neulet.2018.01.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/23/2017] [Accepted: 01/12/2018] [Indexed: 01/30/2023]
Abstract
NMDA-type ionotropic glutamate receptors (NMDARs) play an important role in the regulation of synapse development and function in the brain. Recently we have shown that NMDARs are critical for GABAergic synapse development in developing hippocampal neurons. However, it remains unclear whether NMDARs are important for establishment of GABAergic synaptic transmission in other types of neurons in the brain. Here we report that in both cortical pyramidal neurons and midbrain dopamine neurons in ventral tegmental area (VTA), genetic deletion of the GluN1 subunit, which is required for assembly of functional NMDARs, leads to a strong reduction of GABAergic synaptic transmission. These data demonstrate that NMDARs play an important role in the development of GABAergic synaptic transmission in two types of neurons with distinct developmental origins, and suggest that NMDARs are commonly involved in development of GABAergic synaptic transmission in different types of neurons in the brain.
Collapse
Affiliation(s)
- Xinglong Gu
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Wei Lu
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
34
|
TGF-β Signaling in Dopaminergic Neurons Regulates Dendritic Growth, Excitatory-Inhibitory Synaptic Balance, and Reversal Learning. Cell Rep 2017; 17:3233-3245. [PMID: 28009292 DOI: 10.1016/j.celrep.2016.11.068] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 09/26/2016] [Accepted: 11/22/2016] [Indexed: 12/21/2022] Open
Abstract
Neural circuits involving midbrain dopaminergic (DA) neurons regulate reward and goal-directed behaviors. Although local GABAergic input is known to modulate DA circuits, the mechanism that controls excitatory/inhibitory synaptic balance in DA neurons remains unclear. Here, we show that DA neurons use autocrine transforming growth factor β (TGF-β) signaling to promote the growth of axons and dendrites. Surprisingly, removing TGF-β type II receptor in DA neurons also disrupts the balance in TGF-β1 expression in DA neurons and neighboring GABAergic neurons, which increases inhibitory input, reduces excitatory synaptic input, and alters phasic firing patterns in DA neurons. Mice lacking TGF-β signaling in DA neurons are hyperactive and exhibit inflexibility in relinquishing learned behaviors and re-establishing new stimulus-reward associations. These results support a role for TGF-β in regulating the delicate balance of excitatory/inhibitory synaptic input in local microcircuits involving DA and GABAergic neurons and its potential contributions to neuropsychiatric disorders.
Collapse
|
35
|
|
36
|
Wang Q, Yin P, Yu B, Zhao Z, Richter-Levin G, Yu L, Cao X. Down-regulation of dorsal striatal αCaMKII causes striatum-related cognitive and synaptic disorders. Exp Neurol 2017; 298:112-121. [PMID: 28890075 DOI: 10.1016/j.expneurol.2017.09.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 08/25/2017] [Accepted: 09/06/2017] [Indexed: 12/14/2022]
Abstract
Alpha calcium/calmodulin dependent protein kinase II (αCaMKII) is a serine/threonine protein kinase which is expressed abundantly in dorsal striatum and is highly involved in the corticostriatal synaptic plasticity. Nevertheless, it currently remains unclear whether and how αCaMKII plays a in the striatum-related neural disorders. To address the above issue, lentivirus-mediated short hairpin RNA (shRNA) was used to silence the expression of αCaMKII gene in the dorsal striatum of mice. As a consequence of down-regulation of dorsal striatal αCaMKII expression, we observed defective motor skill learning in accelerating rotarod and response learning in water cross maze. Furthermore, impaired corticostriatal basal transmission and long-term potentiation (LTP), which correlated with the deficits in dorsal striatum-related cognition, were also detected in the αCaMKII-shRNA mice. Consistent with the above results, αCaMKII-shRNA mice exhibited a remarkable decline in GluA1-Ser831 and GluA1-Ser845 phosphorylation levels of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR), and a decline in the expression levels of N-methyl-d-aspartic acid receptor (NMDAR) subunits NR1, NR2A and NR2B. Taken together, αCaMKII down-regulation caused dorsal striatum-related cognitive disorders by inhibiting corticostriatal synaptic plasticity, which resulted from dysfunction of AMPARs and NMDARs. Our findings demonstrate for the first time an important role of αCaMKII in striatum-related neural disorders and provide further evidence for the proposition that corticostriatal LTP underlies aspects of dorsal striatum-related cognition.
Collapse
Affiliation(s)
- Qi Wang
- Key Laboratory of Brain Functional Genomics, MOE & STCSM, East China Normal University, Shanghai 200062, China
| | - Pengcheng Yin
- Key Laboratory of Brain Functional Genomics, MOE & STCSM, East China Normal University, Shanghai 200062, China
| | - Bin Yu
- Key Laboratory of Brain Functional Genomics, MOE & STCSM, East China Normal University, Shanghai 200062, China
| | - Zheng Zhao
- Key Laboratory of Brain Functional Genomics, MOE & STCSM, East China Normal University, Shanghai 200062, China
| | - Gal Richter-Levin
- "Sagol" Department of Neurobiology, University of Haifa, Haifa 31905, Israel
| | - Lu Yu
- Department of Chinese Internal Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Xiaohua Cao
- Key Laboratory of Brain Functional Genomics, MOE & STCSM, East China Normal University, Shanghai 200062, China.
| |
Collapse
|
37
|
Goodman J, Ressler RL, Packard MG. Enhancing and impairing extinction of habit memory through modulation of NMDA receptors in the dorsolateral striatum. Neuroscience 2017; 352:216-225. [DOI: 10.1016/j.neuroscience.2017.03.042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/22/2017] [Accepted: 03/26/2017] [Indexed: 01/20/2023]
|
38
|
Abstract
Habits, both good ones and bad ones, are pervasive in animal behavior. Important frameworks have been developed to understand habits through psychological and neurobiological studies. This work has given us a rich understanding of brain networks that promote habits, and has also helped us to understand what constitutes a habitual behavior as opposed to a behavior that is more flexible and prospective. Mounting evidence from studies using neural recording methods suggests that habit formation is not a simple process. We review this evidence and take the position that habits could be sculpted from multiple dissociable changes in neural activity. These changes occur across multiple brain regions and even within single brain regions. This strategy of classifying components of a habit based on different brain signals provides a potentially useful new way to conceive of disorders that involve overly fixed behaviors as arising from different potential dysfunctions within the brain's habit network.
Collapse
Affiliation(s)
- Kyle S Smith
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, New Hampshire, USA
| | - Ann M Graybiel
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
39
|
Marinova Z, Chuang DM, Fineberg N. Glutamate-Modulating Drugs as a Potential Therapeutic Strategy in Obsessive-Compulsive Disorder. Curr Neuropharmacol 2017; 15:977-995. [PMID: 28322166 PMCID: PMC5652017 DOI: 10.2174/1570159x15666170320104237] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 12/27/2016] [Accepted: 03/15/2017] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Obsessive-compulsive disorder (OCD) is a mental disease commonly associated with severe distress and impairment of social functioning. Serotonin reuptake inhibitors and/or cognitive behavioural therapy are the therapy of choice, however up to 40% of patients do not respond to treatment. Glutamatergic signalling has also been implicated in OCD. The aim of the current study was to review the clinical evidence for therapeutic utility of glutamate-modulating drugs as an augmentation or monotherapy in OCD patients. METHODS We conducted a search of the MEDLINE database for clinical studies evaluating the effect of glutamate-modulating drugs in OCD. RESULTS Memantine is the compound most consistently showing a positive effect as an augmentation therapy in OCD. Anti-convulsant drugs (lamotrigine, topiramate) and riluzole may also provide therapeutic benefit to some OCD patients. Finally, ketamine may be of interest due to its potential for a rapid onset of action. CONCLUSION Further randomized placebo-controlled trials in larger study populations are necessary in order to draw definitive conclusions on the utility of glutamate-modulating drugs in OCD. Furthermore, genetic and epigenetic factors, clinical symptoms and subtypes predicting treatment response to glutamate-modulating drugs need to be investigated systematically.
Collapse
Affiliation(s)
- Zoya Marinova
- Department of Psychosomatic Medicine, Clinic Barmelweid, Barmelweid, Switzerland
| | - De-Maw Chuang
- National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, United States
| | - Naomi Fineberg
- Hertfordshire Partnership University NHS Foundation Trust and University of Hertfordshire, Welwyn Garden City, AL8 6HG, United Kingdom
| |
Collapse
|
40
|
Kanaan AS, Gerasch S, García-García I, Lampe L, Pampel A, Anwander A, Near J, Möller HE, Müller-Vahl K. Pathological glutamatergic neurotransmission in Gilles de la Tourette syndrome. Brain 2016; 140:218-234. [DOI: 10.1093/brain/aww285] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/31/2016] [Accepted: 09/12/2016] [Indexed: 11/13/2022] Open
|
41
|
Jastrzębska K, Walczak M, Cieślak PE, Szumiec Ł, Turbasa M, Engblom D, Błasiak T, Parkitna JR. Loss of NMDA receptors in dopamine neurons leads to the development of affective disorder-like symptoms in mice. Sci Rep 2016; 6:37171. [PMID: 27853270 PMCID: PMC5112557 DOI: 10.1038/srep37171] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 10/25/2016] [Indexed: 12/28/2022] Open
Abstract
The role of changes in dopamine neuronal activity during the development of symptoms in affective disorders remains controversial. Here, we show that inactivation of NMDA receptors on dopaminergic neurons in adult mice led to the development of affective disorder-like symptoms. The loss of NMDA receptors altered activity and caused complete NMDA-insensitivity in dopamine-like neurons. Mutant mice exhibited increased immobility in the forced swim test and a decrease in social interactions. Mutation also led to reduced saccharin intake, however the preference of sweet taste was not significantly decreased. Additionally, we found that while mutant mice were slower to learn instrumental tasks, they were able to reach the same performance levels, had normal sensitivity to feedback and showed similar motivation to exert effort as control animals. Taken together these results show that inducing the loss of NMDA receptor-dependent activity in dopamine neurons is associated with development of affective disorder-like symptoms.
Collapse
Affiliation(s)
- Kamila Jastrzębska
- Laboratory of Transgenic Models, Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Krakow, Poland
| | - Magdalena Walczak
- Department of Neurophysiology and Chronobiology, Institute of Zoology, Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland
| | - Przemysław Eligiusz Cieślak
- Laboratory of Transgenic Models, Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Krakow, Poland
| | - Łukasz Szumiec
- Laboratory of Transgenic Models, Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Krakow, Poland
| | - Mateusz Turbasa
- Laboratory of Transgenic Models, Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Krakow, Poland
| | - David Engblom
- Cell Biology, Department of Clinical and Experimental Medicine, Linköping University, SE-581 85, Linköping, Sweden
| | - Tomasz Błasiak
- Department of Neurophysiology and Chronobiology, Institute of Zoology, Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland
| | - Jan Rodriguez Parkitna
- Laboratory of Transgenic Models, Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Krakow, Poland
| |
Collapse
|
42
|
Yau HJ, Wang DV, Tsou JH, Chuang YF, Chen BT, Deisseroth K, Ikemoto S, Bonci A. Pontomesencephalic Tegmental Afferents to VTA Non-dopamine Neurons Are Necessary for Appetitive Pavlovian Learning. Cell Rep 2016; 16:2699-2710. [PMID: 27568569 DOI: 10.1016/j.celrep.2016.08.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 06/28/2016] [Accepted: 07/31/2016] [Indexed: 12/23/2022] Open
Abstract
The ventral tegmental area (VTA) receives phenotypically distinct innervations from the pedunculopontine tegmental nucleus (PPTg). While PPTg-to-VTA inputs are thought to play a critical role in stimulus-reward learning, direct evidence linking PPTg-to-VTA phenotypically distinct inputs in the learning process remains lacking. Here, we used optogenetic approaches to investigate the functional contribution of PPTg excitatory and inhibitory inputs to the VTA in appetitive Pavlovian conditioning. We show that photoinhibition of PPTg-to-VTA cholinergic or glutamatergic inputs during cue presentation dampens the development of anticipatory approach responding to the food receptacle during the cue. Furthermore, we employed in vivo optetrode recordings to show that photoinhibition of PPTg cholinergic or glutamatergic inputs significantly decreases VTA non-dopamine (non-DA) neural activity. Consistently, photoinhibition of VTA non-DA neurons disrupts the development of cue-elicited anticipatory approach responding. Taken together, our study reveals a crucial regulatory mechanism by PPTg excitatory inputs onto VTA non-DA neurons during appetitive Pavlovian conditioning.
Collapse
Affiliation(s)
- Hau-Jie Yau
- Synaptic Plasticity Section, Intramural Research Program, National Institute on Drug Abuse, NIH, U.S. Department of Health and Human Services, Baltimore, MD 21224, USA; Graduate Institute of Brain and Mind Sciences, National Taiwan University, Taipei 10051, Taiwan
| | - Dong V Wang
- Neurocircuitry of Motivation Section, Intramural Research Program, National Institute on Drug Abuse, NIH, U.S. Department of Health and Human Services, Baltimore, MD 21224, USA
| | - Jen-Hui Tsou
- Synaptic Plasticity Section, Intramural Research Program, National Institute on Drug Abuse, NIH, U.S. Department of Health and Human Services, Baltimore, MD 21224, USA
| | - Yi-Fang Chuang
- Institute of Public Health, National Yang-Ming University, Taipei 112, Taiwan
| | - Billy T Chen
- Synaptic Plasticity Section, Intramural Research Program, National Institute on Drug Abuse, NIH, U.S. Department of Health and Human Services, Baltimore, MD 21224, USA; Ionis Pharmaceuticals Inc., Carlsbad, CA 92010, USA
| | - Karl Deisseroth
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering and Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Satoshi Ikemoto
- Neurocircuitry of Motivation Section, Intramural Research Program, National Institute on Drug Abuse, NIH, U.S. Department of Health and Human Services, Baltimore, MD 21224, USA
| | - Antonello Bonci
- Synaptic Plasticity Section, Intramural Research Program, National Institute on Drug Abuse, NIH, U.S. Department of Health and Human Services, Baltimore, MD 21224, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Psychiatry, Johns Hopkins University, Baltimore, MD 21287, USA.
| |
Collapse
|
43
|
Stelly CE, Pomrenze MB, Cook JB, Morikawa H. Repeated social defeat stress enhances glutamatergic synaptic plasticity in the VTA and cocaine place conditioning. eLife 2016; 5. [PMID: 27374604 PMCID: PMC4931908 DOI: 10.7554/elife.15448] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 06/07/2016] [Indexed: 11/13/2022] Open
Abstract
Enduring memories of sensory cues associated with drug intake drive addiction. It is well known that stressful experiences increase addiction vulnerability. However, it is not clear how repeated stress promotes learning of cue-drug associations, as repeated stress generally impairs learning and memory processes unrelated to stressful experiences. Here, we show that repeated social defeat stress in rats causes persistent enhancement of long-term potentiation (LTP) of NMDA receptor-mediated glutamatergic transmission in the ventral tegmental area (VTA). Protein kinase A-dependent increase in the potency of inositol 1,4,5-triphosphate-induced Ca2+ signaling underlies LTP facilitation. Notably, defeated rats display enhanced learning of contextual cues paired with cocaine experience assessed using a conditioned place preference (CPP) paradigm. Enhancement of LTP in the VTA and cocaine CPP in behaving rats both require glucocorticoid receptor activation during defeat episodes. These findings suggest that enhanced glutamatergic plasticity in the VTA may contribute, at least partially, to increased addiction vulnerability following repeated stressful experiences. DOI:http://dx.doi.org/10.7554/eLife.15448.001 Daily stress increases the likelihood that people who take drugs will become addicted. A very early step in the development of addiction is learning that certain people, places, or paraphernalia are associated with obtaining drugs. These ‘cues’ – drug dealers, bars, cigarette advertisements, etc. – become powerful motivators to seek out drugs and can trigger relapse in recovering addicts. It is thought that learning happens when synapses (the connections between neurons in the brain) that relay information about particular cues become stronger. However, it is not clear how stress promotes the learning of cue-drug associations. Stelly et al. investigated whether repeated episodes of stress make it easier to strengthen synapses on dopamine neurons, which are involved in processing rewards and addiction. For the experiments, rats were repeatedly exposed to a stressful situation – an encounter with an unfamiliar aggressive rat – every day for five days. Stelly et al. found that these stressed rats formed stronger associations between the drug cocaine and the place where they were given the drug (the cue). Furthermore, a mechanism that strengthens synapses was more sensitive in the stressed rats than in unstressed rats. These changes persisted for 10-30 days after the stressful situation, suggesting that stress might begin a period of time during which the individual is more vulnerable to addiction. The experiments also show that a hormone called corticosterone – which is released during stressful experiences – is necessary for stress to trigger the changes in the synapses and behavior of the rats. However, corticosterone must work with other factors because giving this hormone to unstressed rats was not sufficient to trigger the changes seen in the stressed rats. Future experiments will investigate what these other stress factors are and how they work together with corticosterone. DOI:http://dx.doi.org/10.7554/eLife.15448.002
Collapse
Affiliation(s)
- Claire E Stelly
- Department of Neuroscience, University of Texas, Austin, United States.,Waggoner Center for Alcohol and Addiction Research, University of Texas, Austin, United States
| | - Matthew B Pomrenze
- Waggoner Center for Alcohol and Addiction Research, University of Texas, Austin, United States.,Division of Pharmacology and Toxicology, University of Texas, Austin, United States
| | - Jason B Cook
- Department of Neuroscience, University of Texas, Austin, United States.,Waggoner Center for Alcohol and Addiction Research, University of Texas, Austin, United States
| | - Hitoshi Morikawa
- Department of Neuroscience, University of Texas, Austin, United States.,Waggoner Center for Alcohol and Addiction Research, University of Texas, Austin, United States
| |
Collapse
|
44
|
Xie K, Fox GE, Liu J, Tsien JZ. 512-Channel and 13-Region Simultaneous Recordings Coupled with Optogenetic Manipulation in Freely Behaving Mice. Front Syst Neurosci 2016; 10:48. [PMID: 27378865 PMCID: PMC4905953 DOI: 10.3389/fnsys.2016.00048] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 05/23/2016] [Indexed: 01/19/2023] Open
Abstract
The development of technologies capable of recording both single-unit activity and local field potentials (LFPs) over a wide range of brain circuits in freely behaving animals is the key to constructing brain activity maps. Although mice are the most popular mammalian genetic model, in vivo neural recording has been traditionally limited to smaller channel count and fewer brain structures because of the mouse’s small size and thin skull. Here, we describe a 512-channel tetrode system that allows us to record simultaneously over a dozen cortical and subcortical structures in behaving mice. This new technique offers two major advantages – namely, the ultra-low cost and the do-it-yourself flexibility for targeting any combination of many brain areas. We show the successful recordings of both single units and LFPs from 13 distinct neural circuits of the mouse brain, including subregions of the anterior cingulate cortices, retrosplenial cortices, somatosensory cortices, secondary auditory cortex, hippocampal CA1, dentate gyrus, subiculum, lateral entorhinal cortex, perirhinal cortex, and prelimbic cortex. This 512-channel system can also be combined with Cre-lox neurogenetics and optogenetics to further examine interactions between genes, cell types, and circuit dynamics across a wide range of brain structures. Finally, we demonstrate that complex stimuli – such as an earthquake and fear-inducing foot-shock – trigger firing changes in all of the 13 brain regions recorded, supporting the notion that neural code is highly distributed. In addition, we show that localized optogenetic manipulation in any given brain region could disrupt network oscillations and caused changes in single-unit firing patterns in a brain-wide manner, thereby raising the cautionary note of the interpretation of optogenetically manipulated behaviors.
Collapse
Affiliation(s)
- Kun Xie
- Brain and Behavior Discovery Institute-Department of Neurology, Medical College of Georgia, Augusta University, Augusta GA, USA
| | - Grace E Fox
- Brain and Behavior Discovery Institute-Department of Neurology, Medical College of Georgia, Augusta University, Augusta GA, USA
| | - Jun Liu
- Brain and Behavior Discovery Institute-Department of Neurology, Medical College of Georgia, Augusta University, AugustaGA, USA; The Brain Decoding Center, Banna Biomedical Research Institute, Yunnan Academy of Science and TechnologyYunnan, China
| | - Joe Z Tsien
- Brain and Behavior Discovery Institute-Department of Neurology, Medical College of Georgia, Augusta University, Augusta GA, USA
| |
Collapse
|
45
|
Functional Connectome Analysis of Dopamine Neuron Glutamatergic Connections in Forebrain Regions. J Neurosci 2016; 35:16259-71. [PMID: 26658874 DOI: 10.1523/jneurosci.1674-15.2015] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED In the ventral tegmental area (VTA), a subpopulation of dopamine neurons express vesicular glutamate transporter 2 and make glutamatergic connections to nucleus accumbens (NAc) and olfactory tubercle (OT) neurons. However, their glutamatergic connections across the forebrain have not been explored systematically. To visualize dopamine neuron forebrain projections and to enable photostimulation of their axons independent of transmitter status, we virally transfected VTA neurons with channelrhodopsin-2 fused to enhanced yellow fluorescent protein (ChR2-EYFP) and used DAT(IREScre) mice to restrict expression to dopamine neurons. ChR2-EYFP-expressing neurons almost invariably stained for tyrosine hydroxylase, identifying them as dopaminergic. Dopamine neuron axons visualized by ChR2-EYFP fluorescence projected most densely to the striatum, moderately to the amygdala and entorhinal cortex (ERC), sparsely to prefrontal and cingulate cortices, and rarely to the hippocampus. Guided by ChR2-EYFP fluorescence, we recorded systematically from putative principal neurons in target areas and determined the incidence and strength of glutamatergic connections by activating all dopamine neuron terminals impinging on recorded neurons with wide-field photostimulation. This revealed strong glutamatergic connections in the NAc, OT, and ERC; moderate strength connections in the central amygdala; and weak connections in the cingulate cortex. No glutamatergic connections were found in the dorsal striatum, hippocampus, basolateral amygdala, or prefrontal cortex. These results indicate that VTA dopamine neurons elicit widespread, but regionally distinct, glutamatergic signals in the forebrain and begin to define the dopamine neuron excitatory functional connectome. SIGNIFICANCE STATEMENT Dopamine neurons are important for the control of motivated behavior and are involved in the pathophysiology of several major neuropsychiatric disorders. Recent studies have shown that some ventral midbrain dopamine neurons are capable of glutamate cotransmission. With conditional expression of channelrhodopsin in dopamine neurons, we systematically explored dopamine neuron connections in the forebrain and identified regionally specific dopamine neuron excitatory connections. Establishing that only a subset of forebrain regions receive excitatory connections from dopamine neurons will help to determine the function of dopamine neuron glutamate cotransmission, which likely involves transmission of precise temporal signals and enhancement of the dynamic range of dopamine neuron signals.
Collapse
|
46
|
Degoulet M, Stelly CE, Ahn KC, Morikawa H. L-type Ca²⁺ channel blockade with antihypertensive medication disrupts VTA synaptic plasticity and drug-associated contextual memory. Mol Psychiatry 2016; 21:394-402. [PMID: 26100537 PMCID: PMC4689680 DOI: 10.1038/mp.2015.84] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 04/14/2015] [Accepted: 04/30/2015] [Indexed: 02/08/2023]
Abstract
Drug addiction is driven, in part, by powerful and enduring memories of sensory cues associated with drug intake. As such, relapse to drug use during abstinence is frequently triggered by an encounter with drug-associated cues, including the drug itself. L-type Ca(2+) channels (LTCCs) are known to regulate different forms of synaptic plasticity, the major neural substrate for learning and memory, in various brain areas. Long-term potentiation (LTP) of NMDA receptor (NMDAR)-mediated glutamatergic transmission in the ventral tegmental area (VTA) may contribute to the increased motivational valence of drug-associated cues triggering relapse. In this study, using rat brain slices, we found that isradipine, a general LTCC antagonist used as antihypertensive medication, not only blocks the induction of NMDAR LTP but also promotes the reversal of previously induced LTP in the VTA. In behaving rats, isradipine injected into the VTA suppressed the acquisition of cocaine-paired contextual cue memory assessed using a conditioned place preference (CPP) paradigm. Furthermore, administration of isradipine or a CaV1.3 subtype-selective LTCC antagonist (systemic or intra-VTA) before a single extinction or reinstatement session, while having no immediate effect at the time of administration, abolished previously acquired cocaine and alcohol (ethanol) CPP on subsequent days. Notably, CPP thus extinguished cannot be reinstated by drug re-exposure, even after 2 weeks of withdrawal. These results suggest that LTCC blockade during exposure to drug-associated cues may cause unlearning of the increased valence of those cues, presumably via reversal of glutamatergic synaptic plasticity in the VTA.
Collapse
Affiliation(s)
- Mickael Degoulet
- Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | - Claire E. Stelly
- Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | - Kee-Chan Ahn
- Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | - Hitoshi Morikawa
- Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA,Corresponding author: Hitoshi Morikawa, Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, 2400 Speedway, Austin, TX 78712, USA., Tel: 1-512-232-9299, Fax: 1-512-471-3878,
| |
Collapse
|
47
|
Keiflin R, Janak PH. Dopamine Prediction Errors in Reward Learning and Addiction: From Theory to Neural Circuitry. Neuron 2016; 88:247-63. [PMID: 26494275 DOI: 10.1016/j.neuron.2015.08.037] [Citation(s) in RCA: 201] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Midbrain dopamine (DA) neurons are proposed to signal reward prediction error (RPE), a fundamental parameter in associative learning models. This RPE hypothesis provides a compelling theoretical framework for understanding DA function in reward learning and addiction. New studies support a causal role for DA-mediated RPE activity in promoting learning about natural reward; however, this question has not been explicitly tested in the context of drug addiction. In this review, we integrate theoretical models with experimental findings on the activity of DA systems, and on the causal role of specific neuronal projections and cell types, to provide a circuit-based framework for probing DA-RPE function in addiction. By examining error-encoding DA neurons in the neural network in which they are embedded, hypotheses regarding circuit-level adaptations that possibly contribute to pathological error signaling and addiction can be formulated and tested.
Collapse
Affiliation(s)
- Ronald Keiflin
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Patricia H Janak
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD 21218, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
48
|
Lee JC, Wang LP, Tsien JZ. Dopamine Rebound-Excitation Theory: Putting Brakes on PTSD. Front Psychiatry 2016; 7:163. [PMID: 27729874 PMCID: PMC5037223 DOI: 10.3389/fpsyt.2016.00163] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/14/2016] [Indexed: 01/19/2023] Open
Abstract
It is not uncommon for humans or animals to experience traumatic events in their lifetimes. However, the majority of individuals are resilient to long-term detrimental changes turning into anxiety and depression, such as post-traumatic stress disorder (PTSD). What underlying neural mechanism accounts for individual variability in stress resilience? Hyperactivity in fear circuits, such as the amygdalar system, is well-known to be the major pathophysiological basis for PTSD, much like a "stuck accelerator." Interestingly, increasing evidence demonstrates that dopamine (DA) - traditionally known for its role in motivation, reward prediction, and addiction - is also crucial in regulating fear learning and anxiety. Yet, how dopaminergic (DAergic) neurons control stress resilience is unclear, especially given that DAergic neurons have multiple subtypes with distinct temporal dynamics. Here, we propose the Rebound-Excitation Theory, which posits that DAergic neurons' rebound-excitation at the termination of fearful experiences serves as an important "brake" by providing intrinsic safety-signals to fear-processing neural circuits in a spatially and temporally controlled manner. We discuss how DAergic neuron rebound-excitation may be regulated by genetics and experiences, and how such physiological properties may be used as a brain-activity biomarker to predict and confer individual resilience to stress and anxiety.
Collapse
Affiliation(s)
- Jason C Lee
- Department of Neurology, Brain and Behavior Discovery Institute, Medical College of Georgia, Augusta University , Augusta, GA , USA
| | - Lei Philip Wang
- Department of Neurology, Brain and Behavior Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, USA; Department of Psychiatry, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Joe Z Tsien
- Department of Neurology, Brain and Behavior Discovery Institute, Medical College of Georgia, Augusta University , Augusta, GA , USA
| |
Collapse
|
49
|
Luo SX, Huang EJ. Dopaminergic Neurons and Brain Reward Pathways: From Neurogenesis to Circuit Assembly. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 186:478-88. [PMID: 26724386 DOI: 10.1016/j.ajpath.2015.09.023] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/01/2015] [Accepted: 09/23/2015] [Indexed: 11/26/2022]
Abstract
Midbrain dopaminergic (DA) neurons in the substantia nigra pars compacta and ventral tegmental area regulate extrapyramidal movement and important cognitive functions, including motivation, reward associations, and habit learning. Dysfunctions in DA neuron circuitry have been implicated in several neuropsychiatric disorders, including addiction and schizophrenia, whereas selective degeneration of DA neurons in substantia nigra pars compacta is a key neuropathological feature in Parkinson disease. Efforts to understand these disorders have focused on dissecting the underlying causes, as well as developing therapeutic strategies to replenish dopamine deficiency. In particular, the promise of cell replacement therapies for clinical intervention has led to extensive research in the identification of mechanisms involved in DA neuron development. It is hoped that a comprehensive understanding of these mechanisms will lead to therapeutic strategies that improve the efficiency of DA neuron production, engraftment, and function. This review provides a comprehensive discussion on how Wnt/β-catenin and sonic hedgehog-Smoothened signaling mechanisms control the specification and expansion of DA progenitors and the differentiation of DA neurons. We also discuss how mechanisms involving transforming growth factor-β and transcriptional cofactor homeodomain interacting protein kinase 2 regulate the survival and maturation of DA neurons in early postnatal life. These results not only reveal fundamental mechanisms regulating DA neuron development, but also provide important insights to their potential contributions to neuropsychiatric and neurodegenerative diseases.
Collapse
Affiliation(s)
- Sarah X Luo
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, California; Department of Pathology, University of California San Francisco, San Francisco, California
| | - Eric J Huang
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, California; Department of Pathology, University of California San Francisco, San Francisco, California; Pathology Service 113B, San Francisco Veterans Affairs Medical Center, San Francisco, California.
| |
Collapse
|
50
|
Sommer S, Hauber W. N-methyl-D-aspartate receptors in the ventral tegmental area mediate the excitatory influence of Pavlovian stimuli on instrumental performance. Brain Struct Funct 2015; 221:4399-4409. [PMID: 26691586 DOI: 10.1007/s00429-015-1170-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/07/2015] [Indexed: 10/22/2022]
Abstract
Pavlovian stimuli predictive of food can markedly amplify instrumental responding for food. This effect is termed Pavlovian-instrumental transfer (PIT). The ventral tegmental area (VTA) plays a key role in mediating PIT, however, it is yet unknown whether N-methyl-D-aspartate (NMDA)-type glutamate receptors in the VTA are involved in PIT. Here, we examined the effects of an NMDA-receptor blockade in the VTA on PIT. Immediately prior to PIT testing, rats were subjected to intra-VTA infusions of vehicle or of the NMDA-receptor antagonist 2-amino-5-phosphonopentanoic acid (AP-5) (1, 5 µg/side). In rats that received AP-5 at the lower dose, the PIT effect was intact, i.e. presentation of the Pavlovian stimulus enhanced instrumental responding. By contrast, in rats that received AP-5 at the higher dose, the PIT effect was blocked. The data suggest that NMDA receptors in the VTA mediate the activating effects of Pavlovian stimuli on instrumental responding.
Collapse
Affiliation(s)
- Susanne Sommer
- Department Animal Physiology, University of Stuttgart, Pfaffenwaldring 57, 70550, Stuttgart, Germany
| | - Wolfgang Hauber
- Department Animal Physiology, University of Stuttgart, Pfaffenwaldring 57, 70550, Stuttgart, Germany.
| |
Collapse
|