51
|
Dias FP, Carvalho Crespo LGS, Leite Junior JB, Samuels RI, Coimbra NC, Carey RJ, Carrera MP. Morphine reward effects and morphine behavioral sensitization: The adventitious association of morphine activation of brain reward effects with ongoing spontaneous activity. Pharmacol Biochem Behav 2021; 209:173244. [PMID: 34363828 DOI: 10.1016/j.pbb.2021.173244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 07/22/2021] [Accepted: 07/30/2021] [Indexed: 12/01/2022]
Abstract
The development of sensitization is one of the hallmarks of addictive drugs such as morphine. We administered morphine (10 mg/kg; MOR) to induce locomotor sensitization and ERK activation in the VTA and NAc. In the first experiment, four groups of rats received five daily 30 min sessions in an open-field, and locomotion was measured. For the first four sessions, one group received MOR pre-test (MOR-P); a second group received vehicle pre-test (MOR-UP) and MOR 30 min post-test; the remaining 2 groups received vehicle (VEH) pre-test. On the fifth session, the MOR-P, MOR-UP, and one VEH group received MOR pre-test and the remaining VEH group received VEH. Sensitization emerged in the first 5 min and progressed over to the second and third 5 min blocks only in the MOR-P group. For the second experiment, 4 groups received MOR and 4 groups VEH, and were then returned to their home cage and after 5, 15, 30 or 60 min post-injection, were euthanized for ERK measurements in VTA and NAc. ERK activation increased and peaked at 5 min post injection in the MOR group and then declined to VEH levels by 30 min. Another two groups received either MOR or VEH immediately before a 5 min arena test and ERK was measured immediately post-test. MOR had no effect on locomotion but increased ERK in the VTA and NAc. The peak ERK activation in VTA reflected activation of reward systems by morphine that reinforced locomotor behavior and with repeated treatments, induced a sensitization effect.
Collapse
Affiliation(s)
- Fabiolla Patusco Dias
- Behavioral Pharmacology Group, Laboratory of Animal Morphology and Pathology, State University of North Fluminense Darcy Ribeiro, Avenida Alberto Lamego, 2000, Campos dos Goytacazes 28013-602, RJ, Brazil
| | - Luiz Gustavo Soares Carvalho Crespo
- Behavioral Pharmacology Group, Laboratory of Animal Morphology and Pathology, State University of North Fluminense Darcy Ribeiro, Avenida Alberto Lamego, 2000, Campos dos Goytacazes 28013-602, RJ, Brazil
| | - Joaquim Barbosa Leite Junior
- Behavioral Pharmacology Group, Laboratory of Animal Morphology and Pathology, State University of North Fluminense Darcy Ribeiro, Avenida Alberto Lamego, 2000, Campos dos Goytacazes 28013-602, RJ, Brazil
| | - Richard Ian Samuels
- Department of Entomology and Plant Pathology, State University of North Fluminense Darcy Ribeiro, Campos dos Goytacazes, RJ, Brazil
| | - Norberto Cysne Coimbra
- Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo, SP, Brazil
| | - Robert J Carey
- Department of Psychiatry SUNY Upstate Medical University, 800 Irving Avenue, Syracuse, NY 13210, USA
| | - Marinete Pinheiro Carrera
- Behavioral Pharmacology Group, Laboratory of Animal Morphology and Pathology, State University of North Fluminense Darcy Ribeiro, Avenida Alberto Lamego, 2000, Campos dos Goytacazes 28013-602, RJ, Brazil.
| |
Collapse
|
52
|
Nakamura Y, Longueville S, Nishi A, Hervé D, Girault JA, Nakamura Y. Dopamine D1 receptor-expressing neurons activity is essential for locomotor and sensitizing effects of a single injection of cocaine. Eur J Neurosci 2021; 54:5327-5340. [PMID: 34273137 DOI: 10.1111/ejn.15394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 07/05/2021] [Accepted: 07/13/2021] [Indexed: 11/29/2022]
Abstract
Dopamine D1 receptors play an important role in the effects of cocaine. Here, we investigated the role of neurons which express these receptors (D1-neurons) in the acute locomotor effects of cocaine and the locomotor sensitization observed after a second injection of this drug, using the previously established two-injection protocol of sensitization. We inhibited D1-neurons using double transgenic mice conditionally expressing the inhibitory Gi-coupled designer receptor exclusively activated by designer drugs (Gi-DREADD) in D1-neurons. Chemogenetic inhibition of D1-neurons by a low dose of clozapine (0.1 mg/kg) decreased the cocaine-induced expression of Fos in striatal neurons. It diminished the basal locomotor activity and acute hyper-locomotion induced by cocaine (20 mg/kg). Clozapine 0.1 mg/kg had no effect by itself and did not alter cocaine effects in wild-type mice. Inhibition of D1-neurons during the first cocaine administration prevented the sensitization of the locomotor response in response to a second cocaine administration 10 days later. On Day 11, inhibition of D1-neurons by clozapine stimulation of Gi-DREADD blocked cocaine-induced locomotion including in sensitized mice, whereas on Day 12, in the absence of clozapine and D1-neurons inhibition, all mice displayed a sensitized response to cocaine. These results show that chemogenetic inhibition of D1-neurons decreases spontaneous and cocaine-induced locomotor activity. It prevents sensitization induction and blocks sensitized locomotion in a two-injection protocol of sensitization but does not reverse established sensitization. Our study further supports the central role of D1-neurons in mediating the acute locomotor effects of cocaine and its sensitization.
Collapse
Affiliation(s)
- Yukari Nakamura
- INSERM UMR-S 1270, Paris, France.,Faculty of Sciences and Engineering, Sorbonne University, Paris, France.,Institut du Fer à Moulin, Paris, France.,Department of Pharmacology, Kurume University School of Medicine, Kurume, Japan
| | - Sophie Longueville
- INSERM UMR-S 1270, Paris, France.,Faculty of Sciences and Engineering, Sorbonne University, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Akinori Nishi
- Department of Pharmacology, Kurume University School of Medicine, Kurume, Japan
| | - Denis Hervé
- INSERM UMR-S 1270, Paris, France.,Faculty of Sciences and Engineering, Sorbonne University, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Jean-Antoine Girault
- INSERM UMR-S 1270, Paris, France.,Faculty of Sciences and Engineering, Sorbonne University, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Yuki Nakamura
- INSERM UMR-S 1270, Paris, France.,Faculty of Sciences and Engineering, Sorbonne University, Paris, France.,Institut du Fer à Moulin, Paris, France
| |
Collapse
|
53
|
Liu L, Liu M, Zhao W, Zhao YL, Wang Y. Levo-tetrahydropalmatine: A new potential medication for methamphetamine addiction and neurotoxicity. Exp Neurol 2021; 344:113809. [PMID: 34256045 DOI: 10.1016/j.expneurol.2021.113809] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/23/2021] [Accepted: 07/08/2021] [Indexed: 10/20/2022]
Abstract
Levo-tetrahydropalmatine (l-THP) is mainly derived from the dried tuber of the Papaveraceae plant Corydalis, also called Corydalis B, which is a drug with analgesic, hypnotic, sedative and other effects. Methamphetamine (METH) belongs to the central nervous stimulant and is a highly addictive drug. It is an urgent problem to study the mechanism of methamphetamine neurotoxicity and to search for the therapeutic targets of the METH addiction. This review is aimed to discuss the pharmacological mechanism and the protective effects of l-THP on METH-induced neurotoxicity, and to explore the therapeutic prospects of l-THP for METH addiction to provide an innovative application of l-THP in clinic. It was found that exposure to METH leads to the compulsive drug-seeking and drug-taking behavior, which is ultimately resulted in METH addiction and neurotoxicity. L-THP has the inhibitory effects on the incidence, maintenance and relapse of METH addiction. L-THP can effectively enhance the plasticity of nerve cells and improve the function of nerve cells where brain-derived neurotrophic factor (BDNF) and its pathways play a protective role. Therefore, l-THP has the potential to become an important therapeutic drug for METH addiction and neurotoxicity.
Collapse
Affiliation(s)
- Lian Liu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Ming Liu
- Department of Drug Control, Criminal Investigation Police University of China, Shenyang, Liaoning 110854, PR China
| | - Wei Zhao
- Department of Drug Control, Criminal Investigation Police University of China, Shenyang, Liaoning 110854, PR China
| | - Yuan-Ling Zhao
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Yun Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, PR China.
| |
Collapse
|
54
|
Cutando L, Puighermanal E, Castell L, Tarot P, Bertaso F, Bonnavion P, Kerchove d'Exaerde A, Isingrini E, Galante M, Dallerac G, Pascoli V, Lüscher C, Giros B, Valjent E. Regulation of GluA1 phosphorylation by d-amphetamine and methylphenidate in the cerebellum. Addict Biol 2021; 26:e12995. [PMID: 33368923 DOI: 10.1111/adb.12995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/21/2020] [Accepted: 11/13/2020] [Indexed: 01/14/2023]
Abstract
Prescription stimulants, such as d-amphetamine or methylphenidate are used to treat suffering from attention-deficit hyperactivity disorder (ADHD). They potently release dopamine (DA) and norepinephrine (NE) and cause phosphorylation of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit GluA1 in the striatum. Whether other brain regions are also affected remains elusive. Here, we demonstrate that d-amphetamine and methylphenidate increase phosphorylation at Ser845 (pS845-GluA1) in the membrane fraction of mouse cerebellum homogenate. We identify Bergmann glial cells as the source of pS845-GluA1 and demonstrate a requirement for intact NE release. Consequently, d-amphetamine-induced pS845-GluA1 was prevented by β1-adenoreceptor antagonist, whereas the blockade of DA D1 receptor had no effect. Together, these results indicate that NE regulates GluA1 phosphorylation in Bergmann glial cells in response to prescription stimulants.
Collapse
Affiliation(s)
- Laura Cutando
- IGF University of Montpellier, CNRS, Inserm Montpellier France
| | - Emma Puighermanal
- IGF University of Montpellier, CNRS, Inserm Montpellier France
- Neurosciences Institute, Department of Cell Biology, Physiology and Immunology Autonomous University of Barcelona Barcelona Spain
| | - Laia Castell
- IGF University of Montpellier, CNRS, Inserm Montpellier France
| | - Pauline Tarot
- IGF University of Montpellier, CNRS, Inserm Montpellier France
| | | | - Patricia Bonnavion
- Laboratory of Neurophysiology, ULB Neuroscience Institute Université Libre de Bruxelles (ULB) Brussels Belgium
| | - Alban Kerchove d'Exaerde
- Laboratory of Neurophysiology, ULB Neuroscience Institute Université Libre de Bruxelles (ULB) Brussels Belgium
| | - Elsa Isingrini
- Integrative Neuroscience and Cognition Center University of Paris, CNRS Paris France
- Department of Psychiatry, Douglas Hospital McGill University Montreal Quebec Canada
| | - Micaela Galante
- Pharmacologie et Biochimie de la Synapse, Institut des Neurosciences Paris‐Saclay University of Paris‐Saclay, University of Paris‐Sud, CNRS, UMR Orsay France
| | - Glenn Dallerac
- Pharmacologie et Biochimie de la Synapse, Institut des Neurosciences Paris‐Saclay University of Paris‐Saclay, University of Paris‐Sud, CNRS, UMR Orsay France
| | - Vincent Pascoli
- Department of Basic Neurosciences, Medical Faculty University of Geneva Geneva Switzerland
| | - Christian Lüscher
- Department of Basic Neurosciences, Medical Faculty University of Geneva Geneva Switzerland
| | - Bruno Giros
- Integrative Neuroscience and Cognition Center University of Paris, CNRS Paris France
- Department of Psychiatry, Douglas Hospital McGill University Montreal Quebec Canada
| | | |
Collapse
|
55
|
Mahaman YAR, Huang F, Embaye KS, Wang X, Zhu F. The Implication of STEP in Synaptic Plasticity and Cognitive Impairments in Alzheimer's Disease and Other Neurological Disorders. Front Cell Dev Biol 2021; 9:680118. [PMID: 34195199 PMCID: PMC8236946 DOI: 10.3389/fcell.2021.680118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 05/06/2021] [Indexed: 12/31/2022] Open
Abstract
STriatal-Enriched protein tyrosine Phosphatase (STEP) is a tyrosine phosphatase that has been implicated in Alzheimer’s disease (AD), the most common form of dementia, and many other neurological diseases. The protein level and activity of STEP have been found to be elevated in most of these disorders, and specifically in AD as a result of dysregulation of different pathways including PP2B/DARPP32/PP1, PKA as well as impairments of both proteasomal and lysosomal systems. The upregulation in STEP leads to increased binding to, and dephosphorylation of, its substrates which are mainly found to be synaptic plasticity and thus learning and memory related proteins. These proteins include kinases like Fyn, Pyk2, ERK1/2 and both NMDA and AMPA receptor subunits GluN2B and GluA2. The dephosphorylation of these molecules results in inactivation of these kinases and internalization of NMDA and AMPA receptor complexes leading to synapse loss and cognitive impairments. In this study, we aim to review STEP regulation and its implications in AD as well as other neurological disorders and then summarize data on targeting STEP as therapeutic strategy in these diseases.
Collapse
Affiliation(s)
- Yacoubou Abdoul Razak Mahaman
- Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital, Shenzhen University, Shenzhen, China.,Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Huang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kidane Siele Embaye
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaochuan Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Feiqi Zhu
- Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital, Shenzhen University, Shenzhen, China
| |
Collapse
|
56
|
Lin R, Learman LN, Na CH, Renuse S, Chen KT, Chen PY, Lee GH, Xiao B, Resnick SM, Troncoso JC, Szumlinski KK, Linden DJ, Park JM, Savonenko A, Pandey A, Worley PF. Persistently Elevated mTOR Complex 1-S6 Kinase 1 Disrupts DARPP-32-Dependent D 1 Dopamine Receptor Signaling and Behaviors. Biol Psychiatry 2021; 89:1058-1072. [PMID: 33353667 PMCID: PMC8076344 DOI: 10.1016/j.biopsych.2020.10.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/17/2020] [Accepted: 10/22/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND The serine-threonine kinase mTORC1 (mechanistic target of rapamycin complex 1) is essential for normal cell function but is aberrantly activated in the brain in both genetic-developmental and sporadic diseases and is associated with a spectrum of neuropsychiatric symptoms. The underlying molecular mechanisms of cognitive and neuropsychiatric symptoms remain controversial. METHODS The present study examines behaviors in transgenic models that express Rheb, the most proximal known activator of mTORC1, and profiles striatal phosphoproteomics in a model with persistently elevated mTORC1 signaling. Biochemistry, immunohistochemistry, electrophysiology, and behavior approaches are used to examine the impact of persistently elevated mTORC1 on D1 dopamine receptor (D1R) signaling. The effect of persistently elevated mTORC1 was confirmed using D1-Cre to elevate mTORC1 activity in D1R neurons. RESULTS We report that persistently elevated mTORC1 signaling blocks canonical D1R signaling that is dependent on DARPP-32 (dopamine- and cAMP-regulated neuronal phosphoprotein). The immediate downstream effector of mTORC1, ribosomal S6 kinase 1 (S6K1), phosphorylates and activates DARPP-32. Persistent elevation of mTORC1-S6K1 occludes dynamic D1R signaling downstream of DARPP-32 and blocks multiple D1R responses, including dynamic gene expression, D1R-dependent corticostriatal plasticity, and D1R behavioral responses including sociability. Candidate biomarkers of mTORC1-DARPP-32 occlusion are increased in the brain of human disease subjects in association with elevated mTORC1-S6K1, supporting a role for this mechanism in cognitive disease. CONCLUSIONS The mTORC1-S6K1 intersection with D1R signaling provides a molecular framework to understand the effects of pathological mTORC1 activation on behavioral symptoms in neuropsychiatric disease.
Collapse
Affiliation(s)
- Raozhou Lin
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Lisa N. Learman
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Chan-Hyun Na
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Santosh Renuse
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First ST SW, Rochester, MN 55905, USA.,Center for Individualized Medicine, Mayo Clinic, 200 First ST SW, Rochester, MN 55905, USA
| | - Kevin T. Chen
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Po Yu Chen
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Gum-Hwa Lee
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Bo Xiao
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Susan M. Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD 21224, USA
| | - Juan C. Troncoso
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Karen K. Szumlinski
- Department of Psychological and Brain Sciences and the Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA
| | - David J. Linden
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Joo-Min Park
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Republic of Korea
| | - Alena Savonenko
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First ST SW, Rochester, MN 55905, USA.,Center for Individualized Medicine, Mayo Clinic, 200 First ST SW, Rochester, MN 55905, USA
| | - Paul F. Worley
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Corresponding author. Phone: 410-502-5489
| |
Collapse
|
57
|
ERK1/2 kinases and dopamine D2 receptors participate in the anticonvulsant effects of a new derivative of benzoylpyridine oxime and valproic acid. Eur J Pharmacol 2021; 903:174150. [PMID: 33961874 DOI: 10.1016/j.ejphar.2021.174150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/26/2021] [Accepted: 04/30/2021] [Indexed: 01/11/2023]
Abstract
Inhibition of the activity of extracellular signal-regulated kinases (ERK1/2) induced by the activation of the dopamine D2 receptor signalling cascade may be a promising pharmacological target. The aim of this work was to study the involvement of ERK1/2 and dopamine D2 receptor in the mechanism of the anticonvulsant action of valproic acid (VA) and a new benzoylpyridine oxime derivative (GIZH-298), which showed antiepileptic activity in different models of epilepsy. We showed that subchronic exposure to maximal electroshock seizures (MES) for 5 days reduced the density of dopamine D2 receptors in the striatum of mice. GIZH-298 counteracted the decrease in the number of dopamine D2 receptors associated with MES and increased the number of ligand binding sites of dopamine D2 receptors in mice without MES. The affinity of dopamine D2 receptors to the ligand was not changed by GIZH-298. MES caused an increase in ERK1/2 and synapsin I phosphorylation in the striatum while GIZH-298, similar to VA, reduced the levels of both phospho-ERK1/2 and phosphosynapsin I after MES, which correlated with the decrease in the intensity of seizure in mice. In addition, GIZH-298 suppressed ERK1/2 phosphorylation in SH-SY5Y human neuroblastoma cells at therapeutic concentrations, while VA inhibited ERK1/2 phosphorylation in vivo but not in vitro. The data obtained expand the understanding of the mechanisms of action of VA and GIZH-298, which involve regulating the activity of ERK1/2 kinases, probably by modulating dopamine D2 receptors in limbic structures, as well as (in the case of GIZH-298) directly inhibiting of the ERK1/2 cascade.
Collapse
|
58
|
Yu W, Pati D, Pina MM, Schmidt KT, Boyt KM, Hunker AC, Zweifel LS, McElligott ZA, Kash TL. Periaqueductal gray/dorsal raphe dopamine neurons contribute to sex differences in pain-related behaviors. Neuron 2021; 109:1365-1380.e5. [PMID: 33740416 PMCID: PMC9990825 DOI: 10.1016/j.neuron.2021.03.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/02/2021] [Accepted: 02/26/2021] [Indexed: 02/06/2023]
Abstract
Sex differences in pain severity, response, and pathological susceptibility are widely reported, but the neural mechanisms that contribute to these outcomes remain poorly understood. Here we show that dopamine (DA) neurons in the ventrolateral periaqueductal gray/dorsal raphe (vlPAG/DR) differentially regulate pain-related behaviors in male and female mice through projections to the bed nucleus of the stria terminalis (BNST). We find that activation of vlPAG/DRDA+ neurons or vlPAG/DRDA+ terminals in the BNST reduces nociceptive sensitivity during naive and inflammatory pain states in male mice, whereas activation of this pathway in female mice leads to increased locomotion in the presence of salient stimuli. We additionally use slice physiology and genetic editing approaches to demonstrate that vlPAG/DRDA+ projections to the BNST drive sex-specific responses to pain through DA signaling, providing evidence of a novel ascending circuit for pain relief in males and contextual locomotor response in females.
Collapse
Affiliation(s)
- Waylin Yu
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Dipanwita Pati
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Melanie M Pina
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Karl T Schmidt
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kristen M Boyt
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Avery C Hunker
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Larry S Zweifel
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA; Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA
| | - Zoe A McElligott
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Thomas L Kash
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
59
|
Domenici MR, Mallozzi C, Pepponi R, Casella I, Chiodi V, Ferrante A, Popoli P. Insight into the Role of the STriatal-Enriched Protein Tyrosine Phosphatase (STEP) in A 2A Receptor-Mediated Effects in the Central Nervous System. Front Pharmacol 2021; 12:647742. [PMID: 33953681 PMCID: PMC8090931 DOI: 10.3389/fphar.2021.647742] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/26/2021] [Indexed: 12/18/2022] Open
Abstract
The STriatal-Enriched protein tyrosine phosphatase STEP is a brain-specific tyrosine phosphatase that plays a pivotal role in the mechanisms of learning and memory, and it has been demonstrated to be involved in several neuropsychiatric diseases. Recently, we found a functional interaction between STEP and adenosine A2A receptor (A2AR), a subtype of the adenosine receptor family widely expressed in the central nervous system, where it regulates motor behavior and cognition, and plays a role in cell survival and neurodegeneration. Specifically, we demonstrated the involvement of STEP in A2AR-mediated cocaine effects in the striatum and, more recently, we found that in the rat striatum and hippocampus, as well as in a neuroblastoma cell line, the overexpression of the A2AR, or its stimulation, results in an increase in STEP activity. In the present article we will discuss the functional implication of this interaction, trying to examine the possible mechanisms involved in this relation between STEP and A2ARs.
Collapse
Affiliation(s)
- Maria Rosaria Domenici
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanitá, Rome, Italy
| | - Cinzia Mallozzi
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | - Rita Pepponi
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanitá, Rome, Italy
| | - Ida Casella
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanitá, Rome, Italy
| | - Valentina Chiodi
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanitá, Rome, Italy
| | - Antonella Ferrante
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanitá, Rome, Italy
| | - Patrizia Popoli
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanitá, Rome, Italy
| |
Collapse
|
60
|
Crittenden JR, Gipson TA, Smith AC, Bowden HA, Yildirim F, Fischer KB, Yim M, Housman DE, Graybiel AM. Striatal transcriptome changes linked to drug-induced repetitive behaviors. Eur J Neurosci 2021; 53:2450-2468. [PMID: 33759265 DOI: 10.1111/ejn.15116] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/23/2020] [Accepted: 01/09/2021] [Indexed: 11/30/2022]
Abstract
Disruptive or excessive repetitive motor patterns (stereotypies) are cardinal symptoms in numerous neuropsychiatric disorders. Stereotypies are also evoked by psychomotor stimulants such as amphetamine. The acquisition of motor sequences is paralleled by changes in activity patterns in the striatum, and stereotypies have been linked to abnormal plasticity in these reinforcement-related circuits. Here, we designed experiments in mice to identify transcriptomic changes that underlie striatal plasticity occurring alongside the development of drug-induced stereotypic behavior. We identified three schedules of amphetamine treatment inducing different degrees of stereotypy and used bulk RNAseq to compare striatal gene expression changes among groups of mice treated with the different drug-dose schedules and vehicle-treated, cage-mate controls. Mice were identified as naïve, sensitized, or tolerant to drug-induced stereotypy. All drug-treated groups exhibited expression changes in genes that encode members of the extracellular signal-regulated kinase (ERK) cascades known to regulate psychomotor stimulant responses. In the sensitized group with the most prolonged stereotypy, we found dysregulation of 20 genes that were not changed in other groups. Gene set enrichment analysis indicated highly significant overlap with genes regulated by neuregulin 1 (Nrg1). Nrg1 is known to be a schizophrenia and autism susceptibility gene that encodes a ligand for Erb-B receptors, which are involved in neuronal migration, myelination, and cell survival, including that of dopamine-containing neurons. Stimulant abuse is a risk factor for schizophrenia onset, and these two disorders share behavioral stereotypy phenotypes. Our results raise the possibility that drug-induced sensitization of the Nrg1 signaling pathway might underlie these links.
Collapse
Affiliation(s)
- Jill R Crittenden
- McGovern Institute for Brain Research, The Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Brain and Cognitive Sciences, The Massachusetts Institute of Technology, Cambridge, MA, USA.,Institute for Integrative Cancer Research, The Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Theresa A Gipson
- Institute for Integrative Cancer Research, The Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anne C Smith
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA
| | - Hilary A Bowden
- McGovern Institute for Brain Research, The Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Brain and Cognitive Sciences, The Massachusetts Institute of Technology, Cambridge, MA, USA.,Institute for Integrative Cancer Research, The Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ferah Yildirim
- Department of Neuropsychiatry, Department of Psychiatry and Psychotherapy, and NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Kyle B Fischer
- McGovern Institute for Brain Research, The Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Brain and Cognitive Sciences, The Massachusetts Institute of Technology, Cambridge, MA, USA.,Institute for Integrative Cancer Research, The Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Michael Yim
- McGovern Institute for Brain Research, The Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Brain and Cognitive Sciences, The Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David E Housman
- Institute for Integrative Cancer Research, The Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ann M Graybiel
- McGovern Institute for Brain Research, The Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Brain and Cognitive Sciences, The Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
61
|
The Role of CaMKII and ERK Signaling in Addiction. Int J Mol Sci 2021; 22:ijms22063189. [PMID: 33804804 PMCID: PMC8004038 DOI: 10.3390/ijms22063189] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
Nicotine is the predominant addictive compound of tobacco and causes the acquisition of dependence through its interactions with nicotinic acetylcholine receptors and various neurotransmitter releases in the central nervous system. The Ca2+/calmodulin-dependent protein kinase II (CaMKII) and extracellular signal-regulated kinase (ERK) play a pivotal role in synaptic plasticity in the hippocampus. CaMKII is involved in long-term potentiation induction, which underlies the consolidation of learning and memory; however, the roles of CaMKII in nicotine and other psychostimulant-induced addiction still require further investigation. This article reviews the molecular mechanisms and crucial roles of CaMKII and ERK in nicotine and other stimulant drug-induced addiction. We also discuss dopamine (DA) receptor signaling involved in nicotine-induced addiction in the brain reward circuitry. In the last section, we introduce the association of polyunsaturated fatty acids and cellular chaperones of fatty acid-binding protein 3 in the context of nicotine-induced addiction in the mouse nucleus accumbens and provide a novel target for the treatment of drug abuse affecting dopaminergic systems.
Collapse
|
62
|
Porru S, López-Cruz L, Carratalá-Ros C, Salamone JD, Acquas E, Correa M. Impact of Caffeine on Ethanol-Induced Stimulation and Sensitization: Changes in ERK and DARPP-32 Phosphorylation in Nucleus Accumbens. Alcohol Clin Exp Res 2021; 45:608-619. [PMID: 33471948 DOI: 10.1111/acer.14553] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 01/05/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND Caffeine is frequently consumed with ethanol to reduce the impairing effects induced by ethanol, including psychomotor slowing or incoordination. Both drugs modulate dopamine (DA)-related markers in accumbens (Acb), and Acb DA is involved in voluntary locomotion and locomotor sensitization. The present study determined whether caffeine can affect locomotion induced by acute and repeated ethanol administration in adult male CD-1 mice. METHODS Acute administration of caffeine (7.5 to 30.0 mg/kg) was evaluated for its effects on acute ethanol-induced (1.5 to 3.5 g/kg) changes in open-field horizontal locomotion, supported rearing, and rearing not supported by the wall. DA receptor-dependent phosphorylation markers were assessed: extracellular signal-regulated kinase (pERK), and dopamine-and cAMP-regulated phosphoprotein Mr32kDa phosphorylated at threonine 75 site (pDARPP-32-Thr75) in Acb core and shell. Acutely administered caffeine was also evaluated in ethanol-sensitized (1.5 g/kg) mice. RESULTS Acute ethanol decreased both types of rearing. Caffeine increased supported rearing but did not block ethanol -induced decreases in rearing. Both substances increased horizontal locomotion in a biphasic manner, and caffeine potentiated ethanol-induced locomotion. Although ethanol administered repeatedly induced sensitization of locomotion and unsupported rearing, acute administration of caffeine to ethanol-sensitized mice in an ethanol-free state resulted in blunted stimulant effects compared with those seen in ethanol-naïve mice. Ethanol increased pERK immunoreactivity in both subregions of the Acb, but coadministration with caffeine blunted this increase. There were no effects on pDARPP-32(Thr75) immunoreactivity. CONCLUSIONS The present results demonstrated that, after the first administration, caffeine potentiated the stimulating actions of ethanol, but did not counteract its suppressant or ataxic effects. Moreover, our results show that caffeine has less activating effects in ethanol-sensitized animals.
Collapse
Affiliation(s)
- Simona Porru
- Àrea de Psicobiologia, Campus de Riu Sec, Universitat Jaume I, Castelló, Spain.,Department of Life and Environmental Sciences, University of Cagliari, Monserrato, Italy
| | - Laura López-Cruz
- Àrea de Psicobiologia, Campus de Riu Sec, Universitat Jaume I, Castelló, Spain
| | - Carla Carratalá-Ros
- Àrea de Psicobiologia, Campus de Riu Sec, Universitat Jaume I, Castelló, Spain
| | - John D Salamone
- Department of Psychology, University of Connecticut, Storrs, Connecticut, USA
| | - Elio Acquas
- Department of Life and Environmental Sciences, University of Cagliari, Monserrato, Italy
| | - Mercè Correa
- Àrea de Psicobiologia, Campus de Riu Sec, Universitat Jaume I, Castelló, Spain
| |
Collapse
|
63
|
Melis C, Beauvais G, Muntean BS, Cirnaru MD, Otrimski G, Creus-Muncunill J, Martemyanov KA, Gonzalez-Alegre P, Ehrlich ME. Striatal Dopamine Induced ERK Phosphorylation Is Altered in Mouse Models of Monogenic Dystonia. Mov Disord 2021; 36:1147-1157. [PMID: 33458877 DOI: 10.1002/mds.28476] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Similar to some monogenic forms of dystonia, levodopa-induced dyskinesia is a hyperkinetic movement disorder with abnormal nigrostriatal dopaminergic neurotransmission. Molecularly, it is characterized by hyper-induction of phosphorylation of extracellular signal-related kinase in response to dopamine in medium spiny neurons of the direct pathway. OBJECTIVES The objective of this study was to determine if mouse models of monogenic dystonia exhibit molecular features of levodopa-induced dyskinesia. METHODS Western blotting and quantitative immunofluorescence was used to assay baseline and/or dopamine-induced levels of the phosphorylated kinase in the striatum in mouse models of DYT1, DYT6, and DYT25 expressing a reporter in dopamine D1 receptor-expressing projection neurons. Cyclic adenosine monophosphate (cAMP) immunoassay and adenylyl cyclase activity assays were also performed. RESULTS In DYT1 and DYT6 models, blocking dopamine reuptake with cocaine leads to enhanced extracellular signal-related kinase phosphorylation in dorsomedial striatal medium spiny neurons in the direct pathway, which is abolished by pretreatment with the N-methyl-d-aspartate antagonist MK-801. Phosphorylation is decreased in a model of DYT25. Levels of basal and stimulated cAMP and adenylyl cyclase activity were normal in the DYT1 and DYT6 mice and decreased in the DYT25 mice. Oxotremorine induced increased abnormal movements in the DYT1 knock-in mice. CONCLUSIONS The increased dopamine induction of extracellular signal-related kinase phosphorylation in 2 genetic types of dystonia, similar to what occurs in levodopa-induced dyskinesia, and its decrease in a third, suggests that abnormal signal transduction in response to dopamine in the postsynaptic nigrostriatal pathway might be a point of convergence for dystonia and other hyperkinetic movement disorders, potentially offering common therapeutic targets. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Chiara Melis
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Genevieve Beauvais
- Raymond G. Perelman Center for Cellular and Molecular Therapy, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Brian S Muntean
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, USA
| | - Maria-Daniela Cirnaru
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Garrett Otrimski
- Raymond G. Perelman Center for Cellular and Molecular Therapy, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jordi Creus-Muncunill
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, USA
| | - Pedro Gonzalez-Alegre
- Raymond G. Perelman Center for Cellular and Molecular Therapy, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Neurology, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michelle E Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Departments of Pediatrics and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
64
|
Ohene-Nyako M, Persons AL, Napier TC. Hippocampal blood-brain barrier of methamphetamine self-administering HIV-1 transgenic rats. Eur J Neurosci 2021; 53:416-429. [PMID: 32725911 PMCID: PMC9949894 DOI: 10.1111/ejn.14925] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 07/19/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022]
Abstract
Combined antiretroviral therapy for HIV infection reduces plasma viral load and prolongs life. However, the brain is a viral reservoir, and pathologies such as cognitive decline and blood-brain barrier (BBB) disruption persist. Methamphetamine abuse is prevalent among HIV-infected individuals. Methamphetamine and HIV toxic proteins can disrupt the BBB, but it is unclear if there exists a common pathway by which HIV proteins and methamphetamine induce BBB damage. Also unknown are the BBB effects imposed by chronic exposure to HIV proteins in the comorbid context of chronic methamphetamine abuse. To evaluate these scenarios, we trained HIV-1 transgenic (Tg) and non-Tg rats to self-administer methamphetamine using a 21-day paradigm that produced an equivalency dose range at the low end of the amounts self-titrated by humans. Markers of BBB integrity were measured for the hippocampus, a brain region involved in cognitive function. Outcomes revealed that tight junction proteins, claudin-5 and occludin, were reduced in Tg rats independent of methamphetamine, and this co-occurred with increased levels of lipopolysaccharide, albumin (indicating barrier breakdown) and matrix metalloproteinase-9 (MMP-9; indicating barrier matrix disruption); reductions in GFAP (indicating astrocytic dysfunction); and microglial activation (indicating inflammation). Evaluations of markers for two signaling pathways that regulate MMP-9 transcription, NF-κB and ERK/∆FosB revealed an overall genotype effect for NF-κB. Methamphetamine did not alter measurements from Tg rats, but in non-Tg rats, methamphetamine reduced occludin and GFAP, and increased MMP-9 and NF-κB. Study outcomes suggest that BBB dysregulation resulting from chronic exposure to HIV-1 proteins or methamphetamine both involve NF-κB/MMP-9.
Collapse
Affiliation(s)
- Michael Ohene-Nyako
- Department of Pharmacology, Rush University, Chicago, IL, USA,Department of Physician Assistant Studies, Rush University, Chicago, IL, USA
| | - Amanda L. Persons
- Department of Physician Assistant Studies, Rush University, Chicago, IL, USA,Department of Psychiatry and Behavioral Sciences, Rush University, Chicago, IL, USA,Center for Compulsive Behavior and Addiction, Rush University, Chicago, IL, USA
| | - T. Celeste Napier
- Department of Pharmacology, Rush University, Chicago, IL, USA,Department of Physician Assistant Studies, Rush University, Chicago, IL, USA,Center for Compulsive Behavior and Addiction, Rush University, Chicago, IL, USA
| |
Collapse
|
65
|
Qiao X, Zhu Y, Dang W, Wang R, Sun M, Chen Y, Shi Y, Zhang L. Dual-specificity phosphatase 15 (DUSP15) in the nucleus accumbens is a novel negative regulator of morphine-associated contextual memory. Addict Biol 2021; 26:e12884. [PMID: 32043707 DOI: 10.1111/adb.12884] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 01/18/2020] [Accepted: 01/23/2020] [Indexed: 12/21/2022]
Abstract
Drug relapse among addicts often occurs due to the learned association between drug-paired cues and the rewarding effects of these drugs, such as morphine. Contextual memory associated with morphine has a central role in maintenance and relapse. We showed that morphine-conditioned place preference (CPP) activates extracellular-regulated protein kinase (ERK) in the nucleus accumbens (NAc). The main enzymes that mediate ERK dephosphorylation are members of the dual-specificity phosphatase (DUSP) superfamily. It is unclear which members regulate the morphine CPP-induced activation of ERK. After screening, DUSP15 was found to be decreased during both morphine CPP expression and the reinstatement period. Intra-NAc infusions of AAV-DUSP15 (overexpression) not only prevented the expression of morphine-induced CPP but also facilitated extinction, inhibited reinstatement, and abolished ERK activation. However, after repeated morphine exposure and withdrawal in mice, there was no change in the expression of p-ERK and DUSP15, and the overexpression of DUSP15 in the NAc did not improve the impaired spatial memory or anxiety-like behaviour induced by morphine. Together, these findings indicate that DUSP15 not only prevents the expression of drug-paired contextual memory but also promotes the extinction of existing addiction memories, thus providing a novel therapeutic target for the treatment of drug addiction.
Collapse
Affiliation(s)
- Xiaomeng Qiao
- Department of Forensic Medicine, School of Basic Medical Sciences Zhengzhou University China
| | - Yongsheng Zhu
- College of Forensic Science, School of Medicine, Xi'an Jiaotong University China
| | - Wei Dang
- The Sixth Ward, Xi'an Mental Health Center China
| | - Runzhi Wang
- Department of Forensic Medicine, School of Basic Medical Sciences Zhengzhou University China
| | - Mizhu Sun
- Department of Forensic Medicine, School of Basic Medical Sciences Zhengzhou University China
| | - Yuanyuan Chen
- College of Forensic Science, School of Medicine, Xi'an Jiaotong University China
| | - Yuhui Shi
- College of Forensic Science, School of Medicine, Xi'an Jiaotong University China
| | - Lirong Zhang
- Department of Pharmacology, School of Basic Medical Sciences Zhengzhou University China
| |
Collapse
|
66
|
Abstract
DARPP-32 (dopamine- and cAMP-regulated phosphoprotein with an apparent Mr of 32,000), now also known as phosphoprotein phosphatase 1 regulatory subunit 1B (PPP1R1B), is a potent inhibitor of protein phosphatase 1 (PP1, also known as PPP1) when phosphorylated at Thr34 by cAMP-dependent protein kinase (PKA). DARPP-32 exhibits a remarkable regional distribution in brain, roughly similar to that of dopamine innervation. Its discovery was a culmination of the long-standing effort of Paul Greengard to understand the mechanisms through which neurotransmitters such as dopamine exert their effects on target neurons. DARPP-32 is particularly enriched in striatal projection neurons where it is regulated by numerous signals through which it integrates and amplifies responses to many stimuli. Molecular studies of DARPP-32 have revealed that its regulation and function are more complex than anticipated. It is phosphorylated on multiple sites by several protein kinases that modulate DARPP-32 properties. Primarily, when phosphorylated at Thr34 DARPP-32 is a potent inhibitor of PP1, whereas when phosphorylated at Thr75 by Cdk5 it inhibits PKA. Phosphorylation at serine residues by CK1 and CK2 modulates its intracellular localization and its sensitivity to kinases or phosphatases. Modeling studies provide evidence that the signaling pathways including DARPP-32 are endowed of strong robustness and bistable properties favoring switch-like responses. Thus DARPP-32 combined with a set of other distinct signaling molecules enriched in striatal projection neurons plays a key role in the characteristic properties and physiological function of these neurons.
Collapse
|
67
|
Cocaine-Dependent Acquisition of Locomotor Sensitization and Conditioned Place Preference Requires D1 Dopaminergic Signaling through a Cyclic AMP, NCS-Rapgef2, ERK, and Egr-1/Zif268 Pathway. J Neurosci 2020; 41:711-725. [PMID: 33268547 DOI: 10.1523/jneurosci.1497-20.2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 11/08/2020] [Accepted: 11/15/2020] [Indexed: 11/21/2022] Open
Abstract
Elucidation of the mechanism of dopamine signaling to ERK that underlies plasticity in dopamine D1 receptor-expressing neurons leading to acquired cocaine preference is incomplete. NCS-Rapgef2 is a novel cAMP effector, expressed in neuronal and endocrine cells in adult mammals, that is required for D1 dopamine receptor-dependent ERK phosphorylation in mouse brain. In this report, we studied the effects of abrogating NCS-Rapgef2 expression on cAMP-dependent ERK→Egr-1/Zif268 signaling in cultured neuroendocrine cells; in D1 medium spiny neurons of NAc slices; and in either male or female mouse brain in a region-specific manner. NCS-Rapgef2 gene deletion in the NAc in adult mice, using adeno-associated virus-mediated expression of cre recombinase, eliminated cocaine-induced ERK phosphorylation and Egr-1/Zif268 upregulation in D1-medium spiny neurons and cocaine-induced behaviors, including locomotor sensitization and conditioned place preference. Abrogation of NCS-Rapgef2 gene expression in mPFC and BLA, by crossing mice bearing a floxed Rapgef2 allele with a cre mouse line driven by calcium/calmodulin-dependent kinase IIα promoter also eliminated cocaine-induced phospho-ERK activation and Egr-1/Zif268 induction, but without effect on the cocaine-induced behaviors. Our results indicate that NCS-Rapgef2 signaling to ERK in dopamine D1 receptor-expressing neurons in the NAc, but not in corticolimbic areas, contributes to cocaine-induced locomotor sensitization and conditioned place preference. Ablation of cocaine-dependent ERK activation by elimination of NCS-Rapgef2 occurred with no effect on phosphorylation of CREB in D1 dopaminoceptive neurons of NAc. This study reveals a new cAMP-dependent signaling pathway for cocaine-induced behavioral adaptations, mediated through NCS-Rapgef2/phospho-ERK activation, independently of PKA/CREB signaling.SIGNIFICANCE STATEMENT ERK phosphorylation in dopamine D1 receptor-expressing neurons exerts a pivotal role in psychostimulant-induced neuronal gene regulation and behavioral adaptation, including locomotor sensitization and drug preference in rodents. In this study, we examined the role of dopamine signaling through the D1 receptor via a novel pathway initiated through the cAMP-activated guanine nucleotide exchange factor NCS-Rapgef2 in mice. NCS-Rapgef2 in the NAc is required for activation of ERK and Egr-1/Zif268 in D1 dopaminoceptive neurons after acute cocaine administration, and subsequent enhanced locomotor response and drug seeking behavior after repeated cocaine administration. This novel component in dopamine signaling provides a potential new target for intervention in psychostimulant-shaped behaviors, and new understanding of how D1-medium spiny neurons encode the experience of psychomotor stimulant exposure.
Collapse
|
68
|
Porru S, Maccioni R, Bassareo V, Peana AT, Salamone JD, Correa M, Acquas E. Effects of caffeine on ethanol-elicited place preference, place aversion and ERK phosphorylation in CD-1 mice. J Psychopharmacol 2020; 34:1357-1370. [PMID: 33103552 DOI: 10.1177/0269881120965892] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Epidemiological studies indicate a rise in the combined consumption of caffeinated and alcoholic beverages, which can lead to increased risk of alcoholic-beverage overconsumption. However, the effects of the combination of caffeine and ethanol in animal models related to aspects of drug addiction are still underexplored. AIMS To characterize the pharmacological interaction between caffeine and ethanol and establish if caffeine can affect the ability of ethanol (2 g/kg) to elicit conditioned place preference and conditioned place aversion, we administered caffeine (3 or 15 mg/kg) to male CD-1 mice before saline or ethanol. Moreover, we determined if these doses of caffeine could affect ethanol (2 g/kg) elicited extracellular signal-regulated kinase phosphorylation in brain areas, nucleus accumbens, bed nucleus of stria terminalis, central nucleus of the amygdala, and basolateral amygdala, previously associated with this type of associative learning. RESULTS In the place-conditioning paradigm, caffeine did not have an effect on its own, whereas ethanol elicited significant conditioned-place preference and conditioned-place aversion. Caffeine (15 mg/kg) significantly prevented the acquisition of ethanol-elicited conditioned-place preference and, at both doses, also prevented the acquisition of ethanol-elicited conditioned-place aversion. Moreover, both doses of caffeine also prevented ethanol-elicited extracellular signal-regulated kinase phosphorylation expression in all brain areas examined. CONCLUSIONS The present data indicate a functional antagonistic action of caffeine and ethanol on associative learning and extracellular signal-regulated kinase phosphorylation after an acute interaction. These results could provide exciting grounds for further studies, also in a translational perspective, of their pharmacological interaction modulating other processes involved in drug consumption and addiction.
Collapse
Affiliation(s)
- Simona Porru
- Department of Life and Environmental Sciences, University of Cagliari, Monserrato, Italy.,Department of Psychobiology, University Jaume I, Castelló, Spain
| | - Riccardo Maccioni
- Department of Life and Environmental Sciences, University of Cagliari, Monserrato, Italy
| | - Valentina Bassareo
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Alessandra T Peana
- Department of Chemistry and Pharmacy, University of Sassari, Sassari, Italy
| | - John D Salamone
- Department of Psychological Sciences, University of Connecticut, Storrs, USA
| | - Mercè Correa
- Department of Psychobiology, University Jaume I, Castelló, Spain
| | - Elio Acquas
- Department of Life and Environmental Sciences, University of Cagliari, Monserrato, Italy.,Centre of Excellence on Neurobiology of Addiction, University of Cagliari, Cagliari, Italy
| |
Collapse
|
69
|
Longueville S, Nakamura Y, Brami-Cherrier K, Coura R, Hervé D, Girault JA. Long-lasting tagging of neurons activated by seizures or cocaine administration in Egr1-CreER T2 transgenic mice. Eur J Neurosci 2020; 53:1450-1472. [PMID: 33226686 DOI: 10.1111/ejn.15060] [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: 10/13/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 11/29/2022]
Abstract
Permanent tagging of neuronal ensembles activated in specific experimental situations is an important objective to study their properties and adaptations. In the context of learning and memory, these neurons are referred to as engram neurons. Here, we describe and characterize a novel mouse line, Egr1-CreERT2 , which carries a transgene in which the promoter of the immediate early gene Egr1 drives the expression of the CreERT2 recombinase that is only active in the presence of tamoxifen metabolite, 4-hydroxy-tamoxifen (4-OHT). Egr1-CreERT2 mice were crossed with various reporter mice, Cre-dependently expressing a fluorescent protein. Without tamoxifen or 4-OHT, no or few tagged neurons were observed. Epileptic seizures induced by pilocarpine or pentylenetetrazol in the presence of tamoxifen or 4-OHT elicited the persistent tagging of many neurons and some astrocytes in the dentate gyrus of hippocampus, where Egr1 is transiently induced by seizures. One week after cocaine and 4-OHT administration, these mice displayed a higher number of tagged neurons in the dorsal striatum than saline/4-OHT controls, with differences between reporter lines. Cocaine-induced tagging required ERK activation and tagged neurons were more likely than others to exhibit ERK phosphorylation or Fos induction after a second injection. Interestingly neurons tagged in saline-treated mice also had an increased propensity to express Fos, suggesting the existence of highly responsive striatal neurons susceptible to be re-activated by cocaine repeated administration, which may contribute to the behavioral adaptations. Our report validates a novel transgenic mouse model for permanently tagging activated neurons and studying long-term alterations of Egr1-expressing cells.
Collapse
Affiliation(s)
- Sophie Longueville
- Inserm UMR-S 1270, Paris, France.,Sciences and Engineering Faculty, Sorbonne Université, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Yuki Nakamura
- Inserm UMR-S 1270, Paris, France.,Sciences and Engineering Faculty, Sorbonne Université, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Karen Brami-Cherrier
- Inserm UMR-S 1270, Paris, France.,Sciences and Engineering Faculty, Sorbonne Université, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Renata Coura
- Inserm UMR-S 1270, Paris, France.,Sciences and Engineering Faculty, Sorbonne Université, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Denis Hervé
- Inserm UMR-S 1270, Paris, France.,Sciences and Engineering Faculty, Sorbonne Université, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Jean-Antoine Girault
- Inserm UMR-S 1270, Paris, France.,Sciences and Engineering Faculty, Sorbonne Université, Paris, France.,Institut du Fer à Moulin, Paris, France
| |
Collapse
|
70
|
Mongi-Bragato B, Avalos MP, Guzmán AS, García-Keller C, Bollati FA, Cancela LM. Endogenous enkephalin is necessary for cocaine-induced alteration in glutamate transmission within the nucleus accumbens. Eur J Neurosci 2020; 53:1441-1449. [PMID: 33159343 DOI: 10.1111/ejn.15035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 11/30/2022]
Abstract
Altered glutamate transmission within the nucleus accumbens (NAc) has been proposed as a central mechanism underlying behavioural sensitisation associated with repeated cocaine exposure. In addition to glutamate, enkephalin, an endogenous opioid peptide derived from proenkephalin, is necessary for the neuroadaptations associated with chronic cocaine. However, the influence of enkephalin on long-term changes in glutamate transmission within the NAc associated with cocaine-induced sensitisation has not been described. This study used knockout proenkephalin mice (KO) to study the influence of endogenous enkephalin on the adaptations in glutamate neurotransmission associated with repeated cocaine treatment. Wild-type (WT) and KO mice were treated with daily cocaine injections for 9 days to induce sensitisation. On days 15 and 21, the animals received a cocaine challenge and locomotor sensitisation was evaluated, and microdialysis was performed to determine accumbens glutamate content on day 21. No expression of behavioural sensitisation to cocaine was evidenced in the KO mice. Consistently, these showed no changes in glutamate transmission in the NAc associated with repeated cocaine. This study reveals the central role of enkephalin in regulating the glutamate mechanisms associated with cocaine sensitisation.
Collapse
Affiliation(s)
- Bethania Mongi-Bragato
- Departamento de Farmacología, Facultad de Ciencias Químicas, Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María Paula Avalos
- Departamento de Farmacología, Facultad de Ciencias Químicas, Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Andrea S Guzmán
- Departamento de Farmacología, Facultad de Ciencias Químicas, Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Constanza García-Keller
- Departamento de Farmacología, Facultad de Ciencias Químicas, Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina.,Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA
| | - Flavia A Bollati
- Departamento de Farmacología, Facultad de Ciencias Químicas, Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Liliana M Cancela
- Departamento de Farmacología, Facultad de Ciencias Químicas, Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
71
|
Christensen KR, Nairn AC. cAMP-regulated phosphoproteins DARPP-32, ARPP16/19, and RCS modulate striatal signal transduction through protein kinases and phosphatases. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2020; 90:39-65. [PMID: 33706938 DOI: 10.1016/bs.apha.2020.09.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Decades of research led by Paul Greengard identified protein phosphorylation as a ubiquitous and vital post-translational modification involved in many neuronal signaling pathways. In particular, his discovery that second messenger-regulated protein phosphorylation plays a central role in the propagation and transduction of signals in the nervous system has been essential in understanding the molecular mechanisms of neuronal communication. The establishment of dopamine (DA) as an essential neurotransmitter in the central nervous system, combined with observations that DA activates G-protein-coupled receptors to control the production of cyclic adenosine monophosphate (cAMP) in postsynaptic neurons, has provided fundamental insight into the regulation of neurotransmission. Notably, DA signaling in the striatum is involved in many neurological functions such as control of locomotion, reward, addiction, and learning, among others. This review focuses on the history, characterization, and function of cAMP-mediated regulation of serine/threonine protein phosphatases and their role in DA-mediated signaling in striatal neurons. Several small, heat- and acid-stable proteins, including DARPP-32, RCS, and ARPP-16/19, were discovered by the Greengard laboratory to be regulated by DA- and cAMP signaling, and found to undergo a complex but coordinated sequence of phosphorylation and dephosphorylation events. These studies have contributed significantly to the establishment of protein phosphorylation as a ubiquitous and vital process in signal propagation in neurons, paradigm shifting discoveries at the time. Understanding DA-mediated signaling in the context of signal propagation has led to numerous insights into human conditions and the development of treatments and therapies.
Collapse
Affiliation(s)
- Kyle R Christensen
- Department of Psychiatry, Yale University, School of Medicine, New Haven, CT, United States
| | - Angus C Nairn
- Department of Psychiatry, Yale University, School of Medicine, New Haven, CT, United States.
| |
Collapse
|
72
|
Kim B, Yun J, Park B. Methamphetamine-Induced Neuronal Damage: Neurotoxicity and Neuroinflammation. Biomol Ther (Seoul) 2020; 28:381-388. [PMID: 32668144 PMCID: PMC7457172 DOI: 10.4062/biomolther.2020.044] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/23/2020] [Accepted: 06/25/2020] [Indexed: 01/11/2023] Open
Abstract
Methamphetamine (METH) is a highly addictive psychostimulant and one of the most widely abused drugs worldwide. The continuous use of METH eventually leads to drug addiction and causes serious health complications, including attention deficit, memory loss and cognitive decline. These neurological complications are strongly associated with METH-induced neurotoxicity and neuroinflammation, which leads to neuronal cell death. The current review investigates the molecular mechanisms underlying METH-mediated neuronal damages. Our analysis demonstrates that the process of neuronal impairment by METH is closely related to oxidative stress, transcription factor activation, DNA damage, excitatory toxicity and various apoptosis pathways. Thus, we reach the conclusion here that METH-induced neuronal damages are attributed to the neurotoxic and neuroinflammatory effect of the drug. This review provides an insight into the mechanisms of METH addiction and contributes to the discovery of therapeutic targets on neurological impairment by METH abuse.
Collapse
Affiliation(s)
- Buyun Kim
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Jangmi Yun
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Byoungduck Park
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| |
Collapse
|
73
|
Calabrese V, Di Maio A, Marino G, Cardinale A, Natale G, De Rosa A, Campanelli F, Mancini M, Napolitano F, Avallone L, Calabresi P, Usiello A, Ghiglieri V, Picconi B. Rapamycin, by Inhibiting mTORC1 Signaling, Prevents the Loss of Striatal Bidirectional Synaptic Plasticity in a Rat Model of L-DOPA-Induced Dyskinesia. Front Aging Neurosci 2020; 12:230. [PMID: 32848709 PMCID: PMC7431470 DOI: 10.3389/fnagi.2020.00230] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 06/30/2020] [Indexed: 12/16/2022] Open
Abstract
Levodopa (L-DOPA) treatment is the main gold-standard therapy for Parkinson disease (PD). Besides good antiparkinsonian effects, prolonged use of this drug is associated to the development of involuntary movements known as L-DOPA-induced dyskinesia (LID). L-DOPA-induced dyskinesia is linked to a sensitization of dopamine (DA) D1 receptors located on spiny projection neurons (SPNs) of the dorsal striatum. Several evidences have shown that the emergence of LID can be related to striatal D1/cAMP/PKA/DARPP-32 and extracellular signal-regulated kinases (ERK1/2) pathway overactivation associated to aberrant N-methyl-d-aspartate (NMDA) receptor function. In addition, within striatum, ERK1/2 is also able to modulate in a D1 receptor-dependent manner the activity of the mammalian target of rapamycin complex 1 (mTORC1) pathway under DA depletion and L-DOPA therapy. Consistently, increased mTORC1 signaling appears during chronic administration of L-DOPA and shows a high correlation with the severity of dyskinesia. Furthermore, the abnormal activation of the D1/PKA/DARPP-32 cascade is paralleled by increased phosphorylation of the GluA1 subunit of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor at the PKA Ser845 site. The GluA1 promotes excitatory AMPA receptor-mediated transmission and may be implicated in the alterations found at the corticostriatal synapses of dyskinetic animals. In our study, we investigated the role of mTORC1 pathway activation in modulating bidirectional striatal synaptic plasticity in L-DOPA-treated parkinsonian rats. Inhibition of mTORC1 by coadministration of rapamycin to L-DOPA was able to limit the magnitude of LID expression, accounting for a therapeutic effect of this drug. In particular, behavioral data showed that, in L-DOPA-treated rats, rapamycin administration induced a selective decrease of distinct components of abnormal involuntary movements (i.e., axial and orolingual dyskinesia). Furthermore, ex vivo patch clamp and intracellular recordings of SPNs revealed that pharmacological inhibition of mTORC1 also resulted associated with a physiological bidirectional plasticity, when compared to dyskinetic rats treated with L-DOPA alone. This study uncovers the important role of mTORC1 inhibition to prevent the loss of striatal bidirectional plasticity under chronic L-DOPA treatment in rodent models of PD.
Collapse
Affiliation(s)
- Valeria Calabrese
- Laboratory of Experimental Neurophysiology, IRCCS San Raffaele Pisana, Rome, Italy.,Department of Medicine, University of Perugia, Perugia, Italy
| | | | - Gioia Marino
- Department of Medicine, University of Perugia, Perugia, Italy.,Laboratory of Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Antonella Cardinale
- Laboratory of Experimental Neurophysiology, IRCCS San Raffaele Pisana, Rome, Italy.,Department of Medicine, University of Perugia, Perugia, Italy
| | - Giuseppina Natale
- Department of Medicine, University of Perugia, Perugia, Italy.,Laboratory of Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Arianna De Rosa
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Federica Campanelli
- Department of Medicine, University of Perugia, Perugia, Italy.,Laboratory of Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Maria Mancini
- Laboratory of Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Francesco Napolitano
- CEINGE Biotecnologie Avanzate, Naples, Italy.,Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | - Luigi Avallone
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | - Paolo Calabresi
- Neurologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandro Usiello
- CEINGE Biotecnologie Avanzate, Naples, Italy.,Department of Environmental, Biological and Pharmaceutical Sciences and Technologies (DISTABIF), University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Veronica Ghiglieri
- Department of Medicine, University of Perugia, Perugia, Italy.,Laboratory of Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Barbara Picconi
- Laboratory of Experimental Neurophysiology, IRCCS San Raffaele Pisana, Rome, Italy.,Università Telematica San Raffaele, Rome, Italy
| |
Collapse
|
74
|
Prud'hon S, Bekadar S, Rastetter A, Guégan J, Cormier-Dequaire F, Lacomblez L, Mangone G, You H, Daniau M, Marie Y, Bertrand H, Lesage S, Tezenas Du Montcel S, Anheim M, Brice A, Danjou F, Corvol JC. Exome Sequencing Reveals Signal Transduction Genes Involved in Impulse Control Disorders in Parkinson's Disease. Front Neurol 2020; 11:641. [PMID: 32793093 PMCID: PMC7385236 DOI: 10.3389/fneur.2020.00641] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 05/29/2020] [Indexed: 01/08/2023] Open
Abstract
Introduction: Impulse control disorders (ICDs) frequently complicate dopamine agonist (DA) therapy in Parkinson's disease (PD). There is growing evidence of a high heritability for ICDs in the general population and in PD. Variants on genes belonging to the reward pathway have been shown to account for part of this heritability. We aimed to identify new pathways associated with ICDs in PD. Methods: Thirty-six Parkinsonian patients on DA therapy with (n = 18) and without ICDs (n = 18) matched on age at PD's onset, and gender was selected to represent the most extreme phenotypes of their category. Exome sequencing was performed, and variants with a strong functional impact in brain-expressed genes were selected. Allele frequencies and their distribution in genes and pathways were analyzed with single variant and SKAT-O tests. The 10 most associated variants, genes, and pathways were retained for replication in the Parkinson's progression markers initiative (PPMI) cohort. Results: None of markers tested passed the significance threshold adjusted for multiple comparisons. However, the “Adenylate cyclase activating” pathway, one of the top associated pathways in the discovery data set (p = 1.6 × 10−3) was replicated in the PPMI cohort and was significantly associated with ICDs in a post hoc pooled analysis (combined p-value 3.3 × 10−5). Two of the 10 most associated variants belonged to genes implicated in cAMP and ERK signaling (rs34193571 in RasGRF2, p = 5 × 10−4; rs1877652 in PDE2A, p = 8 × 10−4) although non-significant after Bonferroni correction. Conclusion: Our results suggest that genes implicated in the signaling pathways linked to G protein-coupled receptors participate to genetic susceptibility to ICDs in PD.
Collapse
Affiliation(s)
- Sabine Prud'hon
- Sorbonne Université, INSERM UMRS 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle, ICM, Paris, France.,Assistance Publique Hôpitaux de Paris, Centre d'Investigation Clinique neurosciences, Department of Neurology, Hôpital Pitié-Salpêtrière, Paris, France
| | - Samir Bekadar
- Sorbonne Université, INSERM UMRS 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle, ICM, Paris, France.,Assistance Publique Hôpitaux de Paris, Centre d'Investigation Clinique neurosciences, Department of Neurology, Hôpital Pitié-Salpêtrière, Paris, France
| | - Agnès Rastetter
- Sorbonne Université, INSERM UMRS 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle, ICM, Paris, France
| | - Justine Guégan
- Sorbonne Université, INSERM UMRS 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle, ICM, Paris, France
| | - Florence Cormier-Dequaire
- Sorbonne Université, INSERM UMRS 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle, ICM, Paris, France.,Assistance Publique Hôpitaux de Paris, Centre d'Investigation Clinique neurosciences, Department of Neurology, Hôpital Pitié-Salpêtrière, Paris, France
| | - Lucette Lacomblez
- Assistance Publique Hôpitaux de Paris, Centre d'Investigation Clinique neurosciences, Department of Neurology, Hôpital Pitié-Salpêtrière, Paris, France.,Assistance Publique Hôpitaux de Paris, Department of Pharmacology, Hôpital Pitié-Salpêtrière, Paris, France
| | - Graziella Mangone
- Sorbonne Université, INSERM UMRS 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle, ICM, Paris, France.,Assistance Publique Hôpitaux de Paris, Centre d'Investigation Clinique neurosciences, Department of Neurology, Hôpital Pitié-Salpêtrière, Paris, France
| | - Hana You
- Sorbonne Université, INSERM UMRS 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle, ICM, Paris, France.,Assistance Publique Hôpitaux de Paris, Centre d'Investigation Clinique neurosciences, Department of Neurology, Hôpital Pitié-Salpêtrière, Paris, France
| | - Mailys Daniau
- Sorbonne Université, INSERM UMRS 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle, ICM, Paris, France
| | - Yannick Marie
- Sorbonne Université, INSERM UMRS 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle, ICM, Paris, France
| | - Hélène Bertrand
- Sorbonne Université, INSERM UMRS 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle, ICM, Paris, France
| | - Suzanne Lesage
- Sorbonne Université, INSERM UMRS 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle, ICM, Paris, France
| | - Sophie Tezenas Du Montcel
- Assistance Publique Hôpitaux de Paris, Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Hôpital Pitié-Salpêtrière, Paris, France
| | - Mathieu Anheim
- Hôpitaux Universitaires de Strasbourg, Department of Neurology, Strasbourg, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), UMR 7104 CNRS/Unistra, Inserm U1258, Illkirch, France.,Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Alexis Brice
- Sorbonne Université, INSERM UMRS 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle, ICM, Paris, France.,Assistance Publique Hôpitaux de Paris, Department of Genetics, Hôpital Pitié-Salpêtrière, Paris, France
| | - Fabrice Danjou
- Sorbonne Université, INSERM UMRS 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle, ICM, Paris, France
| | - Jean-Christophe Corvol
- Sorbonne Université, INSERM UMRS 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle, ICM, Paris, France.,Assistance Publique Hôpitaux de Paris, Centre d'Investigation Clinique neurosciences, Department of Neurology, Hôpital Pitié-Salpêtrière, Paris, France
| |
Collapse
|
75
|
Delint-Ramirez I, Segev A, Pavuluri A, Self DW, Kourrich S. Cocaine-Induced Synaptic Redistribution of NMDARs in Striatal Neurons Alters NMDAR-Dependent Signal Transduction. Front Neurosci 2020; 14:698. [PMID: 32760242 PMCID: PMC7371985 DOI: 10.3389/fnins.2020.00698] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/09/2020] [Indexed: 11/25/2022] Open
Abstract
The consequence of repeated cocaine exposure and prolonged abstinence on glutamate receptor expression in the nucleus accumbens has been extensively studied. However, the early effects of cocaine on NMDAR signaling remain unknown. NMDAR signaling depends on the subunit composition, subcellular localization, and the interaction with proteins at the postsynaptic density (PSD), where NMDARs and other proteins form supercomplexes that are responsible for the signaling pathways activated by NMDAR-induced Ca2+ influx. Here, we investigated the effect of cocaine on NMDAR subunit composition and subcellular localization after both intraperitoneal non-contingent cocaine and response-contingent intravenous cocaine self-administration in mice. We found that repeated cocaine exposure, regardless of the route or contingency of drug administration, decreases NMDAR interactions with the PSD and synaptic lipid rafts in the accumbens shell and dorsal striatum. We provide evidence that cocaine triggers an early redistribution of NMDARs from synaptic to extrasynaptic sites, and that this adaptation has implications in the activation of downstream signaling pathways. Thus, consistent with a loss of NMDAR function, cocaine-induced ERK phosphorylation is attenuated. Because early NMDAR activity contributes to the initiation of lasting addiction-relevant neuroadaptations, these data may hold clues into cellular mechanisms responsible for the development of cocaine addiction.
Collapse
Affiliation(s)
- Ilse Delint-Ramirez
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Amir Segev
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Asha Pavuluri
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - David W Self
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Saïd Kourrich
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Département des Sciences Biologiques-CERMO-FC, Université du Québec à Montréal, Montreal, QC, Canada
| |
Collapse
|
76
|
Salery M, Trifilieff P, Caboche J, Vanhoutte P. From Signaling Molecules to Circuits and Behaviors: Cell-Type-Specific Adaptations to Psychostimulant Exposure in the Striatum. Biol Psychiatry 2020; 87:944-953. [PMID: 31928716 DOI: 10.1016/j.biopsych.2019.11.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/30/2019] [Accepted: 11/01/2019] [Indexed: 12/17/2022]
Abstract
Addiction is characterized by a compulsive pattern of drug seeking and consumption and a high risk of relapse after withdrawal that are thought to result from persistent adaptations within brain reward circuits. Drugs of abuse increase dopamine (DA) concentration in these brain areas, including the striatum, which shapes an abnormal memory trace of drug consumption that virtually highjacks reward processing. Long-term neuronal adaptations of gamma-aminobutyric acidergic striatal projection neurons (SPNs) evoked by drugs of abuse are critical for the development of addiction. These neurons form two mostly segregated populations, depending on the DA receptor they express and their output projections, constituting the so-called direct (D1 receptor) and indirect (D2 receptor) SPN pathways. Both SPN subtypes receive converging glutamate inputs from limbic and cortical regions, encoding contextual and emotional information, together with DA, which mediates reward prediction and incentive values. DA differentially modulates the efficacy of glutamate synapses onto direct and indirect SPN pathways by recruiting distinct striatal signaling pathways, epigenetic and genetic responses likely involved in the transition from casual drug use to addiction. Herein we focus on recent studies that have assessed psychostimulant-induced alterations in a cell-type-specific manner, from remodeling of input projections to the characterization of specific molecular events in each SPN subtype and their impact on long-lasting behavioral adaptations. We discuss recent evidence revealing the complex and concerted action of both SPN populations on drug-induced behavioral responses, as these studies can contribute to the design of future strategies to alleviate specific behavioral components of addiction.
Collapse
Affiliation(s)
- Marine Salery
- Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Pierre Trifilieff
- NutriNeuro, Unité Mixte de Recherche (UMR) 1286, Institut National de la Recherche Agronomique, Bordeaux Institut Polytechnique, University of Bordeaux, Bordeaux, France
| | - Jocelyne Caboche
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Sorbonne Université, Faculty of Sciences, Paris, France; Centre National de la Recherche Scientifique, UMR8246, Paris, France; Institut National de la Santé et de la Recherche Médicale, U1130, Paris France.
| | - Peter Vanhoutte
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Sorbonne Université, Faculty of Sciences, Paris, France; Centre National de la Recherche Scientifique, UMR8246, Paris, France; Institut National de la Santé et de la Recherche Médicale, U1130, Paris France
| |
Collapse
|
77
|
Won S, Roche KW. Regulation of glutamate receptors by striatal-enriched tyrosine phosphatase 61 (STEP 61 ). J Physiol 2020; 599:443-451. [PMID: 32170729 DOI: 10.1113/jp278703] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/25/2020] [Indexed: 12/26/2022] Open
Abstract
Phosphorylation regulates glutamate receptor trafficking. The cytosolic C-terminal domains of both NMDA receptors (NMDARs) and AMPA receptors (AMPARs) have distinct motifs, which are substrates for serine/threonine and tyrosine phosphorylation. Decades of research have shown how phosphorylation of glutamate receptors mediates protein binding and receptor trafficking, ultimately controlling synaptic transmission and plasticity. STEP is a protein tyrosine phosphatase (also known as PTPN5), with several isoforms resulting from alternative splicing. Targets of STEP include a variety of important synaptic substrates, among which are the tyrosine kinase Fyn and glutamate receptors. In particular, STEP61 , the longest isoform, dephosphorylates the NMDAR subunit GluN2B and strongly regulates the expression of NMDARs at synapses. This interplay between STEP, Fyn and GluN2B-containing NMDARs has been characterized by multiple groups. More recently, STEP61 was shown to bind to AMPARs in a subunit-specific manner and differentially regulate synaptic NMDARs and AMPARs. Because of its many effects on synaptic proteins, STEP has been implicated in regulating excitatory synapses during plasticity and playing a role in synaptic dysfunction in a variety of neurological disorders. In this review, we will highlight the ways in which STEP61 differentially regulates NMDARs and AMPARs, as well as its role in plasticity and disease.
Collapse
Affiliation(s)
- Sehoon Won
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Katherine W Roche
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
78
|
Bendová Z, Pačesová D, Novotný J. The day-night differences in ERK1/2, GSK3β activity and c-Fos levels in the brain, and the responsiveness of various brain structures to morphine. J Comp Neurol 2020; 528:2471-2495. [PMID: 32170720 DOI: 10.1002/cne.24906] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/17/2020] [Accepted: 03/09/2020] [Indexed: 11/12/2022]
Abstract
As with other drugs or pharmaceuticals, opioids differ in their rewarding or analgesic effects depending on when they are applied. In the previous study, we have demonstrated the day/night difference in the sensitivity of the major circadian clock in the suprachiasmatic nucleus to a low dose of morphine, and showed the bidirectional effect of morphine on pERK1/2 and pGSK3β levels in the suprachiasmatic nucleus depending on the time of administration. The main aim of this study was to identify other brain structures that respond differently to morphine depending on the time of its administration. Using immunohistochemistry, we identified 44 structures that show time-of-day specific changes in c-Fos level and activity of ERK1/2 and GSK3β kinases in response to a single dose of 1 mg/kg morphine. Furthermore, comparison among control groups revealed the differences in the spontaneous levels of all markers with a generally higher level during the night, that is, in the active phase of the day. We thus provide further evidence for diurnal variations in the activity of brain regions outside the suprachiasmatic nucleus indicated by the temporal changes in the molecular substrate. We suggest that these changes are responsible for generating diurnal variation in the reward behavior or analgesic effect of opioid administration.
Collapse
Affiliation(s)
- Zdeňka Bendová
- Faculty of Science, Charles University, Prague, Czech Republic.,Department of Sleep Medicine and Chronobiology, National Institute of Mental Health, Klecany, Czech Republic
| | - Dominika Pačesová
- Faculty of Science, Charles University, Prague, Czech Republic.,Department of Sleep Medicine and Chronobiology, National Institute of Mental Health, Klecany, Czech Republic
| | - Jiří Novotný
- Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
79
|
Laboute T, Gandía J, Pellissier LP, Corde Y, Rebeillard F, Gallo M, Gauthier C, Léauté A, Diaz J, Poupon A, Kieffer BL, Le Merrer J, Becker JA. The orphan receptor GPR88 blunts the signaling of opioid receptors and multiple striatal GPCRs. eLife 2020; 9:50519. [PMID: 32003745 PMCID: PMC7012601 DOI: 10.7554/elife.50519] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 01/30/2020] [Indexed: 12/16/2022] Open
Abstract
GPR88 is an orphan G protein-coupled receptor (GPCR) considered as a promising therapeutic target for neuropsychiatric disorders; its pharmacology, however, remains scarcely understood. Based on our previous report of increased delta opioid receptor activity in Gpr88 null mice, we investigated the impact of GPR88 co-expression on the signaling of opioid receptors in vitro and revealed that GPR88 inhibits the activation of both their G protein- and β-arrestin-dependent signaling pathways. In Gpr88 knockout mice, morphine-induced locomotor sensitization, withdrawal and supra-spinal analgesia were facilitated, consistent with a tonic inhibitory action of GPR88 on µOR signaling. We then explored GPR88 interactions with more striatal versus non-neuronal GPCRs, and revealed that GPR88 can decrease the G protein-dependent signaling of most receptors in close proximity, but impedes β-arrestin recruitment by all receptors tested. Our study unravels an unsuspected buffering role of GPR88 expression on GPCR signaling, with intriguing consequences for opioid and striatal functions.
Collapse
Affiliation(s)
- Thibaut Laboute
- Deficits of Reward GPCRs and Sociability, Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, Université de Tours, Inserm, Nouzilly, France
| | - Jorge Gandía
- Deficits of Reward GPCRs and Sociability, Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, Université de Tours, Inserm, Nouzilly, France
| | - Lucie P Pellissier
- Deficits of Reward GPCRs and Sociability, Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, Université de Tours, Inserm, Nouzilly, France.,Biology and Bioinformatics of Signalling Systems, Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, Université de Tours, Nouzilly, France
| | - Yannick Corde
- Deficits of Reward GPCRs and Sociability, Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, Université de Tours, Inserm, Nouzilly, France
| | - Florian Rebeillard
- Cellular Biology and Molecular Pharmacology of central Receptors, Centre de Psychiatrie et Neurosciences, Inserm UMR_S894 - Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Maria Gallo
- Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona Biomedical Research Park, Barcelona, Spain
| | - Christophe Gauthier
- Biology and Bioinformatics of Signalling Systems, Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, Université de Tours, Nouzilly, France
| | - Audrey Léauté
- Deficits of Reward GPCRs and Sociability, Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, Université de Tours, Inserm, Nouzilly, France
| | - Jorge Diaz
- Cellular Biology and Molecular Pharmacology of central Receptors, Centre de Psychiatrie et Neurosciences, Inserm UMR_S894 - Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Anne Poupon
- Biology and Bioinformatics of Signalling Systems, Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, Université de Tours, Nouzilly, France
| | - Brigitte L Kieffer
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Canada.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, Inserm U1258, Université de Strasbourg, 1 rue Laurent Fries, Illkirch, France
| | - Julie Le Merrer
- Deficits of Reward GPCRs and Sociability, Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, Université de Tours, Inserm, Nouzilly, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, Inserm U1258, Université de Strasbourg, 1 rue Laurent Fries, Illkirch, France
| | - Jérôme Aj Becker
- Deficits of Reward GPCRs and Sociability, Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, Université de Tours, Inserm, Nouzilly, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, Inserm U1258, Université de Strasbourg, 1 rue Laurent Fries, Illkirch, France
| |
Collapse
|
80
|
Mercatelli D, Bezard E, Eleopra R, Zaveri NT, Morari M. Managing Parkinson's disease: moving ON with NOP. Br J Pharmacol 2020; 177:28-47. [PMID: 31648371 PMCID: PMC6976791 DOI: 10.1111/bph.14893] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/12/2019] [Accepted: 09/25/2019] [Indexed: 01/08/2023] Open
Abstract
The opioid-like neuropeptide nociceptin/orphanin FQ (N/OFQ) and its receptor (NOP receptor) contribute to Parkinson's disease (PD) and motor complications associated with levodopa therapy. The N/OFQ-NOP receptor system is expressed in cortical and subcortical motor areas and, notably, in dopaminergic neurons of the substantia nigra compacta. Dopamine depletion, as in rodent models of PD results in up-regulation of N/OFQ transmission in the substantia nigra and down-regulation of N/OFQ transmission in the striatum. Consistent with this, NOP receptor antagonists relieve motor deficits in PD models by reinstating the physiological balance between excitatory and inhibitory inputs impinging on nigro-thalamic GABAergic neurons. NOP receptor antagonists also counteract the degeneration of nigrostriatal dopaminergic neurons, possibly by attenuating the excitotoxicity or modulating the immune response. Conversely, NOP receptor agonists attenuate levodopa-induced dyskinesia by attenuating the hyperactivation of striatal D1 receptor signalling in neurons of the direct striatonigral pathway. The N/OFQ-NOP receptor system might represent a novel target in the therapy of PD.
Collapse
Affiliation(s)
- Daniela Mercatelli
- Department of Medical Sciences, Section of PharmacologyUniversity of Ferrara and National Institute of NeuroscienceFerraraItaly
| | - Erwan Bezard
- Institut des Maladies Neurodégénératives, UMR 5293Université de BordeauxBordeauxFrance
- Institut des Maladies Neurodégénératives, Centre National de la Recherche Scientifique, UMR 5293BordeauxFrance
| | - Roberto Eleopra
- Neurology Unit 1Fondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
| | - Nurulain T. Zaveri
- Astraea Therapeutics, Medicinal Chemistry DivisionMountain ViewCaliforniaUSA
| | - Michele Morari
- Department of Medical Sciences, Section of PharmacologyUniversity of Ferrara and National Institute of NeuroscienceFerraraItaly
| |
Collapse
|
81
|
Morella I, Hallum H, Brambilla R. Dopamine D1 and Glutamate Receptors Co-operate With Brain-Derived Neurotrophic Factor (BDNF) and TrkB to Modulate ERK Signaling in Adult Striatal Slices. Front Cell Neurosci 2020; 14:564106. [PMID: 33304241 PMCID: PMC7701236 DOI: 10.3389/fncel.2020.564106] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 10/19/2020] [Indexed: 11/13/2022] Open
Abstract
In the striatum, the input nucleus of the basal ganglia, the extracellular-signal-regulated kinase (ERK) pathway, necessary for various forms of behavioral plasticity, is triggered by the combined engagement of dopamine D1 and ionotropic glutamate receptors. In this study, we investigated the potential crosstalk between glutamatergic, dopaminergic, and brain-derived neurotrophic factor (BDNF)-TrkB inputs to ERK cascade by using an ex vivo model of mouse striatal slices. Our results confirmed that the concomitant stimulation of D1 and glutamate receptors is necessary to activate ERK in striatal medium spiny neurons (MSNs). Moreover, we found that ERK activation is significantly enhanced when BDNF is co-applied either with glutamate or the D1 agonist SKF38393, supporting the idea of possible integration between BDNF, glutamate, and D1R-mediated signaling. Interestingly, ERK activation via BDNF-TrkB is upregulated upon blockade of either AMPAR/NMDAR or D1 receptors, suggesting a negative regulatory action of these two neurotransmitter systems on BDNF-mediated signaling. However, the observed enhancement of ERK1/2 phosphorylation does not result in corresponding downstream signaling changes at the nuclear level. Conversely, the TrkB antagonist cyclotraxin B partially prevents glutamate- and D1-mediated ERK activation. Altogether, these results suggest a complex and unexpected interaction among dopaminergic, glutamatergic, and BDNF receptor systems to modulate the ERK pathway in striatal neurons.
Collapse
Affiliation(s)
- Ilaria Morella
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom.,Division of Neuroscience, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Harriet Hallum
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom.,Division of Neuroscience, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Riccardo Brambilla
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom.,Division of Neuroscience, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
82
|
Striatal overexpression of β-arrestin2 counteracts L-dopa-induced dyskinesia in 6-hydroxydopamine lesioned Parkinson's disease rats. Neurochem Int 2019; 131:104543. [DOI: 10.1016/j.neuint.2019.104543] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/16/2019] [Accepted: 09/02/2019] [Indexed: 02/07/2023]
|
83
|
Yasuda RP. Adenosine STEPs on synaptic function: An Editorial for 'The activity of the STriatal-enriched protein tyrosine phosphatase in neuronal cells is modulated by adenosine A2A receptor on' page 284. J Neurochem 2019; 152:270-272. [PMID: 31724181 DOI: 10.1111/jnc.14901] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/16/2019] [Accepted: 10/19/2019] [Indexed: 12/13/2022]
Abstract
This is an Editorial Highlight of a manuscript by Mallozzi et al. (2019) in the current issue of the Journal of Neurochemistry, in which the authors detail the biochemical pathway that leads to synaptic depression by cocaine. This pathway requires the adenosine A2A receptor and STEP phosphatases. Activation of the adenosine A2A receptor leads to an increase in intracellular calcium, activation of STEP by dephosphorylation, inhibition of excitatory ionotropic glutamate receptors by dephosphorylation of phospho-tyrosine residues and subsequent internalization of the ionotropic glutamate receptors. This adenosine A2A receptor pathway could lead to potential drug targets for neurologic and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Robert P Yasuda
- Department of Pharmacology & Physiology, Georgetown University, Washington, DC, USA
| |
Collapse
|
84
|
Gangarossa G, Castell L, Castro L, Tarot P, Veyrunes F, Vincent P, Bertaso F, Valjent E. Contrasting patterns of ERK activation in the tail of the striatum in response to aversive and rewarding signals. J Neurochem 2019; 151:204-226. [PMID: 31245856 DOI: 10.1111/jnc.14804] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/13/2019] [Accepted: 06/19/2019] [Indexed: 01/08/2023]
Abstract
The caudal part of the striatum, also named the tail of the striatum (TS), defines a fourth striatal domain. Determining whether rewarding, aversive and salient stimuli regulate the activity of striatal spiny projections neurons (SPNs) of the TS is therefore of paramount importance to understand its functions, which remain largely elusive. Taking advantage of genetically encoded biosensors (A-kinase activity reporter 3) to record protein kinase A signals and by analyzing the distribution of dopamine D1R- and D2R-SPNs in the TS, we characterized three subterritories: a D2R/A2aR-lacking, a D1R/D2R-intermingled and a D1R/D2R-SPNs-enriched area (corresponding to the amygdalostriatal transition). In addition, we provide evidence that the distribution of D1R- and D2R-SPNs in the TS is evolutionarily conserved (mouse, rat, gerbil). The in vivo analysis of extracellular signal-regulated kinase (ERK) phosphorylation in these TS subterritories in response to distinct appetitive, aversive and pharmacological stimuli revealed that SPNs of the TS are not recruited by stimuli triggering innate aversive responses, fasting, satiety, or palatable signals whereas a reduction in ERK phosphorylation occurred following learned avoidance. In contrast, D1R-SPNs of the intermingled and D2R/A2aR-lacking areas were strongly activated by both D1R agonists and psychostimulant drugs (d-amphetamine, cocaine, 3,4-methyl enedioxy methamphetamine, or methylphenidate), but not by hallucinogens. Finally, a similar pattern of ERK activation was observed by blocking selectively dopamine reuptake. Together, our results reveal that the caudal TS might participate in the processing of specific reward signals and discrete aversive stimuli. Cover Image for this issue: doi: 10.1111/jnc.14526. Open Science: This manuscript was awarded with the Open Materials Badge For more information see: https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Giuseppe Gangarossa
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France.,Université de Paris, BFA, UMR 8251, CNRS, Paris, France
| | - Laia Castell
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Liliana Castro
- Sorbonne Université, CNRS, Biological Adaptation and Ageing, Paris, France
| | - Pauline Tarot
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Frederic Veyrunes
- Institut des Sciences de l'Evolution de Montpellier, ISEM, Université de Montpellier, CNRS, EPHE, IRD, Montpellier, France
| | - Pierre Vincent
- Sorbonne Université, CNRS, Biological Adaptation and Ageing, Paris, France
| | - Federica Bertaso
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Emmanuel Valjent
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| |
Collapse
|
85
|
The expression of DARPP-32 in adult male zebra finches (Taenopygia guttata). Brain Struct Funct 2019; 224:2939-2972. [PMID: 31473781 DOI: 10.1007/s00429-019-01947-0] [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: 04/09/2019] [Accepted: 08/20/2019] [Indexed: 10/26/2022]
Abstract
Although the catecholaminergic circuitry in the zebra finch brain has been well studied, there is little information regarding the postsynaptic targets of dopamine. To answer this question, we looked at overall patterns of immunoreactivity for DARPP-32 (a dopamine and cAMP-regulated phosphoprotein, present mostly in dopaminoceptive neurons) in adult male zebra finches. Our results demonstrated that as in mammals and other avian species, DARPP-32 expression was highest in both medial and lateral striatum. Interestingly, a specific pattern of immunoreactivity was observed in the song control system, with 'core' song control regions, that is, LMANcore (lateral magnocellular nucleus of the anterior nidopallium), RA (nucleus robustus arcopallialis) and HVC being less immunoreactive for DARPP-32 than 'shell' areas such as LMANshell, RAcup, AId (intermediate arcopallium) and HVCshelf. Our results suggest that whereas dopamine may modulate the shell pathways at various levels of the AFP, dopaminergic modulation of the core pathway occurs mainly through Area X, a basal ganglia nucleus. Further, secondary sensory cortices including the perientopallial belt, Fields L1 and L3 had higher DARPP-32-immunoreactivity than primary sensory cortical areas such as the pallial basolateral nucleus, entopallium proper and Field L2, corresponding to somatosensory, visual and auditory systems, respectively. We also found DARPP-32-rich axon terminals surrounding dopaminergic neurons in the ventral tegmental area-substantia nigra complex which in turn project to the striatum, suggesting that there may be a reciprocal modulation between these regions. Overall, DARPP-32 expression appears to be higher in areas involved in integrating sensory information, which further supports the role of this protein as a molecular integrator of different signal processing pathways.
Collapse
|
86
|
Liao JF, Cheng YF, Li SW, Lee WT, Hsu CC, Wu CC, Jeng OJ, Wang S, Tsai YC. Lactobacillus plantarum PS128 ameliorates 2,5-Dimethoxy-4-iodoamphetamine-induced tic-like behaviors via its influences on the microbiota-gut-brain-axis. Brain Res Bull 2019; 153:59-73. [PMID: 31351942 DOI: 10.1016/j.brainresbull.2019.07.027] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/21/2019] [Accepted: 07/23/2019] [Indexed: 12/18/2022]
Abstract
We previously reported a novel psychobiotic strain of Lactobacillus plantarum PS128 (PS128) which could ameliorate anxiety-like& depression-like behaviors and modulate cerebral dopamine (DA) and serotonin (5-HT) in mice. Here, we examine the possibility of using PS128 administration to improve tic-like behaviors by using a 5-HT2A and 5-HT2C receptor agonist 2,5-Dimethoxy-4-iodoamphetamine (DOI). PS128 was orally administered to male Wistar rat for 2 weeks before two daily DOI injections. We recorded the behaviors immediately after the second DOI injection and compared the results with control and haloperidol treatment groups. PS128 significantly reduced tic-like behaviors and pre-pulse inhibition deficit in a threshold-dose of 109 CFU per day. Brain tissue analysis showed that DOI induced abnormal DA efflux in the striatum and prefrontal cortex, while PS128 ingestion improved DA metabolism and increased norepinephrine (NE) levels in these two regions. In addition, PS128 ingestion increased DA transporter and β-arrestin expressions and decreased DOI-induced phosphorylation of DA and cAMP regulated phosphoprotein of molecular weight 32 kDa (DARPP-32) at Thr34 and extracellular regulated protein kinases (ERK). PS128 ingestion also modulated peripheral 5-HT levels and shaped the cecal microbiota composition, which helps to alleviate DOI-induced dysbiosis. These results suggested that PS128 ameliorated DOI-induced tic-like hyper-active behaviors via stabilizing cerebral dopaminergic pathways through its modulation of host's microbiota-gut-brain axis. Thus, we believe there are potentials for utilizing psychobiotics to improve syndromes caused by DA dysregulation in DA-related neurological disorders and movement disorders such as Tourette syndrome.
Collapse
Affiliation(s)
- Jian-Fu Liao
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, 155, Section 2, Linong Street, Beitou Dist., Taipei City 11221, Taiwan, ROC.
| | - Yun-Fang Cheng
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, 155, Section 2, Linong Street, Beitou Dist., Taipei City 11221, Taiwan, ROC; Microbiome Research Center, National Yang-Ming University, 155, Section 2, Linong Street, Beitou Dist., Taipei City 11221, Taiwan, ROC; Bened Biomedical Co., Ltd., 2F-2, No. 129, Sec. 2, Zhongshan N. Rd., Zhongshan Dist., Taipei City 104, Taiwan, ROC.
| | - Shiao-Wen Li
- Molecular Medicine Research Center, Chang Gung University, No. 259, Wenhua 1st Rd., Guishan District, Taoyuan City 33302, Taiwan, ROC.
| | - Wang-Tso Lee
- Department of Pediatrics, National Taiwan University Hospital, No. 8, Zhongshan S. Rd., Zhongzheng Dist., Taipei City 100, Taiwan, ROC.
| | - Chih-Chieh Hsu
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, 155, Section 2, Linong Street, Beitou Dist., Taipei City 11221, Taiwan, ROC; Bened Biomedical Co., Ltd., 2F-2, No. 129, Sec. 2, Zhongshan N. Rd., Zhongshan Dist., Taipei City 104, Taiwan, ROC.
| | - Chien-Chen Wu
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, 155, Section 2, Linong Street, Beitou Dist., Taipei City 11221, Taiwan, ROC; Bened Biomedical Co., Ltd., 2F-2, No. 129, Sec. 2, Zhongshan N. Rd., Zhongshan Dist., Taipei City 104, Taiwan, ROC.
| | - One-Jang Jeng
- Bened Biomedical Co., Ltd., 2F-2, No. 129, Sec. 2, Zhongshan N. Rd., Zhongshan Dist., Taipei City 104, Taiwan, ROC.
| | - Sabrina Wang
- Institute of Anatomy and Cell Biology, National Yang-Ming University, 155, Section 2, Linong Street, Beitou Dist., Taipei City 11221, Taiwan, ROC.
| | - Ying-Chieh Tsai
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, 155, Section 2, Linong Street, Beitou Dist., Taipei City 11221, Taiwan, ROC; Microbiome Research Center, National Yang-Ming University, 155, Section 2, Linong Street, Beitou Dist., Taipei City 11221, Taiwan, ROC.
| |
Collapse
|
87
|
The effects of proteasome on baseline and methamphetamine-dependent dopamine transmission. Neurosci Biobehav Rev 2019; 102:308-317. [DOI: 10.1016/j.neubiorev.2019.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 04/29/2019] [Accepted: 05/09/2019] [Indexed: 12/16/2022]
|
88
|
Modulation and functions of dopamine receptor heteromers in drugs of abuse-induced adaptations. Neuropharmacology 2019; 152:42-50. [DOI: 10.1016/j.neuropharm.2018.12.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/27/2018] [Accepted: 12/03/2018] [Indexed: 12/18/2022]
|
89
|
Medial prefrontal cortex ERK and conditioning: Evidence for the association of increased medial prefrontal cortex ERK with the presence/absence of apomorphine conditioned behavior using a unique post-trial conditioning/extinction protocol. Behav Brain Res 2019; 365:56-65. [DOI: 10.1016/j.bbr.2019.02.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 02/05/2019] [Accepted: 02/12/2019] [Indexed: 11/17/2022]
|
90
|
Valvassori SS, Gava FF, Dal-Pont GC, Simões HL, Damiani-Neves M, Andersen ML, Boeck CR, Quevedo J. Effects of lithium and valproate on ERK/JNK signaling pathway in an animal model of mania induced by amphetamine. Heliyon 2019; 5:e01541. [PMID: 31193305 PMCID: PMC6525279 DOI: 10.1016/j.heliyon.2019.e01541] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 04/11/2019] [Accepted: 04/16/2019] [Indexed: 12/13/2022] Open
Abstract
Bipolar disorder (BD) is a severe and chronic psychiatric disorder, characterized by recurrent mood episodes of depression and mania. Some studies have indicated that there are ERK and JNK pathways alterations in the brain from bipolar patients. The animal model of mania induced by dextroamphetamine (d-AMPH) has been considered an excellent model to study intracellular alterations related to BD. The present study aimed to evaluate the effects of lithium (Li) and valproate (VPA) on the behavioral and ERK1/2/JNK1/2 signaling pathway in an animal model of mania induced by d-AMPH. Wistar rats were first given d-AMPH or saline (Sal) for 14 days, and then, between the 8th and 14th days, the rats were treated with Li, VPA, or Sal. The open-field test was used to evaluate the locomotion and exploration behaviors of rats. The levels of phosphorylated ERK1/2 and JNK1/2 were assessed in the hippocampus and frontal cortex of the rats. Li and VPA reversed the increased of locomotion and exploration induced by d-AMPH. The treatment with VPA or AMPH per se decreased the levels of pERK1 in the hippocampus. The treatment with VPA in the animals submitted to the administration of d-AMPH decreased the levels of ERK1, JNK-1, and JNK-2 phosphorylated in the hippocampus of the animals. The treatment with Li decreased the JNK-1 phosphorylated in the hippocampus of the animals submitted to the animal model of mania induced by d-AMPH. Although the association of VPA plus amphetamine alters some proteins involved in the JNK pathway in the hippocampus, these alterations were very random and seemed that were not related to the d-AMPH-induced manic-like behavior. These results suggest that the manic-like effects induced by d-AMPH and the antimanic effects of mood stabilizers, Li and VPA, are not related to the alteration on ERK1/2 and JNK1/2 pathways.
Collapse
Affiliation(s)
- Samira S Valvassori
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Fernanda F Gava
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Gustavo C Dal-Pont
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Henio Leonardo Simões
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Marcela Damiani-Neves
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Monica Levy Andersen
- Master's Degree in Health and Life Sciences, Postgraduate Program in Nanosciences, Franciscan University, Santa Maria, RS, Brazil
| | | | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.,Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.,Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.,Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
91
|
Belmer A, Beecher K, Jacques A, Patkar OL, Sicherre F, Bartlett SE. Axonal Non-segregation of the Vesicular Glutamate Transporter VGLUT3 Within Serotonergic Projections in the Mouse Forebrain. Front Cell Neurosci 2019; 13:193. [PMID: 31133811 PMCID: PMC6523995 DOI: 10.3389/fncel.2019.00193] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/17/2019] [Indexed: 11/13/2022] Open
Abstract
A subpopulation of raphe 5-HT neurons expresses the vesicular glutamate transporter VGLUT3 with the co-release of glutamate and serotonin proposed to play a pivotal role in encoding reward- and anxiety-related behaviors. Serotonin axons are identifiable by immunolabeling of either serotonin (5-HT) or the plasma membrane 5-HT transporter (SERT), with SERT labeling demonstrated to be only partially overlapping with 5-HT staining. Studies investigating the colocalization or segregation of VGLUT3 within SERT or 5-HT immunolabeled boutons have led to inconsistent results. Therefore, we combined immunohistochemistry, high resolution confocal imaging, and 3D-reconstruction techniques to map and quantify the distribution of VGLUT3 immunoreactive boutons within 5-HT vs. SERT-positive axons in various regions of the mouse forebrain, including the prefrontal cortex, nucleus accumbens core and shell, bed nucleus of the stria terminalis, dorsal striatum, lateral septum, basolateral and central amygdala, and hippocampus. Our results demonstrate that about 90% of 5-HT boutons are colocalized with SERT in almost all the brain regions studied, which therefore reveals that VGLUT3 and SERT do not segregate. However, in the posterior part of the NAC shell, we confirmed the presence of a subtype of 5-HT immunoreactive axons that lack the SERT. Interestingly, about 90% of the 5-HT/VGLUT3 boutons were labeled for the SERT in this region, suggesting that VGLUT3 is preferentially located in SERT immunoreactive 5-HT boutons. This work demonstrates that VGLUT3 and SERT cannot be used as specific markers to classify the different subtypes of 5-HT axons.
Collapse
Affiliation(s)
- Arnauld Belmer
- Translational Research Institute, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Kate Beecher
- Translational Research Institute, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Angela Jacques
- Translational Research Institute, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Omkar L Patkar
- QIMR Berghofer Medical Research Institute, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Florian Sicherre
- Biologie Integrative et Physiologie, Université Pierre et Marie Curie, Paris, France
| | - Selena E Bartlett
- Translational Research Institute, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
92
|
Krawczyk MC, Millan J, Blake MG, Feld M, Boccia MM. Relevance of ERK1/2 Post-retrieval Participation on Memory Processes: Insights in Their Particular Role on Reconsolidation and Persistence of Memories. Front Mol Neurosci 2019; 12:95. [PMID: 31057366 PMCID: PMC6478671 DOI: 10.3389/fnmol.2019.00095] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 03/29/2019] [Indexed: 12/12/2022] Open
Abstract
Back in 1968, Misanin and his group posited that reactivation of consolidated memories could support changes in that trace, similar to what might happen during the consolidation process. Not until 2000, when Nader et al. (2000) studied the behavioral effect of a protein synthesis inhibitor on retrieved memories, could this previous statement be taken under consideration once again; suggesting that consolidated memories can become labile after reactivation. The process of strengthening after memory labilization was named memory reconsolidation. In recent years, many studies pointed towards a critical participation of the extracellular signal-regulated kinase (ERK)/mitogen activated protein kinases (MAPKs) pathway in different memory processes (e.g., consolidation, extinction, reconsolidation, among others). In this review article, we will focus on how this system might be modulating the processes triggered after retrieval of well-consolidated memories in mice.
Collapse
Affiliation(s)
- Maria C Krawczyk
- Laboratorio de Neurofarmacología de los Procesos de Memoria, Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Julieta Millan
- Laboratorio de Neurofarmacología de los Procesos de Memoria, Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Mariano G Blake
- Instituto de Fisiología y Biofísica (IFIBIO UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Mariana Feld
- CONICET-Universidad de Buenos Aires, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CABA, Argentina
| | - Mariano M Boccia
- Laboratorio de Neurofarmacología de los Procesos de Memoria, Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| |
Collapse
|
93
|
Stevenson RA, Hoffman JL, Maldonado-Devincci AM, Faccidomo S, Hodge CW. MGluR5 activity is required for the induction of ethanol behavioral sensitization and associated changes in ERK MAP kinase phosphorylation in the nucleus accumbens shell and lateral habenula. Behav Brain Res 2019; 367:19-27. [PMID: 30914307 DOI: 10.1016/j.bbr.2019.03.038] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/21/2019] [Accepted: 03/21/2019] [Indexed: 11/26/2022]
Abstract
Metabotropic glutamate receptor subtype-5 (mGluR5) activity regulates a variety of behavioral pathologies associated with alcohol addiction. The main goal of this study was to determine if mGluR5 regulates the induction of ethanol-induced locomotor sensitization, which is a model of experience-dependent plasticity following initial exposure to drugs of abuse. The extracellular signal-regulated kinase (ERK1/2) pathway is downstream of mGluR5 and implicated in alcohol addiction; however, its role in sensitization remains unexplored. We sought to determine if mGluR5-mediated changes in ethanol-induced sensitization are associated with changes in ERK1/2 phosphorylation (pERK1/2) in specific brain regions. Adult male DBA/2 J mice were tested for acute locomotor response to ethanol (0 or 2 g/kg, IP) followed by a 9-day induction period in which the mGluR5 antagonist MPEP (0 or 30 mg/kg, IP) was administered prior to ethanol (0 or 2.5 g/kg, IP). One day later, ethanol (2 g/kg) produced a robust within- and between-group increase in locomotor activity, indicating sensitization in mice that received MPEP (0 mg/kg) during induction. MPEP (30 mg/kg) treatment during induction resulted in locomotor response to ethanol (2 g/kg) challenge that was equivalent to an acute response, indicating full blockade of sensitization. Sensitization was associated with increased pERK1/2 immunoreactivity (IR) in nucleus accumbens shell (AcbSh) and a reduction in lateral habenula (LHb), both of which were blocked by MPEP treatment during induction. Sensitization was also associated with mGluR5-independent increases in pERK1/2 IR in the nucleus accumbens core and decreases in the dentate gyrus and lateral septum. These data indicate that mGluR5 activity is required for the induction of ethanol locomotor sensitization and associated changes in ERK1/2 phosphorylation in the AcbSh and LHb, which raises the hypothesis that mGluR5-mediated cell signaling in these brain regions may mediate the induction of sensitization. Elucidating mechanisms of sensitization may increase understanding of how ethanol hijacks behavioral functions during the development of addiction.
Collapse
Affiliation(s)
- Rebekah A Stevenson
- Center for Alcohol Studies, Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States; Department of Biology, Bucknell University, Lewisburg, PA, 17837, United States
| | - Jessica L Hoffman
- Center for Alcohol Studies, Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Antoniette M Maldonado-Devincci
- Center for Alcohol Studies, Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States; Department of Psychology, North Carolina A&T State University, Greensboro, NC, 27411, United States
| | - Sara Faccidomo
- Center for Alcohol Studies, Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Clyde W Hodge
- Center for Alcohol Studies, Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States.
| |
Collapse
|
94
|
Dobbs LK, Kaplan AR, Bock R, Phamluong K, Shin JH, Bocarsly ME, Eberhart L, Ron D, Alvarez VA. D1 receptor hypersensitivity in mice with low striatal D2 receptors facilitates select cocaine behaviors. Neuropsychopharmacology 2019; 44:805-816. [PMID: 30504927 PMCID: PMC6372593 DOI: 10.1038/s41386-018-0286-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 11/16/2018] [Accepted: 11/23/2018] [Indexed: 11/17/2022]
Abstract
Vulnerability for cocaine abuse in humans is associated with low dopamine D2 receptor (D2R) availability in the striatum. The mechanisms driving this vulnerability are poorly understood. In this study, we found that downregulating D2R expression selectively in striatal indirect-pathway neurons triggers a multitude of changes in D1 receptor (D1R)-expressing direct-pathway neurons, which comprise the other main subpopulation of striatal projection neurons. These changes include a leftward shift in the dose-response to a D1-like agonist that indicates a behavioral D1R hypersensitivity, a shift from PKA to ERK intracellular signaling cascades upon D1R activation, and a reduction in the density of bridging collaterals from D1R-expressing neurons to pallidal areas. We hypothesize that the D1R hypersensitivity underlies abuse vulnerability by facilitating the behavioral responses to repeated cocaine, such as locomotor sensitization and drug self-administration. We found evidence that littermate control mice develop D1R hypersensitivity after they are sensitized to cocaine. Indeed, D1-like agonist and cocaine cross-sensitize in control littermates and this effect was potentiated in mice lacking striatal D2Rs from indirect-pathway neurons. To our surprise, mice with low striatal D2Rs acquired cocaine self-administration similarly to littermate controls and showed no significant change in motivation to take cocaine but lower seeking. These findings indicate that downregulation of striatal D2Rs triggers D1R hypersensitivity to facilitate cocaine locomotor sensitization, which by itself was not associated with greater cocaine taking or seeking under the conditions tested.
Collapse
Affiliation(s)
- Lauren K Dobbs
- Laboratory on the Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
- Center on Compulsive Behaviors, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Alanna R Kaplan
- Laboratory on the Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Roland Bock
- Laboratory on the Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
- Center on Compulsive Behaviors, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Khanhky Phamluong
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - J Hoon Shin
- Laboratory on the Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Miriam E Bocarsly
- Laboratory on the Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
- Postdoctoral Research Associate Program, National Institute of General Medical Sciences, NIH, Bethesda, MD, USA
| | - Lindsay Eberhart
- Laboratory on the Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Dorit Ron
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Veronica A Alvarez
- Laboratory on the Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA.
- Center on Compulsive Behaviors, Intramural Research Program, NIH, Bethesda, MD, USA.
- National Institute on Drug Abuse, Intramural Research Program, NIH, Baltimore, MD, USA.
| |
Collapse
|
95
|
Valjent E, Biever A, Gangarossa G, Puighermanal E. Dopamine signaling in the striatum. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2019; 116:375-396. [PMID: 31036297 DOI: 10.1016/bs.apcsb.2019.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The striatum integrates dopamine-mediated reward signals to generate appropriate behavior in response to glutamate-mediated sensory cues. Such associative learning relies on enduring neural plasticity in striatal GABAergic spiny projection neurons which, when altered, can lead to the development of a wide variety of pathological states. Considerable progress has been made in our understanding of the intracellular signaling mechanisms in dopamine-related behaviors and pathologies. Through the prism of the regulation of histone H3 and ribosomal protein S6 phosphorylation, we review how dopamine-mediated signaling events regulate gene transcription and mRNA translation. Particularly, we focus on the intracellular cascades controlling these phosphorylations downstream of the modulation of dopamine receptors by psychostimulants, antipsychotics and l-DOPA. Finally, we highlight the importance to precisely determine in which neuronal populations these signaling events occur in order to understand how they participate in remodeling neural circuits and altering dopamine-related behaviors.
Collapse
Affiliation(s)
- Emmanuel Valjent
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France.
| | - Anne Biever
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | - Giuseppe Gangarossa
- Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Emma Puighermanal
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Autonomous University of Barcelona, Barcelona, Spain
| |
Collapse
|
96
|
Klein MO, Battagello DS, Cardoso AR, Hauser DN, Bittencourt JC, Correa RG. Dopamine: Functions, Signaling, and Association with Neurological Diseases. Cell Mol Neurobiol 2019; 39:31-59. [PMID: 30446950 DOI: 10.1007/s10571-018-0632-3] [Citation(s) in RCA: 497] [Impact Index Per Article: 99.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/02/2018] [Indexed: 02/07/2023]
Abstract
The dopaminergic system plays important roles in neuromodulation, such as motor control, motivation, reward, cognitive function, maternal, and reproductive behaviors. Dopamine is a neurotransmitter, synthesized in both central nervous system and the periphery, that exerts its actions upon binding to G protein-coupled receptors. Dopamine receptors are widely expressed in the body and function in both the peripheral and the central nervous systems. Dopaminergic signaling pathways are crucial to the maintenance of physiological processes and an unbalanced activity may lead to dysfunctions that are related to neurodegenerative diseases. Unveiling the neurobiology and the molecular mechanisms that underlie these illnesses may contribute to the development of new therapies that could promote a better quality of life for patients worldwide. In this review, we summarize the aspects of dopamine as a catecholaminergic neurotransmitter and discuss dopamine signaling pathways elicited through dopamine receptor activation in normal brain function. Furthermore, we describe the potential involvement of these signaling pathways in evoking the onset and progression of some diseases in the nervous system, such as Parkinson's, Schizophrenia, Huntington's, Attention Deficit and Hyperactivity Disorder, and Addiction. A brief description of new dopaminergic drugs recently approved and under development treatments for these ailments is also provided.
Collapse
Affiliation(s)
- Marianne O Klein
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, 05508-000, Brazil
| | - Daniella S Battagello
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, 05508-000, Brazil
| | - Ariel R Cardoso
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, 05508-000, Brazil
| | - David N Hauser
- Center for Translational Neuroscience, Sanford Burnham Prebys (SBP) Medical Discovery Institute, 10901 North Torrey Pines Rd., La Jolla, CA, 92037, USA
| | - Jackson C Bittencourt
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, 05508-000, Brazil.
- Center for Neuroscience and Behavior, Institute of Psychology, USP, São Paulo, Brazil.
| | - Ricardo G Correa
- Center for Translational Neuroscience, Sanford Burnham Prebys (SBP) Medical Discovery Institute, 10901 North Torrey Pines Rd., La Jolla, CA, 92037, USA.
| |
Collapse
|
97
|
Miyashita T, Kikuchi E, Horiuchi J, Saitoe M. Long-Term Memory Engram Cells Are Established by c-Fos/CREB Transcriptional Cycling. Cell Rep 2018; 25:2716-2728.e3. [DOI: 10.1016/j.celrep.2018.11.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 09/13/2018] [Accepted: 11/01/2018] [Indexed: 12/13/2022] Open
|
98
|
Genetic enhancement of Ras-ERK pathway does not aggravate L-DOPA-induced dyskinesia in mice but prevents the decrease induced by lovastatin. Sci Rep 2018; 8:15381. [PMID: 30337665 PMCID: PMC6194127 DOI: 10.1038/s41598-018-33713-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 10/03/2018] [Indexed: 12/21/2022] Open
Abstract
Increasing evidence supports a close relationship between Ras-ERK1/2 activation in the striatum and L-DOPA-induced dyskinesia (LID). ERK1/2 activation by L-DOPA takes place through the crosstalk between D1R/AC/PKA/DARPP-32 pathway and NMDA/Ras pathway. Compelling genetic and pharmacological evidence indicates that Ras-ERK1/2 inhibition prevents LID onset and may even revert already established dyskinetic symptoms. However, it is currently unclear whether exacerbation of Ras-ERK1/2 activity in the striatum may further aggravate dyskinesia in experimental animal models. Here we took advantage of two genetic models in which Ras-ERK1/2 signaling is hyperactivated, the Nf1+/− mice, in which the Ras inhibitor neurofibromin is reduced, and the Ras-GRF1 overexpressing (Ras-GRF1 OE) transgenic mice in which a specific neuronal activator of Ras is enhanced. Nf1+/− and Ras-GRF1 OE mice were unilaterally lesioned with 6-OHDA and treated with an escalating L-DOPA dosing regimen. In addition, a subset of Nf1+/− hemi-parkinsonian animals was also co-treated with the Ras inhibitor lovastatin. Our results revealed that Nf1+/− and Ras-GRF1 OE mice displayed similar dyskinetic symptoms to their wild-type counterparts. This observation was confirmed by the lack of differences between mutant and wild-type mice in striatal molecular changes associated to LID (i.e., FosB, and pERK1/2 expression). Interestingly, attenuation of Ras activity with lovastatin does not weaken dyskinetic symptoms in Nf1+/− mice. Altogether, these data suggest that ERK1/2-signaling activation in dyskinetic animals is maximal and does not require further genetic enhancement in the upstream Ras pathway. However, our data also demonstrate that such a genetic enhancement may reduce the efficacy of anti-dyskinetic drugs like lovastatin.
Collapse
|
99
|
Mahmoudi M, Pakpour S, Perry G. Drug-Abuse Nanotechnology: Opportunities and Challenges. ACS Chem Neurosci 2018; 9:2288-2298. [PMID: 29851334 DOI: 10.1021/acschemneuro.8b00127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Opioid drug abuse and dependence/addiction are complex disorders regulated by a wide range of interacting networks of genes and pathways that control a variety of phenotypes. Although the field has been extensively progressed since the birth of the National Institute on Drug Abuse in 1974, the fundamental knowledge and involved mechanisms that lead to drug dependence/addiction are poorly understood, and thus, there has been limited success in the prevention of drug addiction and development of therapeutics for definitive treatment and cure of addiction disease. The lack of success in both identification of addiction in at-risk populations and the development of efficient drugs has resulted in a serious social and economic burden from opioid drug abuse with global increasing rate of mortality from drug overdoses. This perspective aims to draw the attention of scientists to the potential role of nanotechnologies, which might pave the way for the development of more practical platforms for either drug development or identification and screening of patients who may be vulnerable to addiction after using opioid drugs.
Collapse
Affiliation(s)
- Morteza Mahmoudi
- Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Sepideh Pakpour
- Infectious Disease & Microbiome, Broad Institute, Cambridge, Massachusetts 02142, United States
- School of Engineering, University of British Columbia, Kelowna, BC V1V 1V7, Canada
| | - George Perry
- Neurosciences Institute and Department of Biology, College of Sciences, University of Texas, San Antonio, Texas 78249, United States
| |
Collapse
|
100
|
Jobson CLM, Renard J, Szkudlarek H, Rosen LG, Pereira B, Wright DJ, Rushlow W, Laviolette SR. Adolescent Nicotine Exposure Induces Dysregulation of Mesocorticolimbic Activity States and Depressive and Anxiety-like Prefrontal Cortical Molecular Phenotypes Persisting into Adulthood. Cereb Cortex 2018; 29:3140-3153. [DOI: 10.1093/cercor/bhy179] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/06/2018] [Indexed: 12/17/2022] Open
Abstract
Abstract
Considerable evidence demonstrates strong comorbidity between nicotine dependence and mood and anxiety disorders. Nevertheless, the neurobiological mechanisms linking adolescent nicotine exposure to mood and anxiety disorders are not known. Disturbances in the mesocorticolimbic dopamine (DA) system, comprising the prefrontal cortex (PFC), ventral tegmental area (VTA), and nucleus accumbens (NAc), are correlates of mood and anxiety-related symptoms and this circuitry is strongly influenced by acute or chronic nicotine exposure. Using a combination of behavioral pharmacology, in vivo neuronal electrophysiology and molecular analyses, we examined and compared the effects of chronic nicotine exposure in rats during adolescence versus adulthood to characterize the mechanisms by which adolescent nicotine may selectively confer increased risk of developing mood and anxiety-related symptoms in later life. We report that exposure to nicotine, selectively during adolescence, induces profound and long-lasting neuronal, molecular and behavioral disturbances involving PFC DA D1R and downstream extracellular-signal-related kinase 1-2 (ERK 1-2) signaling. Remarkably, adolescent nicotine induced a persistent state of hyperactive DA activity in the ventral tegmental area (VTA) concomitant with hyperactive neuronal activity states in the PFC. Our findings identify several unique neuronal and molecular biomarkers that may serve as functional risk mechanisms for the long-lasting neuropsychiatric effects of adolescent smoking behaviors.
Collapse
Affiliation(s)
- Christina L M Jobson
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Justine Renard
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Hanna Szkudlarek
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Laura G Rosen
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Brian Pereira
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Daniel J Wright
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Walter Rushlow
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Steven R Laviolette
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|