1
|
Mohammadkhani A, Mitchell C, James MH, Borgland SL, Dayas CV. Contribution of hypothalamic orexin (hypocretin) circuits to pathologies of motivation. Br J Pharmacol 2024; 181:4430-4449. [PMID: 39317446 PMCID: PMC11458361 DOI: 10.1111/bph.17325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 06/17/2024] [Accepted: 06/28/2024] [Indexed: 09/26/2024] Open
Abstract
The orexin (also known as hypocretin) system, consisting of neuropeptides orexin-A and orexin-B, was discovered over 25 years ago and was immediately identified as a central regulator of sleep and wakefulness. These peptides interact with two G-protein coupled receptors, orexin 1 (OX1) and orexin 2 (OX2) receptors which are capable of coupling to all heterotrimeric G-protein subfamilies, but primarily transduce increases in calcium signalling. Orexin neurons are regulated by a variety of transmitter systems and environmental stimuli that signal reward availability, including food and drug related cues. Orexin neurons are also activated by anticipation, stress, cues predicting motivationally relevant information, including those predicting drugs of abuse, and engage neuromodulatory systems, including dopamine neurons of the ventral tegmental area (VTA) to respond to these signals. As such, orexin neurons have been characterized as motivational activators that coordinate a range of functions, including feeding and arousal, that allow the individual to respond to motivationally relevant information, critical for survival. This review focuses on the role of orexins in appetitive motivation and highlights a role for these neuropeptides in pathologies characterized by inappropriately high levels of motivated arousal (overeating, anxiety and substance use disorders) versus those in which motivation is impaired (depression).
Collapse
Affiliation(s)
- Aida Mohammadkhani
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, Calgary, Alberta, Canada
| | - Caitlin Mitchell
- School of Biomedical Sciences and Pharmacy, University of Newcastle, University Drive, Callaghan, New South Wales, Australia
- The Hunter Medical Research, New Lambton Heights, New South Wales, Australia
| | - Morgan H James
- Department of Psychiatry and Brain Health Institute, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Stephanie L Borgland
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, Calgary, Alberta, Canada
| | - Christopher V Dayas
- School of Biomedical Sciences and Pharmacy, University of Newcastle, University Drive, Callaghan, New South Wales, Australia
- The Hunter Medical Research, New Lambton Heights, New South Wales, Australia
| |
Collapse
|
2
|
Kim YJ, Choi SJ, Hong SI, Park JC, Lee Y, Ma SX, Hur KH, Lee Y, Kim KM, Kim HK, Kim HY, Lee SY, Choi SY, Jang CG. The ion channel TRPA1 is a modulator of the cocaine reward circuit in the nucleus accumbens. Mol Psychiatry 2024; 29:3607-3622. [PMID: 38822069 PMCID: PMC11541219 DOI: 10.1038/s41380-024-02623-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 05/03/2024] [Accepted: 05/21/2024] [Indexed: 06/02/2024]
Abstract
Drug addiction therapies commonly fail because continued drug use promotes the release of excessive and pleasurable dopamine levels. Because the connection between pleasure and drug use becomes hard-wired in the nucleus accumbens (NAc), which interfaces motivation, effective therapies need to modulate this mesolimbic reward system. Here, we report that mice with knockdown of the cation channel TRPA1 (transient receptor potential ankyrin 1) were resistant to the drug-seeking behavior and reward effects of cocaine compared to their wildtype litter mates. In our study, we demonstrate that TRPA1 inhibition in the NAc reduces cocaine activity and dopamine release, and conversely, that TRPA1 is critical for cocaine-induced synaptic strength in dopamine receptor 1-expressing medium spiny neurons. Taken together, our data support that cocaine-induced reward-related behavior and synaptic release of dopamine in the NAc are controlled by TRPA1 and suggest that TRPA1 has therapeutic potential as a target for drug misuse therapies.
Collapse
Affiliation(s)
- Young-Jung Kim
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Su Jeong Choi
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 03080, Republic of Korea
| | - Sa-Ik Hong
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jung-Cheol Park
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 03080, Republic of Korea
| | - Youyoung Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Shi-Xun Ma
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Kwang-Hyun Hur
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Young Lee
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 03080, Republic of Korea
| | - Kyeong-Man Kim
- Pharmacology Laboratory, College of Pharmacy, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyung Kyu Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hee Young Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Seok-Yong Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Se-Young Choi
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 03080, Republic of Korea.
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
3
|
Chen K, Ashtiani KC, Monfared RV, Baldi P, Alachkar A. Circadian cilia transcriptome in mouse brain across physiological and pathological states. Mol Brain 2024; 17:67. [PMID: 39304885 PMCID: PMC11414107 DOI: 10.1186/s13041-024-01143-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024] Open
Abstract
Primary cilia are dynamic sensory organelles that continuously undergo structural modifications in response to environmental and cellular signals, many of which exhibit rhythmic patterns. Building on our previous findings of rhythmic cilia-related gene expression in diurnal primates (baboon), this study extends the investigation to the nocturnal mouse brain to identify circadian patterns of cilia gene expression across brain regions. We used computational techniques and transcriptomic data from four publicly available databases, to examine the circadian expression of cilia-associated genes within six brain areas: brainstem, cerebellum, hippocampus, hypothalamus, striatum, and suprachiasmatic nucleus. Our analysis reveals that a substantial proportion of cilia transcripts exhibit circadian rhythmicity across the examined regions, with notable overrepresentation in the striatum, hippocampus, and cerebellum. We also demonstrate region-specific variations in the abundance and timing of circadian cilia genes' peaks, indicating an adaptation to the distinct physiological roles of each brain region. Additionally, we show that the rhythmic patterns of cilia transcripts are shifted under various physiological and pathological conditions, including modulation of the dopamine system, high-fat diet, and epileptic conditions, indicating the adaptable nature of cilia transcripts' oscillation. While limited to a few mouse brain regions, our study provides initial insights into the distinct circadian patterns of cilia transcripts and highlights the need for future research to expand the mapping across wider brain areas to fully understand the role of cilia's spatiotemporal dynamics in brain functions.
Collapse
Affiliation(s)
- Kiki Chen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, 356A Med Surge II, Irvine, CA, 92697-4625, USA
| | - Kousha Changizi Ashtiani
- Departments of Computer Science, School of Information and Computer Sciences, University of California, Irvine, CA, 92697-4625, USA
| | - Roudabeh Vakil Monfared
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, 356A Med Surge II, Irvine, CA, 92697-4625, USA
| | - Pierre Baldi
- Departments of Computer Science, School of Information and Computer Sciences, University of California, Irvine, CA, 92697-4625, USA.
- Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California, Irvine, CA, 92697, USA.
| | - Amal Alachkar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, 356A Med Surge II, Irvine, CA, 92697-4625, USA.
- Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
4
|
Kawa AB, Hashimoto JG, Beutler MM, Guizzetti M, Wolf ME. Changes in nucleus accumbens core translatome accompanying incubation of cocaine craving. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.15.613147. [PMID: 39345421 PMCID: PMC11429699 DOI: 10.1101/2024.09.15.613147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
In the 'incubation of cocaine craving' model of relapse, rats exhibit progressive intensification (incubation) of cue-induced craving over several weeks of forced abstinence from cocaine self-administration. The expression of incubated craving depends on plasticity of excitatory synaptic transmission in nucleus accumbens core (NAcC) medium spiny neurons (MSN). Previously, we found that the maintenance of this plasticity and the expression of incubation depends on ongoing protein translation, and the regulation of translation is altered after incubation of cocaine craving. Here we used male and female rats that express Cre recombinase in either dopamine D1 receptor- or adenosine 2a (A2a) receptor-expressing MSN to express a GFP-tagged ribosomal protein in a cell-type specific manner, enabling us to use Translating Ribosome Affinity Purification (TRAP) to isolate actively translating mRNAs from both MSN subtypes for analysis by RNA-seq. We compared rats that self-administered saline or cocaine. Saline rats were assessed on abstinence day (AD) 1, while cocaine rats were assessed on AD1 or AD40-50. For both D1-MSN and A2a-MSN, there were few differentially translated genes between saline and cocaine AD1 groups. In contrast, pronounced differences in the translatome were observed between cocaine rats on AD1 and AD40-50, and this was far more robust in D1-MSN. Notably, all comparisons revealed sex differences in translating mRNAs. Sequencing results were validated by qRT-PCR for several genes of interest. This study, the first to combine TRAP-seq, transgenic rats, and a cocaine self-administration paradigm, identifies translating mRNAs linked to incubation of cocaine craving in D1-MSN and A2a-MSN of the NAcC.
Collapse
Affiliation(s)
- Alex B Kawa
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239
| | - Joel G Hashimoto
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239
- VA Portland Health Care System, Portland, OR 97239
| | - Madelyn M Beutler
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239
| | - Marina Guizzetti
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239
- VA Portland Health Care System, Portland, OR 97239
| | - Marina E Wolf
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239
| |
Collapse
|
5
|
Hwang EK, Wunsch AM, Wolf ME. Retinoic acid-mediated homeostatic plasticity drives cell type-specific CP-AMPAR accumulation in nucleus accumbens core and incubation of cocaine craving. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.611703. [PMID: 39314388 PMCID: PMC11419102 DOI: 10.1101/2024.09.12.611703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Incubation of cocaine craving, a translationally relevant model for the persistence of drug craving during abstinence, ultimately depends on strengthening of nucleus accumbens core (NAcc) synapses through synaptic insertion of homomeric GluA1 Ca2+-permeable AMPA receptors (CP-AMPARs). Here we tested the hypothesis that CP-AMPAR upregulation results from a form of homeostatic plasticity, previously characterized in vitro and in other brain regions, that depends on retinoic acid (RA) signaling in dendrites. Under normal conditions, ongoing synaptic transmission maintains intracellular Ca2+ at levels sufficient to suppress RA synthesis. Prolonged blockade of neuronal activity results in disinhibition of RA synthesis, leading to increased GluA1 translation and synaptic insertion of homomeric GluA1 CP-AMPARs. Using slice recordings, we found that increasing RA signaling in NAcc medium spiny neurons (MSN) from drug-naïve rats rapidly upregulates CP-AMPARs, and that this pathway is operative only in MSN expressing the D1 dopamine receptor. In MSN recorded from rats that have undergone incubation of craving, this effect of RA is occluded; instead, interruption of RA signaling in the slice normalizes the incubation-associated elevation of synaptic CP-AMPARs. Paralleling this in vitro finding, interruption of RA signaling in the NAcc of 'incubated rats' normalizes the incubation-associated elevation of cue-induced cocaine seeking. These results suggest that RA signaling becomes tonically active in the NAcc during cocaine withdrawal and, by maintaining elevated CP-AMPAR levels, contributes to the incubation of cocaine craving.
Collapse
Affiliation(s)
- Eun-Kyung Hwang
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, U.S.A. 97212
| | - Amanda M Wunsch
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, U.S.A. 97212
| | - Marina E Wolf
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, U.S.A. 97212
| |
Collapse
|
6
|
Bonnavion P, Varin C, Fakhfouri G, Martinez Olondo P, De Groote A, Cornil A, Lorenzo Lopez R, Pozuelo Fernandez E, Isingrini E, Rainer Q, Xu K, Tzavara E, Vigneault E, Dumas S, de Kerchove d'Exaerde A, Giros B. Striatal projection neurons coexpressing dopamine D1 and D2 receptors modulate the motor function of D1- and D2-SPNs. Nat Neurosci 2024; 27:1783-1793. [PMID: 38965445 DOI: 10.1038/s41593-024-01694-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/28/2024] [Indexed: 07/06/2024]
Abstract
The role of the striatum in motor control is commonly assumed to be mediated by the two striatal efferent pathways characterized by striatal projection neurons (SPNs) expressing dopamine (DA) D1 receptors or D2 receptors (D1-SPNs and D2-SPNs, respectively), without regard to SPNs coexpressing both receptors (D1/D2-SPNs). Here we developed an approach to target these hybrid SPNs in mice and demonstrate that, although these SPNs are less abundant, they have a major role in guiding the motor function of the other two populations. D1/D2-SPNs project exclusively to the external globus pallidus and have specific electrophysiological features with distinctive integration of DA signals. Gain- and loss-of-function experiments indicate that D1/D2-SPNs potentiate the prokinetic and antikinetic functions of D1-SPNs and D2-SPNs, respectively, and restrain the integrated motor response to psychostimulants. Overall, our findings demonstrate the essential role of this population of D1/D2-coexpressing neurons in orchestrating the fine-tuning of DA regulation in thalamo-cortico-striatal loops.
Collapse
Affiliation(s)
- Patricia Bonnavion
- Neurophy Lab, ULB Neuroscience Institute, Université Libre Bruxelles (ULB), Brussels, Belgium
| | - Christophe Varin
- Neurophy Lab, ULB Neuroscience Institute, Université Libre Bruxelles (ULB), Brussels, Belgium
| | - Ghazal Fakhfouri
- Department of Psychiatry, Douglas Hospital, McGill University, Montreal, Quebec, Canada
| | - Pilar Martinez Olondo
- Neurophy Lab, ULB Neuroscience Institute, Université Libre Bruxelles (ULB), Brussels, Belgium
| | - Aurélie De Groote
- Neurophy Lab, ULB Neuroscience Institute, Université Libre Bruxelles (ULB), Brussels, Belgium
| | - Amandine Cornil
- Neurophy Lab, ULB Neuroscience Institute, Université Libre Bruxelles (ULB), Brussels, Belgium
| | - Ramiro Lorenzo Lopez
- Neurophy Lab, ULB Neuroscience Institute, Université Libre Bruxelles (ULB), Brussels, Belgium
| | - Elisa Pozuelo Fernandez
- Neurophy Lab, ULB Neuroscience Institute, Université Libre Bruxelles (ULB), Brussels, Belgium
| | - Elsa Isingrini
- Department of Psychiatry, Douglas Hospital, McGill University, Montreal, Quebec, Canada
- Université Paris Cité, INCC UMR 8002, CNRS, Paris, France
| | - Quentin Rainer
- Department of Psychiatry, Douglas Hospital, McGill University, Montreal, Quebec, Canada
| | - Kathleen Xu
- Department of Psychiatry, Douglas Hospital, McGill University, Montreal, Quebec, Canada
| | - Eleni Tzavara
- Université Paris Cité, INCC UMR 8002, CNRS, Paris, France
- AP-HM, Hôpital Sainte Marguerite, Pôle Psychiatrie Universitaire Solaris, Marseille, France
| | - Erika Vigneault
- Department of Psychiatry, Douglas Hospital, McGill University, Montreal, Quebec, Canada
| | | | - Alban de Kerchove d'Exaerde
- Neurophy Lab, ULB Neuroscience Institute, Université Libre Bruxelles (ULB), Brussels, Belgium.
- WELBIO, WEL Research Institute, Wavre, Belgium.
| | - Bruno Giros
- Department of Psychiatry, Douglas Hospital, McGill University, Montreal, Quebec, Canada.
- Université Paris Cité, INCC UMR 8002, CNRS, Paris, France.
| |
Collapse
|
7
|
Chuhma N, Rayport S. Regional heterogeneity in the membrane properties of mouse striatal neurons. Front Cell Neurosci 2024; 18:1412897. [PMID: 39144155 PMCID: PMC11321984 DOI: 10.3389/fncel.2024.1412897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 06/19/2024] [Indexed: 08/16/2024] Open
Abstract
The cytoarchitecture of the striatum is remarkably homogeneous, in contrast to the regional variation in striatal functions. Whether differences in the intrinsic membrane properties of striatal neurons contribute to regional heterogeneity has not been addressed systematically. We made recordings throughout the young adult mouse striatum under identical conditions, with synaptic input blocked, from four major striatal neuron types, namely, the two subtypes of spiny projection neurons (SPNs), cholinergic interneurons (ChIs), and fast-spiking GABAergic interneurons (FSIs), sampling at least 100 cells per cell type. Regional variation manifested across all cell types. All cell types in the nucleus accumbens (NAc) shell had higher input impedance and increased excitability. Cells in the NAc core were differentiated from the caudate-putamen (CPu) for both SPN subtypes by smaller action potentials and increased excitability. Similarity between the two SPN subtypes showed regional variation, differing more in the NAc than in the CPu. So, in the Str, both the intrinsic properties of interneurons and projection neurons are regionally heterogeneous, with the greatest difference between the NAc and CPu; greater excitability of NAc shell neurons may make the region more susceptible to activity-dependent plasticity.
Collapse
Affiliation(s)
- Nao Chuhma
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
- Department of Psychiatry, Columbia University, New York, NY, United States
| | - Stephen Rayport
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
- Department of Psychiatry, Columbia University, New York, NY, United States
| |
Collapse
|
8
|
Marinescu AM, Labouesse MA. The nucleus accumbens shell: a neural hub at the interface of homeostatic and hedonic feeding. Front Neurosci 2024; 18:1437210. [PMID: 39139500 PMCID: PMC11319282 DOI: 10.3389/fnins.2024.1437210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/16/2024] [Indexed: 08/15/2024] Open
Abstract
Feeding behavior is a complex physiological process regulated by the interplay between homeostatic and hedonic feeding circuits. Among the neural structures involved, the nucleus accumbens (NAc) has emerged as a pivotal region at the interface of these two circuits. The NAc comprises distinct subregions and in this review, we focus mainly on the NAc shell (NAcSh). Homeostatic feeding circuits, primarily found in the hypothalamus, ensure the organism's balance in energy and nutrient requirements. These circuits monitor peripheral signals, such as insulin, leptin, and ghrelin, and modulate satiety and hunger states. The NAcSh receives input from these homeostatic circuits, integrating information regarding the organism's metabolic needs. Conversely, so-called hedonic feeding circuits involve all other non-hunger and -satiety processes, i.e., the sensory information, associative learning, reward, motivation and pleasure associated with food consumption. The NAcSh is interconnected with hedonics-related structures like the ventral tegmental area and prefrontal cortex and plays a key role in encoding hedonic information related to palatable food seeking or consumption. In sum, the NAcSh acts as a crucial hub in feeding behavior, integrating signals from both homeostatic and hedonic circuits, to facilitate behavioral output via its downstream projections. Moreover, the NAcSh's involvement extends beyond simple integration, as it directly impacts actions related to food consumption. In this review, we first focus on delineating the inputs targeting the NAcSh; we then present NAcSh output projections to downstream structures. Finally we discuss how the NAcSh regulates feeding behavior and can be seen as a neural hub integrating homeostatic and hedonic feeding signals, via a functionally diverse set of projection neuron subpopulations.
Collapse
Affiliation(s)
- Alina-Măriuca Marinescu
- Brain, Wire and Behavior Group, Translational Nutritional Biology Laboratory, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Marie A. Labouesse
- Brain, Wire and Behavior Group, Translational Nutritional Biology Laboratory, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
9
|
Mitra S, Werner CT, Shwani T, Lopez AG, Federico D, Higdon K, Li X, Gobira PH, Thomas SA, Martin JA, An C, Chandra R, Maze I, Neve R, Lobo MK, Gancarz AM, Dietz DM. A Novel Role for the Histone Demethylase JMJD3 in Mediating Heroin-Induced Relapse-Like Behaviors. Biol Psychiatry 2024:S0006-3223(24)01452-5. [PMID: 39019389 DOI: 10.1016/j.biopsych.2024.06.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 06/10/2024] [Accepted: 06/25/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND Epigenetic changes that lead to long-term neuroadaptations following opioid exposure are not well understood. We examined how histone demethylase JMJD3 in the nucleus accumbens (NAc) influences heroin seeking after abstinence from self-administration. METHODS Male Sprague Dawley rats were trained to self-administer heroin. Western blotting and quantitative polymerase chain reaction were performed to quantify JMJD3 and bone morphogenetic protein (BMP) pathway expression in the NAc (n = 7-11/group). Pharmacological inhibitors or viral expression vectors were microinfused into the NAc to manipulate JMJD3 or the BMP pathway member SMAD1 (n = 9-11/group). The RiboTag capture method (n = 3-5/group) and viral vectors (n = 7-8/group) were used in male transgenic rats to identify the contributions of D1- and D2-expressing medium spiny neurons in the NAc. Drug seeking was tested by cue-induced response previously paired with drug infusion. RESULTS Levels of JMJD3 and phosphorylated SMAD1/5 in the NAc were increased after 14 days of abstinence from heroin self-administration. Pharmacological and virus-mediated inhibition of JMJD3 or the BMP pathway attenuated cue-induced seeking. Pharmacological inhibition of BMP signaling reduced JMJD3 expression and H3K27me3 levels. JMJD3 bidirectionally affected seeking: expression of the wild-type increased cue-induced seeking whereas expression of a catalytic dead mutant decreased it. JMJD3 expression was increased in D2+ but not D1+ medium spiny neurons. Expression of the mutant JMJD3 in D2+ neurons was sufficient to decrease cue-induced heroin seeking. CONCLUSIONS JMJD3 mediates persistent cellular and behavioral adaptations that underlie heroin relapse, and this activity is regulated by the BMP pathway.
Collapse
Affiliation(s)
- Swarup Mitra
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Craig T Werner
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Treefa Shwani
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Ana Garcia Lopez
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Dale Federico
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Kate Higdon
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Xiaofang Li
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Pedro H Gobira
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Shruthi A Thomas
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Jennifer A Martin
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Chunna An
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Ramesh Chandra
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ian Maze
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Howard Hughes Medical Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Rachel Neve
- Gene Technology Core, Massachusetts General Hospital, Cambridge, Massachusetts
| | - Mary Kay Lobo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Amy M Gancarz
- Department of Psychology, California State University, Bakersfield, Bakersfield, California
| | - David M Dietz
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York.
| |
Collapse
|
10
|
Andraka E, Phillips RA, Brida KL, Day JJ. Chst9 marks a spatially and transcriptionally unique population of Oprm1-expressing neurons in the nucleus accumbens. ADDICTION NEUROSCIENCE 2024; 11:100153. [PMID: 38957401 PMCID: PMC11218735 DOI: 10.1016/j.addicn.2024.100153] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Opioids produce addictive, analgesic, and euphoric effects via actions at mu opioid receptors (μORs). The μOR is encoded by the Oprm1 gene and is expressed in multiple brain regions that regulate reward and motivation, such as the nucleus accumbens (NAc). Oprm1 expression in NAc medium spiny neurons (MSNs) mediates opioid place preference, seeking, and consumption. However, recent single nucleus RNA sequencing (snRNA-seq) studies have revealed that multiple subpopulations of NAc neurons express Oprm1 mRNA, making it unclear which populations mediate diverse behaviors resulting from μOR activation. Using published snRNA-seq datasets from the rat NAc, we identified a novel population of MSNs that express the highest levels of Oprm1 of any NAc cell type. Here, we show that this population is selectively marked by expression of Chst9, a gene encoding a carbohydrate sulfotransferase. Notably, Chst9+ neurons exhibited more abundant expression of Oprm1 as compared to other cell types, and formed discrete cellular clusters along the medial and ventral borders of the NAc shell subregion. Moreover, CHST9 mRNA was also found to mark specific MSN populations in published human and primate snRNA-seq studies, indicating that this unique population may be conserved across species. Together, these results identify a spatially and transcriptionally distinct NAc neuron population characterized by the expression of Chst9. The abundant expression of Oprm1 in this population and the conservation of these cells across species suggests that they may play a key functional role in opioid response and identify this subpopulation as a target for further investigation.
Collapse
Affiliation(s)
- Emma Andraka
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Robert A. Phillips
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Kasey L. Brida
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Jeremy J. Day
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| |
Collapse
|
11
|
Kim YJ, Kook WA, Ma SX, Lee BR, Ko YH, Kim SK, Lee Y, Lee JG, Lee S, Kim KM, Lee SY, Jang CG. The novel psychoactive substance 25E-NBOMe induces reward-related behaviors via dopamine D1 receptor signaling in male rodents. Arch Pharm Res 2024; 47:360-376. [PMID: 38551761 DOI: 10.1007/s12272-024-01491-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 03/20/2024] [Indexed: 04/26/2024]
Abstract
Novel psychoactive substances (NPSs) are new psychotropic drugs designed to evade substance regulatory policies. 25E-NBOMe (2-(4-ethyl-2,5-dimethoxyphenyl)-N-(2-methoxybenzyl)ethanamine) has recently been identified as an NPS, and its recreational misuse has been reported to be rapidly increasing. However, the psychopharmacological effects and mechanisms of 25E-NBOMe have not been studied. We examined the abuse potential of 25E-NBOMe using the conditioned place preference in male mice and self-administration paradigms in male rats. Additionally, immunoblot assay, enzyme-linked immunosorbent assay, and microdialysis were used to determine the molecular effects of 25E-NBOMe in the nucleus accumbens (NAc). Our data demonstrated that 25E-NBOMe induces conditioned place preference, and the dopaminergic signaling in the NAc mediates these. Following 25E-NBOMe administration, expression of dopamine transporter and dopamine D1 receptor (D1DR) were enhanced in the NAc of male mice, and NAc dopamine levels were reduced in both male mice and rats. Induction of intracellular dopaminergic pathways, DARPP32, and phosphorylation of CREB in the NAc of male mice was also observed. Significantly, pharmacological blockade of D1DR or chemogenetic inhibition of D1DR-expressing medium spiny neurons in the NAc attenuated 25E-NBOMe-induced conditioned place preference in male mice. We also examined the hallucinogenic properties of 25E-NBOMe using the head twitch response test in male mice and found that this behavior was mediated by serotonin 2A receptor activity. Our findings demonstrate that D1DR signaling may govern the addictive potential of 25E-NBOMe. Moreover, our study provides new insights into the potential mechanisms of substance use disorder and the improvement of controlled substance management.
Collapse
Affiliation(s)
- Young-Jung Kim
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Wun-A Kook
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Shi-Xun Ma
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Bo-Ram Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yong-Hyun Ko
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Seon-Kyung Kim
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Youyoung Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jae-Gyeong Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Sooyeun Lee
- Analytical Toxicology Laboratory, College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea
| | - Kyeong-Man Kim
- Pharmacology Laboratory, College of Pharmacy, Chonnam National University, Gwangju, 81186, Republic of Korea
| | - Seok-Yong Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
12
|
Espallergues J, Boubaker-Vitre J, Mignon A, Avrillon M, Le Bon-Jego M, Baufreton J, Valjent E. Spatiomolecular Characterization of Dopamine D2 Receptors Cells in the Mouse External Globus Pallidus. Curr Neuropharmacol 2024; 22:1528-1539. [PMID: 37475558 PMCID: PMC11097984 DOI: 10.2174/1570159x21666230720121027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/07/2023] [Accepted: 04/10/2023] [Indexed: 07/22/2023] Open
Abstract
The external globus pallidus (GPe) is part of the basal ganglia circuit and plays a key role in controlling the actions. Although, many evidence indicate that dopamine through its activation of dopamine D2 receptors (D2Rs) modulates the GPe neuronal activity, the precise spatiomolecular characterization of cell populations expressing D2Rs in the mouse GPe is still lacking. By combining single molecule in situ hybridization, cell type-specific imaging analyses, and electrophysiology slice recordings, we found that GPe D2R cells are neurons preferentially localized in the caudal portion of GPe. These neurons comprising pallido-striatal, pallido-nigral, and pallido-cortical neurons segregate into two distinct populations displaying molecular and electrophysiological features of GPe GABAergic PV/NKX2.1 and cholinergic neurons respectively. By clarifying the spatial molecular identity of GPe D2R neurons in the mouse, this work provides the basis for future studies aiming at disentangling the action of dopamine within the GPe.
Collapse
Affiliation(s)
| | | | - Audrey Mignon
- IGF, University Montpellier, CNRS, Inserm, F-34094 Montpellier, France
| | - Maelle Avrillon
- IGF, University Montpellier, CNRS, Inserm, F-34094 Montpellier, France
| | | | - Jerome Baufreton
- University Bordeaux, CNRS, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Emmanuel Valjent
- IGF, University Montpellier, CNRS, Inserm, F-34094 Montpellier, France
| |
Collapse
|
13
|
Phillips RA, Wan E, Tuscher JJ, Reid D, Drake OR, Ianov L, Day JJ. Temporally specific gene expression and chromatin remodeling programs regulate a conserved Pdyn enhancer. eLife 2023; 12:RP89993. [PMID: 37938195 PMCID: PMC10631760 DOI: 10.7554/elife.89993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023] Open
Abstract
Neuronal and behavioral adaptations to novel stimuli are regulated by temporally dynamic waves of transcriptional activity, which shape neuronal function and guide enduring plasticity. Neuronal activation promotes expression of an immediate early gene (IEG) program comprised primarily of activity-dependent transcription factors, which are thought to regulate a second set of late response genes (LRGs). However, while the mechanisms governing IEG activation have been well studied, the molecular interplay between IEGs and LRGs remain poorly characterized. Here, we used transcriptomic and chromatin accessibility profiling to define activity-driven responses in rat striatal neurons. As expected, neuronal depolarization generated robust changes in gene expression, with early changes (1 hr) enriched for inducible transcription factors and later changes (4 hr) enriched for neuropeptides, synaptic proteins, and ion channels. Remarkably, while depolarization did not induce chromatin remodeling after 1 hr, we found broad increases in chromatin accessibility at thousands of sites in the genome at 4 hr after neuronal stimulation. These putative regulatory elements were found almost exclusively at non-coding regions of the genome, and harbored consensus motifs for numerous activity-dependent transcription factors such as AP-1. Furthermore, blocking protein synthesis prevented activity-dependent chromatin remodeling, suggesting that IEG proteins are required for this process. Targeted analysis of LRG loci identified a putative enhancer upstream of Pdyn (prodynorphin), a gene encoding an opioid neuropeptide implicated in motivated behavior and neuropsychiatric disease states. CRISPR-based functional assays demonstrated that this enhancer is both necessary and sufficient for Pdyn transcription. This regulatory element is also conserved at the human PDYN locus, where its activation is sufficient to drive PDYN transcription in human cells. These results suggest that IEGs participate in chromatin remodeling at enhancers and identify a conserved enhancer that may act as a therapeutic target for brain disorders involving dysregulation of Pdyn.
Collapse
Affiliation(s)
- Robert A Phillips
- Department of Neurobiology, University of Alabama at BirminghamBirminghamUnited States
| | - Ethan Wan
- Department of Neurobiology, University of Alabama at BirminghamBirminghamUnited States
| | - Jennifer J Tuscher
- Department of Neurobiology, University of Alabama at BirminghamBirminghamUnited States
| | - David Reid
- Department of Neurobiology, University of Alabama at BirminghamBirminghamUnited States
| | - Olivia R Drake
- Department of Neurobiology, University of Alabama at BirminghamBirminghamUnited States
| | - Lara Ianov
- Department of Neurobiology, University of Alabama at BirminghamBirminghamUnited States
- Civitan International Research Center, University of Alabama at BirminghamBirminghamUnited States
| | - Jeremy J Day
- Department of Neurobiology, University of Alabama at BirminghamBirminghamUnited States
| |
Collapse
|
14
|
Kokane SS, Butler BD, Antonio JH, Armant RJ, Hoch AC, Coelho CS, Brady BN, Chamseddine HH, Perrotti LI. Interactions between estradiol and ERK, but not mTOR, signaling is necessary for enhanced cocaine-induced conditioned place preference in female rats. Pharmacol Biochem Behav 2023; 232:173653. [PMID: 37804867 DOI: 10.1016/j.pbb.2023.173653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/12/2023] [Accepted: 10/03/2023] [Indexed: 10/09/2023]
Abstract
Women rapidly progress from recreational cocaine use to dependence, consume greater quantities of cocaine, experience more positive subjective effects of cocaine and have higher incidences of relapse during abstinence. These effects have been replicated in animal models of cocaine addiction and indicate an enhanced sensitivity and therefore, vulnerability of females to cocaine addiction. Furthermore, it has been demonstrated that estradiol (E2) is a key mediator of the aforementioned effects of cocaine in women and female animals. However, studies identifying the influence of E2 on cocaine-associated reward and its underlying neurobiological mechanisms are lacking. Here, we further explored the influence of E2 on cocaine conditioned place preference in female rats. We show that E2 mediates cocaine-conditioned reward by potentiating cocaine-context associations. In addition, the E2-mediated increases in cocaine-induced CPP are associated with increased activation of ERK1/2 and mTOR proteins in the nucleus accumbens, dorsal striatum, and ventral tegmental area. To assess the involvement of ERK1/2 and mTOR in E2-mediated enhanced cocaine-CPP, we inhibited ERK1/2 and/or mTOR activity during cocaine-conditioning and before CPP-test. Inhibition of ERK1/2 during conditioning blocked cocaine-CPP in females, inhibition mTOR was without effect, and inhibiting ERK1/2 and mTOR before CPP-test blocked cocaine-CPP. In conclusion, we have established that E2 enhances cocaine-conditioned reward by potentiating cocaine-context associations formed during conditioning. Additionally, activation of ERK1/2 during cocaine-conditioning is necessary for the potentiation of cocaine-conditioned reward by E2. SIGNIFICANCE STATEMENT: Studies characterizing the molecular substrates underlying the effects of E2 during the formation of cocaine-context associations are virtually unknown. In this study, we established the influence of E2 during the formation of cocaine-CPP and characterized the role of ERK1/2 and mTOR activity on this effect within significant nodes of the reward pathway. The elucidation of the role of E2 in cocaine-induced intracellular signaling fills a significant gap in our knowledge regarding the mechanisms by which E2 affects intracellular signaling pathways to indicate the motivational salience of a stimulus. These data are crucial to our understanding of how fluctuating hormone levels can render females increasing sensitive to the rewarding effects of cocaine.
Collapse
Affiliation(s)
- Saurabh S Kokane
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Brandon D Butler
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Josimar Hernandez Antonio
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Ross J Armant
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Adam C Hoch
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Clinton S Coelho
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Blake N Brady
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Houda H Chamseddine
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Linda I Perrotti
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA.
| |
Collapse
|
15
|
Andraka E, Phillips RA, Brida KL, Day JJ. Chst9 Marks a Spatially and Transcriptionally Unique Population of Oprm1 -Expressing Neurons in the Nucleus Accumbens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.16.562623. [PMID: 37904940 PMCID: PMC10614864 DOI: 10.1101/2023.10.16.562623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Opioids produce addictive, analgesic, and euphoric effects via actions at mu opioid receptors (μORs). The μOR is encoded by the Oprm1 gene and is expressed in multiple brain regions that regulate reward and motivation, such as the nucleus accumbens (NAc). Oprm1 expression in NAc medium spiny neurons (MSNs) mediates opioid place preference, seeking, and consumption. However, recent single nucleus RNA sequencing (snRNA-seq) studies in rodent, primate, and human NAc have revealed that multiple subpopulations of NAc neurons express Oprm1 mRNA, making it unclear which populations mediate diverse behaviors resulting from μOR activation. Using published snRNA-seq datasets from the rat NAc, we identified a novel population of MSNs that express the highest levels of Oprm1 of any NAc cell type. Here, we show that this population is selectively marked by expression of Chst9 , a gene encoding a carbohydrate sulfotransferase. To validate this observation and characterize spatial localization of this population in the rat NAc, we performed multiplexed RNAscope fluorescence in situ hybridization studies to detect expression of Oprm1 and Chst9 mRNA along with well-validated markers of MSNs. Notably, Chst9 + neurons exhibited more abundant expression of Oprm1 as compared to other cell types, and formed discrete cellular clusters along the medial and ventral borders of the NAc shell subregion. Moreover, CHST9 mRNA was also found to mark specific MSN populations in published human and primate snRNA-seq studies, indicating that this unique population may be conserved across species. Together, these results identify a spatially and transcriptionally distinct NAc neuron population characterized by the expression of Chst9 . The abundant expression of Oprm1 in this population and the conservation of these cells across species suggests that they may play a key functional role in opioid response and identify this subpopulation as a target for further investigation.
Collapse
|
16
|
Conn KA, Alexander S, Burne THJ, Kesby JP. Antagonism of D2 receptors via raclopride ameliorates amphetamine-induced associative learning deficits in male mice. Behav Brain Res 2023; 454:114649. [PMID: 37643667 DOI: 10.1016/j.bbr.2023.114649] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/09/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023]
Abstract
Dopamine levels in the dorsomedial striatum (DMS) are highly dynamic and are thought to underly the encoding of action-outcome associations. Although it is known that amphetamine disrupts the learning that is required for goal-directed action, the role of D1 and D2 receptors in this process has not been established. In this study, we examined the role of D1 and D2 receptor antagonists on learning in response to amphetamine. We used the outcome-specific devaluation task to examine goal-directed action in male C57BL6/J mice treated systemically with either a D1 antagonist (SCH-23990; 0.01 mg/kg) or a D2 antagonist (raclopride; 0.5 mg/kg) and then administered amphetamine (1 mg/kg). The mice were injected repeatedly throughout the instrumental training phase of the task to assess the impact on the learning of action-outcomes, and the subsequent choice test assessing performance of goal-directed action was conducted drug free. Effects of chronic drug administration on locomotor behaviour was assessed before and after the choice test. Treatment during learning with either amphetamine, or the D1 or D2 antagonists, impaired the subsequent performance of goal-directed action. The amphetamine-induced impairment in goal-directed action was reversed in mice treated with raclopride, but not when treated with SCH-23990. By contrast, amphetamine-induced hyperactivity was reversed in mice treated with SCH-23990, but not in mice treated with raclopride. Taken together, these data support the role of a balance of dopamine receptor signalling after amphetamine treatment. While overall D1 receptor availability is necessary to promote learning, in a state of elevated dopamine, modifying D2 receptor function can ameliorate learning deficits.
Collapse
Affiliation(s)
- Kyna-Anne Conn
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Suzy Alexander
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Thomas H J Burne
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4072, Australia; Queensland Centre for Mental Health Research, Wacol, QLD 4076, Australia
| | - James P Kesby
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4072, Australia; Queensland Centre for Mental Health Research, Wacol, QLD 4076, Australia.
| |
Collapse
|
17
|
Sun H, Li Z, Qiu Z, Shen Y, Guo Q, Hu SW, Ding HL, An S, Cao JL. A common neuronal ensemble in nucleus accumbens regulates pain-like behaviour and sleep. Nat Commun 2023; 14:4700. [PMID: 37543693 PMCID: PMC10404280 DOI: 10.1038/s41467-023-40450-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 07/28/2023] [Indexed: 08/07/2023] Open
Abstract
A comorbidity of chronic pain is sleep disturbance. Here, we identify a dual-functional ensemble that regulates both pain-like behaviour induced by chronic constrictive injury or complete Freund's adjuvant, and sleep wakefulness, in the nucleus accumbens (NAc) in mice. Specifically, a select population of NAc neurons exhibits increased activity either upon nociceptive stimulation or during wakefulness. Experimental activation of the ensemble neurons exacerbates pain-like (nociceptive) responses and reduces NREM sleep, while inactivation of these neurons produces the opposite effects. Furthermore, NAc ensemble primarily consists of D1 neurons and projects divergently to the ventral tegmental area (VTA) and preoptic area (POA). Silencing an ensemble innervating VTA neurons selectively increases nociceptive responses without affecting sleep, whereas inhibiting ensemble-innervating POA neurons decreases NREM sleep without affecting nociception. These results suggest a common NAc ensemble that encodes chronic pain and controls sleep, and achieves the modality specificity through its divergent downstream circuit targets.
Collapse
Affiliation(s)
- Haiyan Sun
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
- Department of Pediatrics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China
| | - Zhilin Li
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Zhentong Qiu
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Yu Shen
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Qingchen Guo
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Su-Wan Hu
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Hai-Lei Ding
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Shuming An
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China.
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China.
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, China.
| |
Collapse
|
18
|
Wang X, Yin D, Tang Y, Xiao F, Wang S. Psychiatric adverse reactions to non-selective RET multi-kinase inhibitors: a large-scale pharmacovigilance analysis. Front Pharmacol 2023; 14:1209933. [PMID: 37465525 PMCID: PMC10351039 DOI: 10.3389/fphar.2023.1209933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 06/22/2023] [Indexed: 07/20/2023] Open
Abstract
Objective: The development of non-selective multi-kinase inhibitors (MKIs) has improved the. survival outcomes of patients with cancers. Psychiatric disorders represent an MKIs related AE of particular concern, as they are often ignored and may harm the patient's personal and social functioning. Therefore, we use the public database to describe and evaluate psychiatric adverse events related to various non-selective RET MKIs. Provide evidence for optimizing drug administration in the clinic. Methods: We analyzed spontaneous reports submitted to the Food and Drug Administration Adverse Events Reporting System FDA Adverse Event Reporting System in an observational and retrospective manner. Selecting psychiatric AEs to non-selective RET multikinase inhibitors (sorafenib, lenvatinib, vandetanib, cabozantinib, and sunitinib). We used Reporting Odds Ratio (ROR), Proportional Reporting Ratio (PRR), Bayesian Confidence Propagation Neural Network (BCPNN), and multi-item gamma Poisson shrinker (MGPS) algorithms to analyze suspected adverse reactions of psychiatric related induced by non-selective RET MKIs between January 2004 and September 2022. Results: As of September 2022, 1,108 non-selective RET MKIs ICSRs were related to psychiatric AEs. 706 were ADR ICSRs, and 402 were non-ADR ICSRs. There were more ADR cases in males (69.5%), and 71.8% of the cases were submitted from North America. The age group most frequently affected by psychiatric ADRs was individuals aged 50-64 years for sorafenib, whereas 65-74 years for sunitinib, cabozantinib, and lenvatinib. In all psychiatric ADRs ICSRs, excluding missing data (n = 329), the most common adverse outcome was hospitalization (260/377, 69.0%), and the most serious was death (100/377, 26.5%). What calls for special attention is that the percentage of death rate for sunitinib was highest (24/54, 44.4%) in sunitinib-related psychiatric ADRs ICSRs, (excluding missing data, n = 44), followed by lenvatinib (4/14, 28.6%). Based on ROR, PRR, BCPNN, and MGPS methods, sorafenib, sunitinib, cabozantinib, and lenvatinib are significantly associated with all ADRs, the strongest association was the association between cabozantinib and feeding disorder. Conclusion: Despite the limitations, our study found that, except for vandetanib, other four drugs have been reported to have significant psychiatric side effects. Clinicians need to recognize and monitor these potentially fatal adverse events. If it is suitable for treatment with vandetanib, doctors should choose vandetanib for treatment.
Collapse
Affiliation(s)
- Xuyan Wang
- Central Laboratory, Shanxi Hospital of Integrated Traditional Chinese and Western Medicine, Taiyuan, Shanxi, China
| | - Donghong Yin
- Department of Pharmacy, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yang Tang
- Department of Pharmacy, School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Feng Xiao
- Department of Oncology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Shuyun Wang
- Department of Pharmacy, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
19
|
Phillips RA, Tuscher JJ, Fitzgerald ND, Wan E, Zipperly ME, Duke CG, Ianov L, Day JJ. Distinct subpopulations of D1 medium spiny neurons exhibit unique transcriptional responsiveness to cocaine. Mol Cell Neurosci 2023; 125:103849. [PMID: 36965548 PMCID: PMC10898607 DOI: 10.1016/j.mcn.2023.103849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/13/2023] [Accepted: 03/18/2023] [Indexed: 03/27/2023] Open
Abstract
Drugs of abuse increase extracellular concentrations of dopamine in the nucleus accumbens (NAc), resulting in transcriptional alterations that drive long-lasting cellular and behavioral adaptations. While decades of research have focused on the transcriptional mechanisms by which drugs of abuse influence neuronal physiology and function, few studies have comprehensively defined NAc cell type heterogeneity in transcriptional responses to drugs of abuse. Here, we used single nucleus RNA-seq (snRNA-seq) to characterize the transcriptome of over 39,000 NAc cells from male and female adult Sprague-Dawley rats following acute or repeated cocaine experience. This dataset identified 16 transcriptionally distinct cell populations, including two populations of medium spiny neurons (MSNs) that express the Drd1 dopamine receptor (D1-MSNs). Critically, while both populations expressed classic marker genes of D1-MSNs, only one population exhibited a robust transcriptional response to cocaine. Validation of population-selective transcripts using RNA in situ hybridization revealed distinct spatial compartmentalization of these D1-MSN populations within the NAc. Finally, analysis of published NAc snRNA-seq datasets from non-human primates and humans demonstrated conservation of MSN subtypes across rat and higher order mammals, and further highlighted cell type-specific transcriptional differences across the NAc and broader striatum. These results highlight the utility in using snRNA-seq to characterize both cell type heterogeneity and cell type-specific responses to cocaine and provides a useful resource for cross-species comparisons of NAc cell composition.
Collapse
Affiliation(s)
- Robert A Phillips
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jennifer J Tuscher
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - N Dalton Fitzgerald
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ethan Wan
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Morgan E Zipperly
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Corey G Duke
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lara Ianov
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jeremy J Day
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
20
|
Giua G, Lassalle O, Makrini-Maleville L, Valjent E, Chavis P, Manzoni OJJ. Investigating cell-specific effects of FMRP deficiency on spiny projection neurons in a mouse model of Fragile X syndrome. Front Cell Neurosci 2023; 17:1146647. [PMID: 37323585 PMCID: PMC10264852 DOI: 10.3389/fncel.2023.1146647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/16/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction Fragile X syndrome (FXS), resulting from a mutation in the Fmr1 gene, is the most common monogenic cause of autism and inherited intellectual disability. Fmr1 encodes the Fragile X Messenger Ribonucleoprotein (FMRP), and its absence leads to cognitive, emotional, and social deficits compatible with the nucleus accumbens (NAc) dysfunction. This structure is pivotal in social behavior control, consisting mainly of spiny projection neurons (SPNs), distinguished by dopamine D1 or D2 receptor expression, connectivity, and associated behavioral functions. This study aims to examine how FMRP absence differentially affects SPN cellular properties, which is crucial for categorizing FXS cellular endophenotypes. Methods We utilized a novel Fmr1-/y::Drd1a-tdTomato mouse model, which allows in-situ identification of SPN subtypes in FXS mice. Using RNA-sequencing, RNAScope and ex-vivo patch-clamp in adult male mice NAc, we comprehensively compared the intrinsic passive and active properties of SPN subtypes. Results Fmr1 transcripts and their gene product, FMRP, were found in both SPNs subtypes, indicating potential cell-specific functions for Fmr1. The study found that the distinguishing membrane properties and action potential kinetics typically separating D1- from D2-SPNs in wild-type mice were either reversed or abolished in Fmr1-/y::Drd1a-tdTomato mice. Interestingly, multivariate analysis highlighted the compound effects of Fmr1 ablation by disclosing how the phenotypic traits distinguishing each cell type in wild-type mice were altered in FXS. Discussion Our results suggest that the absence of FMRP disrupts the standard dichotomy characterizing NAc D1- and D2-SPNs, resulting in a homogenous phenotype. This shift in cellular properties could potentially underpin select aspects of the pathology observed in FXS. Therefore, understanding the nuanced effects of FMRP absence on SPN subtypes can offer valuable insights into the pathophysiology of FXS, opening avenues for potential therapeutic strategies.
Collapse
Affiliation(s)
- Gabriele Giua
- INMED, INSERM U1249, Marseille, France
- Aix-Marseille University, Marseille, France
- Cannalab “Cannabinoids Neuroscience Research International Associated Laboratory”, INSERM-Aix-Marseille University/Indiana University, Marseille, France
| | - Olivier Lassalle
- INMED, INSERM U1249, Marseille, France
- Aix-Marseille University, Marseille, France
- Cannalab “Cannabinoids Neuroscience Research International Associated Laboratory”, INSERM-Aix-Marseille University/Indiana University, Marseille, France
| | | | - Emmanuel Valjent
- IGF, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Pascale Chavis
- INMED, INSERM U1249, Marseille, France
- Aix-Marseille University, Marseille, France
- Cannalab “Cannabinoids Neuroscience Research International Associated Laboratory”, INSERM-Aix-Marseille University/Indiana University, Marseille, France
| | - Olivier J. J. Manzoni
- INMED, INSERM U1249, Marseille, France
- Aix-Marseille University, Marseille, France
- Cannalab “Cannabinoids Neuroscience Research International Associated Laboratory”, INSERM-Aix-Marseille University/Indiana University, Marseille, France
| |
Collapse
|
21
|
Sullivan M, Fernandez-Aranda F, Camacho-Barcia L, Harkin A, Macrì S, Mora-Maltas B, Jiménez-Murcia S, O'Leary A, Ottomana AM, Presta M, Slattery D, Scholtz S, Glennon JC. Insulin and Disorders of Behavioural Flexibility. Neurosci Biobehav Rev 2023; 150:105169. [PMID: 37059405 DOI: 10.1016/j.neubiorev.2023.105169] [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: 12/30/2022] [Revised: 04/03/2023] [Accepted: 04/10/2023] [Indexed: 04/16/2023]
Abstract
Behavioural inflexibility is a symptom of neuropsychiatric and neurodegenerative disorders such as Obsessive-Compulsive Disorder, Autism Spectrum Disorder and Alzheimer's Disease, encompassing the maintenance of a behaviour even when no longer appropriate. Recent evidence suggests that insulin signalling has roles apart from its regulation of peripheral metabolism and mediates behaviourally-relevant central nervous system (CNS) functions including behavioural flexibility. Indeed, insulin resistance is reported to generate anxious, perseverative phenotypes in animal models, with the Type 2 diabetes medication metformin proving to be beneficial for disorders including Alzheimer's Disease. Structural and functional neuroimaging studies of Type 2 diabetes patients have highlighted aberrant connectivity in regions governing salience detection, attention, inhibition and memory. As currently available therapeutic strategies feature high rates of resistance, there is an urgent need to better understand the complex aetiology of behaviour and develop improved therapeutics. In this review, we explore the circuitry underlying behavioural flexibility, changes in Type 2 diabetes, the role of insulin in CNS outcomes and mechanisms of insulin involvement across disorders of behavioural inflexibility.
Collapse
Affiliation(s)
- Mairéad Sullivan
- Conway Institute of Biomedical and Biomolecular Research, School of Medicine, University College Dublin, Dublin, Ireland.
| | - Fernando Fernandez-Aranda
- Department of Psychiatry, University Hospital of Bellvitge, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Barcelona, Spain; Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Lucía Camacho-Barcia
- Department of Psychiatry, University Hospital of Bellvitge, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Barcelona, Spain
| | - Andrew Harkin
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland
| | - Simone Macrì
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Bernat Mora-Maltas
- Department of Psychiatry, University Hospital of Bellvitge, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Susana Jiménez-Murcia
- Department of Psychiatry, University Hospital of Bellvitge, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Barcelona, Spain; Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Aet O'Leary
- University Hospital Frankfurt, Frankfurt, Germany
| | - Angela Maria Ottomana
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; Neuroscience Unit, Department of Medicine, University of Parma, 43100 Parma, Italy
| | - Martina Presta
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
| | | | | | - Jeffrey C Glennon
- Conway Institute of Biomedical and Biomolecular Research, School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
22
|
Korlatowicz A, Kolasa M, Pabian P, Solich J, Latocha K, Dziedzicka-Wasylewska M, Faron-Górecka A. Altered Intracellular Signaling Associated with Dopamine D2 Receptor in the Prefrontal Cortex in Wistar Kyoto Rats. Int J Mol Sci 2023; 24:ijms24065941. [PMID: 36983013 PMCID: PMC10056486 DOI: 10.3390/ijms24065941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/13/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Wistar-Kyoto rats (WKY), compared to Wistar rats, are a well-validated animal model for drug-resistant depression. Thanks to this, they can provide information on the potential mechanisms of treatment-resistant depression. Since deep brain stimulation in the prefrontal cortex has been shown to produce rapid antidepressant effects in WKY rats, we focused our study on the prefrontal cortex. Using quantitative autoradiography, we observed a decrease in the binding of [3H] methylspiperone to the dopamine D2 receptor, specifically in that brain region-but not in the striatum, nor the nucleus accumbens-in WKY rats. Further, we focused our studies on the expression level of several components associated with canonical (G proteins), as well as non-canonical, D2-receptor-associated intracellular pathways (e.g., βarrestin2, glycogen synthase kinase 3 beta-Gsk-3β, and β-catenin). As a result, we observed an increase in the expression of mRNA encoding the regulator of G protein signaling 2-RGS2 protein, which is responsible, among other things, for internalizing the D2 dopamine receptor. The increase in RGS2 expression may therefore account for the decreased binding of the radioligand to the D2 receptor. In addition, WKY rats are characterized by the altered signaling of genes associated with the dopamine D2 receptor and the βarrestin2/AKT/Gsk-3β/β-catenin pathway, which may account for certain behavioral traits of this strain and for the treatment-resistant phenotype.
Collapse
Affiliation(s)
- Agata Korlatowicz
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Magdalena Kolasa
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Paulina Pabian
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Joanna Solich
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Katarzyna Latocha
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Marta Dziedzicka-Wasylewska
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Agata Faron-Górecka
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| |
Collapse
|
23
|
He ZX, Xi K, Liu KJ, Yue MH, Wang Y, Yin YY, Liu L, He XX, Yu HL, Xing ZK, Zhu XJ. A Nucleus Accumbens Tac1 Neural Circuit Regulates Avoidance Responses to Aversive Stimuli. Int J Mol Sci 2023; 24:ijms24054346. [PMID: 36901777 PMCID: PMC10001899 DOI: 10.3390/ijms24054346] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/20/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
Neural circuits that control aversion are essential for motivational regulation and survival in animals. The nucleus accumbens (NAc) plays an important role in predicting aversive events and translating motivations into actions. However, the NAc circuits that mediate aversive behaviors remain elusive. Here, we report that tachykinin precursor 1 (Tac1) neurons in the NAc medial shell regulate avoidance responses to aversive stimuli. We show that NAcTac1 neurons project to the lateral hypothalamic area (LH) and that the NAcTac1→LH pathway contributes to avoidance responses. Moreover, the medial prefrontal cortex (mPFC) sends excitatory inputs to the NAc, and this circuit is involved in the regulation of avoidance responses to aversive stimuli. Overall, our study reveals a discrete NAc Tac1 circuit that senses aversive stimuli and drives avoidance behaviors.
Collapse
|
24
|
Anderson EM, Tsvetkov E, Galante A, DeVries D, McCue LM, Wood D, Barry S, Berto S, Lavin A, Taniguchi M, Cowan CW. Epigenetic function during heroin self-administration controls future relapse-associated behavior in a cell type-specific manner. Proc Natl Acad Sci U S A 2023; 120:e2210953120. [PMID: 36745812 PMCID: PMC9963300 DOI: 10.1073/pnas.2210953120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 01/06/2023] [Indexed: 02/08/2023] Open
Abstract
Opioid use produces enduring associations between drug reinforcement/euphoria and discreet or diffuse cues in the drug-taking environment. These powerful associations can trigger relapse in individuals recovering from opioid use disorder (OUD). Here, we sought to determine whether the epigenetic enzyme, histone deacetylase 5 (HDAC5), regulates relapse-associated behavior in an animal model of OUD. We examined the effects of nucleus accumbens (NAc) HDAC5 on both heroin- and sucrose-seeking behaviors using operant self-administration paradigms. We utilized cre-dependent viral-mediated approaches to investigate the cell-type-specific effects of HDAC5 on heroin-seeking behavior, gene expression, and medium spiny neuron (MSN) cell and synaptic physiology. We found that NAc HDAC5 functions during the acquisition phase of heroin self-administration to limit future relapse-associated behavior. Moreover, overexpressing HDAC5 in the NAc suppressed context-associated and reinstated heroin-seeking behaviors, but it did not alter sucrose seeking. We also found that HDAC5 functions within dopamine D1 receptor-expressing MSNs to suppress cue-induced heroin seeking, and within dopamine D2 receptor-expressing MSNs to suppress drug-primed heroin seeking. Assessing cell-type-specific transcriptomics, we found that HDAC5 reduced expression of multiple ion transport genes in both D1- and D2-MSNs. Consistent with this observation, HDAC5 also produced firing rate depression in both MSN classes. These findings revealed roles for HDAC5 during active heroin use in both D1- and D2-MSNs to limit distinct triggers of drug-seeking behavior. Together, our results suggest that HDAC5 might limit relapse vulnerability through regulation of ion channel gene expression and suppression of MSN firing rates during active heroin use.
Collapse
Affiliation(s)
- Ethan M. Anderson
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC29425
| | - Evgeny Tsvetkov
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC29425
| | - Allison Galante
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC29425
| | - Derek DeVries
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC29425
| | - Lauren M. McCue
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC29425
| | - Daniel Wood
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC29425
| | - Sarah Barry
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC29425
| | - Stefano Berto
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC29425
| | - Antonieta Lavin
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC29425
| | - Makoto Taniguchi
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC29425
| | - Christopher W. Cowan
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC29425
| |
Collapse
|
25
|
Psychostimulant Drugs Activate Cell-type Specific and Topographic cFos Expression in the Lumbar Spinal Cord. Neuroscience 2023; 510:9-20. [PMID: 36502959 DOI: 10.1016/j.neuroscience.2022.12.005] [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: 09/11/2022] [Revised: 11/02/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022]
Abstract
Psychostimulant drugs, such as cocaine, d-amphetamine and methylphenidate, alter a wide range of behaviors including locomotor activity and somatosensory perception. These altered behaviors are accompanied by the activation of specific neuronal populations within reward-, emotion- and locomotion-related circuits. However, whether such regulation occurs at the level of the spinal cord, a key node for neural circuits integrating and coordinating sensory and motor functions has never been addressed. By evaluating the temporal and spatial expression pattern of the phosphorylated form of the immediate early gene cFos at Ser32 (pS32-cFos), used as a proxy of neuronal activation, we demonstrate that, in adult male mice, d-amphetamine increases pS32-cFos expression in both inhibitory and excitatory neurons in dorsal and ventral horns at the lumbar spinal cord level. Interestingly, a fraction of neurons activated by a first exposure to d-amphetamine can be re-activated following d-amphetamine re-exposure. Similar expression patterns were observed in response to cocaine and methylphenidate, but not following morphine and dozilcipine administration. Finally, the blockade of dopamine reuptake was sufficient to recapitulate the increase in pS32-cFos expression induced by psychostimulant drugs. Our work provides evidence that cFos expression can be activated in lumbar spinal cord in response to acute psychostimulants administration.
Collapse
|
26
|
Mao LM, Mathur N, Wang JQ. An allosteric potentiator of metabotropic glutamate (mGlu) 2 receptors reduces the cocaine-stimulated ERK1/2 phosphorylation in the mouse striatum. Neurosci Lett 2023; 795:137028. [PMID: 36565803 PMCID: PMC9870709 DOI: 10.1016/j.neulet.2022.137028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/20/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
Metabotropic glutamate (mGlu) receptors are involved in the experience-dependent neuroplasticity in the mesolimbic reward circuit. A Gαi/o-coupled mGlu2 subtype is distributed presynaptically in the striatum. These autoreceptors may have a significant influence over striatal neurons in their intracellular signaling pathways in response to a psychostimulant. Here we explored the effect of pharmacological potentiation of mGlu2 receptors on cocaine-stimulated phosphorylation (activation) of extracellular signal-regulated kinases (ERK) in the mouse striatum in vivo. We found that an mGlu2 selective positive allosteric modulator (PAM) LY487379 after a systemic injection did not alter basal phosphorylation of ERK1/2 or c-Jun N-terminal kinases in the striatum. However, pretreatment with LY487379 blocked the ERK1/2 phosphorylation induced by cocaine in the two subdivisions of the striatum, i.e., the caudate putamen and nucleus accumbens. LY487379 also blocked the cocaine-induced phosphorylation of Elk-1, a transcription factor downstream to the ERK pathway. Additionally, LY487379 reduced locomotor behavioral responses to cocaine. These results demonstrate that the mGlu2 PAM LY487379 possesses the ability to attenuate the activation of the ERK1/2 pathway in striatal neurons and reduce locomotor activity in response to cocaine in vivo.
Collapse
Affiliation(s)
- Li-Min Mao
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - Nirav Mathur
- Department of Anesthesiology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - John Q Wang
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA; Department of Anesthesiology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA.
| |
Collapse
|
27
|
Gerfen CR. Segregation of D1 and D2 dopamine receptors in the striatal direct and indirect pathways: An historical perspective. Front Synaptic Neurosci 2023; 14:1002960. [PMID: 36741471 PMCID: PMC9892636 DOI: 10.3389/fnsyn.2022.1002960] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 12/05/2022] [Indexed: 01/20/2023] Open
Abstract
The direct and indirect striatal pathways form a cornerstone of the circuits of the basal ganglia. Dopamine has opponent affects on the function of these pathways due to the segregation of the D1- and D2-dopamine receptors in the spiny projection neurons giving rise to the direct and indirect pathways. An historical perspective is provided on the discovery of dopamine receptor segregation leading to models of how the direct and indirect affect motor behavior.
Collapse
|
28
|
Phillips RA, Tuscher JJ, Fitzgerald ND, Wan E, Zipperly ME, Duke CG, Ianov L, Day JJ. Distinct subpopulations of D1 medium spiny neurons exhibit unique transcriptional responsiveness to cocaine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.12.523845. [PMID: 36711527 PMCID: PMC9882178 DOI: 10.1101/2023.01.12.523845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Drugs of abuse increase extracellular concentrations of dopamine in the nucleus accumbens (NAc), resulting in transcriptional alterations that drive long-lasting cellular and behavioral adaptations. While decades of research have focused on the transcriptional mechanisms by which drugs of abuse influence neuronal physiology and function, few studies have comprehensively defined NAc cell type heterogeneity in transcriptional responses to drugs of abuse. Here, we used single nucleus RNA-seq (snRNA-seq) to characterize the transcriptome of over 39,000 NAc cells from male and female adult Sprague-Dawley rats following acute or repeated cocaine experience. This dataset identified 16 transcriptionally distinct cell populations, including two populations of medium spiny neurons (MSNs) that express the Drd1 dopamine receptor (D1-MSNs). Critically, while both populations expressed classic marker genes of D1-MSNs, only one population exhibited a robust transcriptional response to cocaine. Validation of population-selective transcripts using RNA in situ hybridization revealed distinct spatial compartmentalization of these D1-MSN populations within the NAc. Finally, analysis of published NAc snRNA-seq datasets from non-human primates and humans demonstrated conservation of MSN subtypes across rat and higher order mammals, and further highlighted cell type-specific transcriptional differences across the NAc and broader striatum. These results highlight the utility in using snRNA-seq to characterize both cell type heterogeneity and cell type-specific responses to cocaine and provides a useful resource for cross-species comparisons of NAc cell composition.
Collapse
Affiliation(s)
- Robert A. Phillips
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jennifer J. Tuscher
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - N. Dalton Fitzgerald
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ethan Wan
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Morgan E. Zipperly
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Corey G. Duke
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lara Ianov
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jeremy J. Day
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
29
|
Nucleus accumbens circuit disinhibits lateral hypothalamus glutamatergic neurons contributing to morphine withdrawal memory in male mice. Nat Commun 2023; 14:71. [PMID: 36604429 PMCID: PMC9814415 DOI: 10.1038/s41467-022-35758-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023] Open
Abstract
The lateral hypothalamus (LH) is physiologically critical in brain functions. The LH also plays an important role in drug addiction. However, neural circuits underlying LH involvement of drug addiction remain obscure. In the present study,our results showed that in male mice, during context-induced expression of morphine withdrawal memory, LH glutamatergic neurons played an important role; dopamine D1 receptor-expressing medium spiny neurons (D1-MSNs) projecting from the core of nucleus accumbens (NAcC) to the LH were an important upstream circuit to activate LH glutamatergic neurons; D1-MSNs projecting from the NAcC to the LH activated LH glutamatergic neurons through inhibiting LH local gamma-aminobutyric acid (GABA) neurons. These results suggest that disinhibited LH glutamatergic neurons by neural circuits from the NAcC importantly contribute to context-induced the expression of morphine withdrawal memory.
Collapse
|
30
|
Pardo M, Gregorio S, Montalban E, Pujadas L, Elias-Tersa A, Masachs N, Vílchez-Acosta A, Parent A, Auladell C, Girault JA, Vila M, Nairn AC, Manso Y, Soriano E. Adult-specific Reelin expression alters striatal neuronal organization: implications for neuropsychiatric disorders. Front Cell Neurosci 2023; 17:1143319. [PMID: 37153634 PMCID: PMC10157100 DOI: 10.3389/fncel.2023.1143319] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/27/2023] [Indexed: 05/10/2023] Open
Abstract
In addition to neuronal migration, brain development, and adult plasticity, the extracellular matrix protein Reelin has been extensively implicated in human psychiatric disorders such as schizophrenia, bipolar disorder, and autism spectrum disorder. Moreover, heterozygous reeler mice exhibit features reminiscent of these disorders, while overexpression of Reelin protects against its manifestation. However, how Reelin influences the structure and circuits of the striatal complex, a key region for the above-mentioned disorders, is far from being understood, especially when altered Reelin expression levels are found at adult stages. In the present study, we took advantage of complementary conditional gain- and loss-of-function mouse models to investigate how Reelin levels may modify adult brain striatal structure and neuronal composition. Using immunohistochemical techniques, we determined that Reelin does not seem to influence the striatal patch and matrix organization (studied by μ-opioid receptor immunohistochemistry) nor the density of medium spiny neurons (MSNs, studied with DARPP-32). We show that overexpression of Reelin leads to increased numbers of striatal parvalbumin- and cholinergic-interneurons, and to a slight increase in tyrosine hydroxylase-positive projections. We conclude that increased Reelin levels might modulate the numbers of striatal interneurons and the density of the nigrostriatal dopaminergic projections, suggesting that these changes may be involved in the protection of Reelin against neuropsychiatric disorders.
Collapse
Affiliation(s)
- Mònica Pardo
- Developmental Neurobiology and Regeneration Laboratory, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Sara Gregorio
- Developmental Neurobiology and Regeneration Laboratory, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Enrica Montalban
- Institut du Fer à Moulin UMR-S 1270, INSERM, Sorbonne University, Paris, France
| | - Lluís Pujadas
- Developmental Neurobiology and Regeneration Laboratory, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Department of Experimental Sciences and Methodology, Faculty of Health Science and Welfare, University of Vic – Central University of Catalonia (UVic-UCC), Vic, Spain
- Tissue Repair and Regeneration Laboratory (TR2Lab), Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central (IRIS-CC), Barcelona, Spain
| | - Alba Elias-Tersa
- Developmental Neurobiology and Regeneration Laboratory, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Núria Masachs
- Developmental Neurobiology and Regeneration Laboratory, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Alba Vílchez-Acosta
- Developmental Neurobiology and Regeneration Laboratory, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Annabelle Parent
- Neurodegenerative Diseases Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
| | - Carme Auladell
- Developmental Neurobiology and Regeneration Laboratory, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Miquel Vila
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Neurodegenerative Diseases Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona (UAB), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
| | - Angus C. Nairn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Yasmina Manso
- Developmental Neurobiology and Regeneration Laboratory, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Yasmina Manso,
| | - Eduardo Soriano
- Developmental Neurobiology and Regeneration Laboratory, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- *Correspondence: Eduardo Soriano,
| |
Collapse
|
31
|
Dwivedi Y, Shelton RC. Genomics in Treatment Development. ADVANCES IN NEUROBIOLOGY 2023; 30:363-385. [PMID: 36928858 DOI: 10.1007/978-3-031-21054-9_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
The Human Genome Project mapped the 3 billion base pairs in the human genome, which ushered in a new generation of genomically focused treatment development. While this has been very successful in other areas, neuroscience has been largely devoid of such developments. This is in large part because there are very few neurological or mental health conditions that are related to single-gene variants. While developments in pharmacogenomics have been somewhat successful, the use of genetic information in practice has to do with drug metabolism and adverse reactions. Studies of drug metabolism related to genetic variations are an important part of drug development. However, outside of cancer biology, the actual translation of genomic information into novel therapies has been limited. Epigenetics, which relates in part to the effects of the environment on DNA, is a promising newer area of relevance to CNS disorders. The environment can induce chemical modifications of DNA (e.g., cytosine methylation), which can be induced by the environment and may represent either shorter- or longer-term changes. Given the importance of environmental influences on CNS disorders, epigenetics may identify important treatment targets in the future.
Collapse
Affiliation(s)
- Yogesh Dwivedi
- Department of Psychiatry and Behavioral Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Richard C Shelton
- Department of Psychiatry and Behavioral Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
32
|
Silvani A, Ghorayeb I, Manconi M, Li Y, Clemens S. Putative Animal Models of Restless Legs Syndrome: A Systematic Review and Evaluation of Their Face and Construct Validity. Neurotherapeutics 2023; 20:154-178. [PMID: 36536233 PMCID: PMC10119375 DOI: 10.1007/s13311-022-01334-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
Restless legs syndrome (RLS) is a sensorimotor disorder that severely affects sleep. It is characterized by an urge to move the legs, which is often accompanied by periodic limb movements during sleep. RLS has a high prevalence in the population and is usually a life-long condition. While its origins remain unclear, RLS is initially highly responsive to treatment with dopaminergic agonists that target D2-like receptors, in particular D2 and D3, but the long-term response is often unsatisfactory. Over the years, several putative animal models for RLS have been developed, mainly based on the epidemiological and neurochemical link with iron deficiency, treatment efficacy of D2-like dopaminergic agonists, or genome-wide association studies that identified risk factors in the patient population. Here, we present the first systematic review of putative animal models of RLS, provide information about their face and construct validity, and report their role in deciphering the underlying pathophysiological mechanisms that may cause or contribute to RLS. We propose that identifying the causal links between genetic risk factors, altered organ functions, and changes to molecular pathways in neural circuitry will eventually lead to more effective new treatment options that bypass the side effects of the currently used therapeutics in RLS, especially for long-term therapy.
Collapse
Affiliation(s)
- Alessandro Silvani
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum - University of Bologna, Ravenna Campus, Ravenna, Italy
| | - Imad Ghorayeb
- Département de Neurophysiologie Clinique, Pôle Neurosciences Cliniques, CHU de Bordeaux, Bordeaux, France
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, UMR 5287, Université de Bordeaux, Bordeaux, France
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, UMR 5287, CNRS, Bordeaux, France
| | - Mauro Manconi
- Sleep Medicine Unit, Neurocenter of Southern Switzerland, EOC, Ospedale Civico, Lugano, Switzerland
- Department of Neurology, University Hospital, Inselspital, Bern, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Yuqing Li
- Department of Neurology, College of Medicine, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Stefan Clemens
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA.
| |
Collapse
|
33
|
Martel AC, Galvan A. Connectivity of the corticostriatal and thalamostriatal systems in normal and parkinsonian states: An update. Neurobiol Dis 2022; 174:105878. [PMID: 36183947 PMCID: PMC9976706 DOI: 10.1016/j.nbd.2022.105878] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 02/06/2023] Open
Abstract
The striatum receives abundant glutamatergic afferents from the cortex and thalamus. These inputs play a major role in the functions of the striatal neurons in normal conditions, and are significantly altered in pathological states, such as Parkinson's disease. This review summarizes the current knowledge of the connectivity of the corticostriatal and thalamostriatal pathways, with emphasis on the most recent advances in the field. We also discuss novel findings regarding structural changes in cortico- and thalamostriatal connections that occur in these connections as a consequence of striatal loss of dopamine in parkinsonism.
Collapse
Affiliation(s)
- Anne-Caroline Martel
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA; Udall Center of Excellence for Parkinson's Disease Research, Emory University, Atlanta, GA, USA
| | - Adriana Galvan
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA; Udall Center of Excellence for Parkinson's Disease Research, Emory University, Atlanta, GA, USA; Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA.
| |
Collapse
|
34
|
Onimus O, Valjent E, Fisone G, Gangarossa G. Haloperidol-Induced Immediate Early Genes in Striatopallidal Neurons Requires the Converging Action of cAMP/PKA/DARPP-32 and mTOR Pathways. Int J Mol Sci 2022; 23:ijms231911637. [PMID: 36232936 PMCID: PMC9569967 DOI: 10.3390/ijms231911637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 09/21/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022] Open
Abstract
Antipsychotics share the common pharmacological feature of antagonizing the dopamine 2 receptor (D2R), which is abundant in the striatum and involved in both the therapeutic and side effects of this drug’s class. The pharmacological blockade of striatal D2R, by disinhibiting the D2R-containing medium-sized spiny neurons (MSNs), leads to a plethora of molecular, cellular and behavioral adaptations, which are central in the action of antipsychotics. Here, we focused on the cell type-specific (D2R-MSNs) regulation of some striatal immediate early genes (IEGs), such as cFos, Arc and Zif268. Taking advantage of transgenic mouse models, pharmacological approaches and immunofluorescence analyses, we found that haloperidol-induced IEGs in the striatum required the synergistic activation of A2a (adenosine) and NMDA (glutamate) receptors. At the intracellular signaling level, we found that the PKA/DARPP-32 and mTOR pathways synergistically cooperate to control the induction of IEGs by haloperidol. By confirming and further expanding previous observations, our results provide novel insights into the regulatory mechanisms underlying the molecular/cellular action of antipsychotics in the striatum.
Collapse
Affiliation(s)
- Oriane Onimus
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France
| | - Emmanuel Valjent
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, Inserm, 34094 Montpellier, France
| | - Gilberto Fisone
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Giuseppe Gangarossa
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France
- Correspondence:
| |
Collapse
|
35
|
Morella I, Pohořalá V, Calpe-López C, Brambilla R, Spanagel R, Bernardi RE. Nicotine self-administration and ERK signaling are altered in RasGRF2 knockout mice. Front Pharmacol 2022; 13:986566. [PMID: 36120353 PMCID: PMC9479000 DOI: 10.3389/fphar.2022.986566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/10/2022] [Indexed: 11/29/2022] Open
Abstract
Ras/Raf/MEK/ERK (Ras-ERK) signaling has been demonstrated to play a role in the effects of drugs of abuse such as cocaine and alcohol, but has not been extensively examined in nicotine-related reward behaviors. We examined the role of Ras Guanine Nucleotide Releasing Factor 2 (RasGRF2), an upstream mediator of the Ras-ERK signaling pathway, on nicotine self-administration (SA) in RasGRF2 KO and WT mice. We first demonstrated that acute nicotine exposure (0.4 mg/kg) resulted in an increase in phosphorylated ERK1/2 (pERK1/2) in the striatum, consistent with previous reports. We also demonstrated that increases in pERK1/2 resulting from acute (0.4 mg/kg) and repeated (0.4 mg/kg, 10 daily injections) exposure to nicotine in WT mice were not present in RasGRF2 KO mice, confirming that RasGRF2 at least partly regulates the activity of the Ras-ERK signaling pathway following nicotine exposure. We then performed intravenous nicotine SA (0.03 mg/kg/infusion for 10 days) in RasGRF2 KO and WT mice. Consistent with a previous report using cocaine SA, RasGRF2 KO mice demonstrated an increase in nicotine SA relative to WT controls. These findings suggest a role for RasGRF2 in the reinforcing effects of nicotine, and implicate the Ras-ERK signaling pathway as a common mediator of the response to drugs of abuse.
Collapse
Affiliation(s)
- Ilaria Morella
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, United Kingdom
- Division of Neuroscience, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Veronika Pohořalá
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Claudia Calpe-López
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Riccardo Brambilla
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, United Kingdom
- Division of Neuroscience, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Rick E. Bernardi
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
36
|
Shang Z, Yang L, Wang Z, Tian Y, Gao Y, Su Z, Guo R, Li W, Liu G, Li X, Yang Z, Li Z, Zhang Z. The transcription factor Zfp503 promotes the D1 MSN identity and represses the D2 MSN identity. Front Cell Dev Biol 2022; 10:948331. [PMID: 36081908 PMCID: PMC9445169 DOI: 10.3389/fcell.2022.948331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/19/2022] [Indexed: 11/23/2022] Open
Abstract
The striatum is primarily composed of two types of medium spiny neurons (MSNs) expressing either D1- or D2-type dopamine receptors. However, the fate determination of these two types of neurons is not fully understood. Here, we found that D1 MSNs undergo fate switching to D2 MSNs in the absence of Zfp503. Furthermore, scRNA-seq revealed that the transcription factor Zfp503 affects the differentiation of these progenitor cells in the lateral ganglionic eminence (LGE). More importantly, we found that the transcription factors Sp8/9, which are required for the differentiation of D2 MSNs, are repressed by Zfp503. Finally, sustained Zfp503 expression in LGE progenitor cells promoted the D1 MSN identity and repressed the D2 MSN identity. Overall, our findings indicated that Zfp503 promotes the D1 MSN identity and represses the D2 MSN identity by regulating Sp8/9 expression during striatal MSN development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Zhenmeiyu Li
- *Correspondence: Zhenmeiyu Li, ; Zhuangzhi Zhang,
| | | |
Collapse
|
37
|
Simmler LD, Li Y, Hadjas LC, Hiver A, van Zessen R, Lüscher C. Dual action of ketamine confines addiction liability. Nature 2022; 608:368-373. [PMID: 35896744 DOI: 10.1038/s41586-022-04993-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 06/17/2022] [Indexed: 12/19/2022]
Abstract
Ketamine is used clinically as an anaesthetic and a fast-acting antidepressant, and recreationally for its dissociative properties, raising concerns of addiction as a possible side effect. Addictive drugs such as cocaine increase the levels of dopamine in the nucleus accumbens. This facilitates synaptic plasticity in the mesolimbic system, which causes behavioural adaptations and eventually drives the transition to compulsion1-4. The addiction liability of ketamine is a matter of much debate, in part because of its complex pharmacology that among several targets includes N-methyl-D-aspartic acid (NMDA) receptor (NMDAR) antagonism5,6. Here we show that ketamine does not induce the synaptic plasticity that is typically observed with addictive drugs in mice, despite eliciting robust dopamine transients in the nucleus accumbens. Ketamine nevertheless supported reinforcement through the disinhibition of dopamine neurons in the ventral tegmental area (VTA). This effect was mediated by NMDAR antagonism in GABA (γ-aminobutyric acid) neurons of the VTA, but was quickly terminated by type-2 dopamine receptors on dopamine neurons. The rapid off-kinetics of the dopamine transients along with the NMDAR antagonism precluded the induction of synaptic plasticity in the VTA and the nucleus accumbens, and did not elicit locomotor sensitization or uncontrolled self-administration. In summary, the dual action of ketamine leads to a unique constellation of dopamine-driven positive reinforcement, but low addiction liability.
Collapse
Affiliation(s)
- Linda D Simmler
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Yue Li
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Lotfi C Hadjas
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Agnès Hiver
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Ruud van Zessen
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Christian Lüscher
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland. .,Service de Neurologie, Department of Clinical Neurosciences, Geneva University Hospital, Geneva, Switzerland.
| |
Collapse
|
38
|
Yuen J, Kouzani AZ, Berk M, Tye SJ, Rusheen AE, Blaha CD, Bennet KE, Lee KH, Shin H, Kim JH, Oh Y. Deep Brain Stimulation for Addictive Disorders-Where Are We Now? Neurotherapeutics 2022; 19:1193-1215. [PMID: 35411483 PMCID: PMC9587163 DOI: 10.1007/s13311-022-01229-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2022] [Indexed: 10/18/2022] Open
Abstract
In the face of a global epidemic of drug addiction, neglecting to develop new effective therapies will perpetuate the staggering human and economic costs of substance use. This review aims to summarize and evaluate the preclinical and clinical studies of deep brain stimulation (DBS) as a novel therapy for refractory addiction, in hopes to engage and inform future research in this promising novel treatment avenue. An electronic database search (MEDLINE, EMBASE, Cochrane library) was performed using keywords and predefined inclusion criteria between 1974 and 6/18/2021 (registered on Open Science Registry). Selected articles were reviewed in full text and key details were summarized and analyzed to understand DBS' therapeutic potential and possible mechanisms of action. The search yielded 25 animal and 22 human studies. Animal studies showed that DBS of targets such as nucleus accumbens (NAc), insula, and subthalamic nucleus reduces drug use and seeking. All human studies were case series/reports (level 4/5 evidence), mostly targeting the NAc with generally positive outcomes. From the limited evidence in the literature, DBS, particularly of the NAc, appears to be a reasonable last resort option for refractory addictive disorders. We propose that future research in objective electrophysiological (e.g., local field potentials) and neurochemical (e.g., extracellular dopamine levels) biomarkers would assist monitoring the progress of treatment and developing a closed-loop DBS system. Preclinical literature also highlighted the prefrontal cortex as a promising DBS target, which should be explored in human research.
Collapse
Affiliation(s)
- Jason Yuen
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong VIC 3216, Australia
| | - Abbas Z Kouzani
- School of Engineering, Deakin University, Geelong VIC 3216, Australia
| | - Michael Berk
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong VIC 3216, Australia
| | - Susannah J Tye
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Psychiatry, University of Minnesota, Minneapolis, MN, 55455, USA
- Department of Psychiatry, Emory University, Atlanta, GA, 30322, USA
| | - Aaron E Rusheen
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Charles D Blaha
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Kevin E Bennet
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
- Division of Engineering, Mayo Clinic, Rochester, MN, 55905, USA
| | - Kendall H Lee
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA
| | - Hojin Shin
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jee Hyun Kim
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong VIC 3216, Australia.
| | - Yoonbae Oh
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
39
|
Li Z, Shang Z, Sun M, Jiang X, Tian Y, Yang L, Wang Z, Su Z, Liu G, Li X, You Y, Yang Z, Xu Z, Zhang Z. Transcription factor Sp9 is a negative regulator of D1-type MSN development. Cell Death Dis 2022; 8:301. [PMID: 35773249 PMCID: PMC9247084 DOI: 10.1038/s41420-022-01088-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 11/09/2022]
Abstract
The striatum is the main input structure of the basal ganglia, receiving information from the cortex and the thalamus and consisting of D1- and D2- medium spiny neurons (MSNs). D1-MSNs and D2-MSNs are essential for motor control and cognitive behaviors and have implications in Parkinson’s Disease. In the present study, we demonstrated that Sp9-positive progenitors produced both D1-MSNs and D2-MSNs and that Sp9 expression was rapidly downregulated in postmitotic D1-MSNs. Furthermore, we found that sustained Sp9 expression in lateral ganglionic eminence (LGE) progenitor cells and their descendants led to promoting D2-MSN identity and repressing D1-MSN identity during striatal development. As a result, sustained Sp9 expression resulted in an imbalance between D1-MSNs and D2-MSNs in the mouse striatum. In addition, the fate-changed D2-like MSNs survived normally in adulthood. Taken together, our findings supported that Sp9 was sufficient to promote D2-MSN identity and repress D1-MSN identity, and Sp9 was a negative regulator of D1-MSN fate.
Collapse
Affiliation(s)
- Zhenmeiyu Li
- Institute of Pediatrics, Children's Hospital of Fudan University, state Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 200032, Shanghai, China
| | - Zicong Shang
- Institute of Pediatrics, Children's Hospital of Fudan University, state Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 200032, Shanghai, China
| | - Mengge Sun
- Institute of Pediatrics, Children's Hospital of Fudan University, state Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 200032, Shanghai, China
| | - Xin Jiang
- Institute of Pediatrics, Children's Hospital of Fudan University, state Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 200032, Shanghai, China
| | - Yu Tian
- Institute of Pediatrics, Children's Hospital of Fudan University, state Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 200032, Shanghai, China
| | - Lin Yang
- Institute of Pediatrics, Children's Hospital of Fudan University, state Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 200032, Shanghai, China
| | - Ziwu Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, state Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 200032, Shanghai, China
| | - Zihao Su
- Institute of Pediatrics, Children's Hospital of Fudan University, state Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 200032, Shanghai, China
| | - Guoping Liu
- Institute of Pediatrics, Children's Hospital of Fudan University, state Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 200032, Shanghai, China
| | - Xiaosu Li
- Institute of Pediatrics, Children's Hospital of Fudan University, state Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 200032, Shanghai, China
| | - Yan You
- Institute of Pediatrics, Children's Hospital of Fudan University, state Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 200032, Shanghai, China
| | - Zhengang Yang
- Institute of Pediatrics, Children's Hospital of Fudan University, state Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 200032, Shanghai, China
| | - Zhejun Xu
- Institute of Pediatrics, Children's Hospital of Fudan University, state Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 200032, Shanghai, China.
| | - Zhuangzhi Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University, state Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 200032, Shanghai, China.
| |
Collapse
|
40
|
Anderson EM, Taniguchi M. Epigenetic Effects of Addictive Drugs in the Nucleus Accumbens. Front Mol Neurosci 2022; 15:828055. [PMID: 35813068 PMCID: PMC9260254 DOI: 10.3389/fnmol.2022.828055] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 05/30/2022] [Indexed: 12/28/2022] Open
Abstract
Substance use induces long-lasting behavioral changes and drug craving. Increasing evidence suggests that epigenetic gene regulation contributes to the development and expression of these long-lasting behavioral alterations. Here we systematically review extensive evidence from rodent models of drug-induced changes in epigenetic regulation and epigenetic regulator proteins. We focus on histone acetylation and histone methylation in a brain region important for drug-related behaviors: the nucleus accumbens. We also discuss how experimentally altering these epigenetic regulators via systemically administered compounds or nucleus accumbens-specific manipulations demonstrate the importance of these proteins in the behavioral effects of drugs and suggest potential therapeutic value to treat people with substance use disorder. Finally, we discuss limitations and future directions for the field of epigenetic studies in the behavioral effects of addictive drugs and suggest how to use these insights to develop efficacious treatments.
Collapse
|
41
|
Burke DA, Alvarez VA. Serotonin receptors contribute to dopamine depression of lateral inhibition in the nucleus accumbens. Cell Rep 2022; 39:110795. [PMID: 35545050 PMCID: PMC9171783 DOI: 10.1016/j.celrep.2022.110795] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 01/09/2022] [Accepted: 04/15/2022] [Indexed: 11/30/2022] Open
Abstract
Dopamine modulation of nucleus accumbens (NAc) circuitry is central to theories of reward seeking and reinforcement learning. Despite decades of effort, the acute dopamine actions on the NAc microcircuitry remain puzzling. Here, we dissect out the direct actions of dopamine on lateral inhibition between medium spiny neurons (MSNs) in mouse brain slices and find that they are pathway specific. Dopamine potently depresses GABAergic transmission from presynaptic dopamine D2 receptor-expressing MSNs (D2-MSNs), whereas it potentiates transmission from presynaptic dopamine D1 receptor-expressing MSNs (D1-MSNs) onto other D1-MSNs. To our surprise, presynaptic D2 receptors mediate only half of the depression induced by endogenous and exogenous dopamine. Presynaptic serotonin 5-HT1B receptors are responsible for a significant component of dopamine-induced synaptic depression. This study clarifies the mechanistic understanding of dopamine actions in the NAc by showing pathway-specific modulation of lateral inhibition and involvement of D2 and 5-HT1B receptors in dopamine depression of D2-MSN synapses.
Collapse
Affiliation(s)
- Dennis A Burke
- Laboratory on Neurobiology of Compulsive Behaviors, NIAAA, National Institutes of Health, Bethesda, MD 20892, USA; Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Veronica A Alvarez
- Laboratory on Neurobiology of Compulsive Behaviors, NIAAA, National Institutes of Health, Bethesda, MD 20892, USA; Intramural Research Program, NIDA, NIH, Baltimore, MD 21224, USA; Center on Compulsive Behaviors, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
42
|
Montalban E, Giralt A, Taing L, Schut EHS, Supiot LF, Castell L, Nakamura Y, de Pins B, Pelosi A, Goutebroze L, Tuduri P, Wang W, Neiburga KD, Vestito L, Castel J, Luquet S, Nairn AC, Hervé D, Heintz N, Martin C, Greengard P, Valjent E, Meye FJ, Gambardella N, Roussarie JP, Girault JA. Translational profiling of mouse dopaminoceptive neurons reveals region-specific gene expression, exon usage, and striatal prostaglandin E2 modulatory effects. Mol Psychiatry 2022; 27:2068-2079. [PMID: 35177825 PMCID: PMC10009708 DOI: 10.1038/s41380-022-01439-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 12/16/2021] [Accepted: 01/05/2022] [Indexed: 01/11/2023]
Abstract
Forebrain dopamine-sensitive (dopaminoceptive) neurons play a key role in movement, action selection, motivation, and working memory. Their activity is altered in Parkinson's disease, addiction, schizophrenia, and other conditions, and drugs that stimulate or antagonize dopamine receptors have major therapeutic applications. Yet, similarities and differences between the various neuronal populations sensitive to dopamine have not been systematically explored. To characterize them, we compared translating mRNAs in the dorsal striatum and nucleus accumbens neurons expressing D1 or D2 dopamine receptor and prefrontal cortex neurons expressing D1 receptor. We identified genome-wide cortico-striatal, striatal D1/D2 and dorso/ventral differences in the translating mRNA and isoform landscapes, which characterize dopaminoceptive neuronal populations. Expression patterns and network analyses identified novel transcription factors with presumptive roles in these differences. Prostaglandin E2 (PGE2) was a candidate upstream regulator in the dorsal striatum. We pharmacologically explored this hypothesis and showed that misoprostol, a PGE2 receptor agonist, decreased the excitability of D2 striatal projection neurons in slices, and diminished their activity in vivo during novel environment exploration. We found that misoprostol also modulates mouse behavior including by facilitating reversal learning. Our study provides powerful resources for characterizing dopamine target neurons, new information about striatal gene expression patterns and regulation. It also reveals the unforeseen role of PGE2 in the striatum as a potential neuromodulator and an attractive therapeutic target.
Collapse
Affiliation(s)
- Enrica Montalban
- Inserm UMR-S 1270, Paris, France.,Faculty of Sciences and Engineering, Sorbonne Université, Paris, France.,Institut du Fer à Moulin, Paris, France.,Université de Paris, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Albert Giralt
- Inserm UMR-S 1270, Paris, France.,Faculty of Sciences and Engineering, Sorbonne Université, Paris, France.,Institut du Fer à Moulin, Paris, France.,Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain.,Production and Validation Center of Advanced Therapies (Creatio), University of Barcelona, Barcelona, Spain
| | - Lieng Taing
- Inserm UMR-S 1270, Paris, France.,Faculty of Sciences and Engineering, Sorbonne Université, Paris, France.,Institut du Fer à Moulin, Paris, France.,UMR1166, Faculté de Médecine, Sorbonne University, Paris, France
| | - Evelien H S Schut
- Department of Translational Neuroscience, Brain Center, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Laura F Supiot
- Department of Translational Neuroscience, Brain Center, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Laia Castell
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France.,Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Yuki Nakamura
- Inserm UMR-S 1270, Paris, France.,Faculty of Sciences and Engineering, Sorbonne Université, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Benoit de Pins
- Inserm UMR-S 1270, Paris, France.,Faculty of Sciences and Engineering, Sorbonne Université, Paris, France.,Institut du Fer à Moulin, Paris, France.,Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Assunta Pelosi
- Inserm UMR-S 1270, Paris, France.,Faculty of Sciences and Engineering, Sorbonne Université, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Laurence Goutebroze
- Inserm UMR-S 1270, Paris, France.,Faculty of Sciences and Engineering, Sorbonne Université, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Pola Tuduri
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Wei Wang
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, USA.,Bioinformatics Resource Center, Rockefeller University, New York, NY, USA
| | - Katrina Daila Neiburga
- Babraham Institute, Cambridge, UK.,Bioinformatics Lab, Riga Stradins University, Riga, Latvia
| | - Letizia Vestito
- Babraham Institute, Cambridge, UK.,University College London, London, UK
| | - Julien Castel
- Université de Paris, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Serge Luquet
- Université de Paris, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Angus C Nairn
- Department of Psychiatry, Yale School of Medicine, Connecticut Mental Health Center, New Haven, CT, USA
| | - Denis Hervé
- Inserm UMR-S 1270, Paris, France.,Faculty of Sciences and Engineering, Sorbonne Université, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Nathaniel Heintz
- Laboratory of Molecular Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Claire Martin
- Université de Paris, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Paul Greengard
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, USA
| | - Emmanuel Valjent
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Frank J Meye
- Department of Translational Neuroscience, Brain Center, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Jean-Pierre Roussarie
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, USA. .,Boston University School of Medicine, Department of Anatomy & Neurobiology, Boston, MA, USA.
| | - Jean-Antoine Girault
- Inserm UMR-S 1270, Paris, France. .,Faculty of Sciences and Engineering, Sorbonne Université, Paris, France. .,Institut du Fer à Moulin, Paris, France.
| |
Collapse
|
43
|
Berland C, Castel J, Terrasi R, Montalban E, Foppen E, Martin C, Muccioli GG, Luquet S, Gangarossa G. Identification of an endocannabinoid gut-brain vagal mechanism controlling food reward and energy homeostasis. Mol Psychiatry 2022; 27:2340-2354. [PMID: 35075269 DOI: 10.1038/s41380-021-01428-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 12/07/2021] [Accepted: 12/23/2021] [Indexed: 12/11/2022]
Abstract
The regulation of food intake, a sine qua non requirement for survival, thoroughly shapes feeding and energy balance by integrating both homeostatic and hedonic values of food. Unfortunately, the widespread access to palatable food has led to the development of feeding habits that are independent from metabolic needs. Among these, binge eating (BE) is characterized by uncontrolled voracious eating. While reward deficit seems to be a major contributor of BE, the physiological and molecular underpinnings of BE establishment remain elusive. Here, we combined a physiologically relevant BE mouse model with multiscale in vivo approaches to explore the functional connection between the gut-brain axis and the reward and homeostatic brain structures. Our results show that BE elicits compensatory adaptations requiring the gut-to-brain axis which, through the vagus nerve, relies on the permissive actions of peripheral endocannabinoids (eCBs) signaling. Selective inhibition of peripheral CB1 receptors resulted in a vagus-dependent increased hypothalamic activity, modified metabolic efficiency, and dampened activity of mesolimbic dopamine circuit, altogether leading to the suppression of palatable eating. We provide compelling evidence for a yet unappreciated physiological integrative mechanism by which variations of peripheral eCBs control the activity of the vagus nerve, thereby in turn gating the additive responses of both homeostatic and hedonic brain circuits which govern homeostatic and reward-driven feeding. In conclusion, we reveal that vagus-mediated eCBs/CB1R functions represent an interesting and innovative target to modulate energy balance and counteract food-reward disorders.
Collapse
Affiliation(s)
- Chloé Berland
- Université de Paris, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France
| | - Julien Castel
- Université de Paris, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France
| | - Romano Terrasi
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, 1200, Brussels, Belgium
| | - Enrica Montalban
- Université de Paris, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France
| | - Ewout Foppen
- Université de Paris, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France
| | - Claire Martin
- Université de Paris, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, 1200, Brussels, Belgium
| | - Serge Luquet
- Université de Paris, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France
| | - Giuseppe Gangarossa
- Université de Paris, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France.
| |
Collapse
|
44
|
Ramírez-Jarquín UN, Sharma M, Shahani N, Li Y, Boregowda S, Subramaniam S. Rhes protein transits from neuron to neuron and facilitates mutant huntingtin spreading in the brain. SCIENCE ADVANCES 2022; 8:eabm3877. [PMID: 35319973 PMCID: PMC8942366 DOI: 10.1126/sciadv.abm3877] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 02/02/2022] [Indexed: 05/12/2023]
Abstract
Rhes (RASD2) is a thyroid hormone-induced gene that regulates striatal motor activity and promotes neurodegeneration in Huntington disease (HD) and tauopathy. Rhes moves and transports the HD protein, polyglutamine-expanded huntingtin (mHTT), via tunneling nanotube (TNT)-like membranous protrusions between cultured neurons. However, similar intercellular Rhes transportation in the intact brain was unknown. Here, we report that Rhes induces TNT-like protrusions in the striatal medium spiny neurons (MSNs) and transported between dopamine-1 receptor (D1R)-MSNs and D2R-MSNs of intact striatum and organotypic brain slices. Notably, mHTT is robustly transported within the striatum and from the striatum to the cortical areas in the brain, and Rhes deletion diminishes such transport. Moreover, Rhes moves to the cortical regions following restricted expression in the MSNs of the striatum. Thus, Rhes is a first striatum-enriched protein demonstrated to move and transport mHTT between neurons and brain regions, providing new insights into interneuronal protein transport in the brain.
Collapse
Affiliation(s)
| | - Manish Sharma
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Neelam Shahani
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Yuqing Li
- Department of Neurology, University of Florida, Gainesville, FL 32610, USA
| | - Siddaraju Boregowda
- Department of Molecular Medicine, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Srinivasa Subramaniam
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| |
Collapse
|
45
|
Kim H, Nam MH, Jeong S, Lee H, Oh SJ, Kim J, Choi N, Seong J. Visualization of differential GPCR crosstalk in DRD1-DRD2 heterodimer upon different dopamine levels. Prog Neurobiol 2022; 213:102266. [DOI: 10.1016/j.pneurobio.2022.102266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/13/2021] [Accepted: 03/24/2022] [Indexed: 12/25/2022]
|
46
|
Allichon MC, Ortiz V, Pousinha P, Andrianarivelo A, Petitbon A, Heck N, Trifilieff P, Barik J, Vanhoutte P. Cell-Type-Specific Adaptions in Striatal Medium-Sized Spiny Neurons and Their Roles in Behavioral Responses to Drugs of Abuse. Front Synaptic Neurosci 2022; 13:799274. [PMID: 34970134 PMCID: PMC8712310 DOI: 10.3389/fnsyn.2021.799274] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/26/2021] [Indexed: 12/21/2022] Open
Abstract
Drug addiction is defined as a compulsive pattern of drug-seeking- and taking- behavior, with recurrent episodes of abstinence and relapse, and a loss of control despite negative consequences. Addictive drugs promote reinforcement by increasing dopamine in the mesocorticolimbic system, which alters excitatory glutamate transmission within the reward circuitry, thereby hijacking reward processing. Within the reward circuitry, the striatum is a key target structure of drugs of abuse since it is at the crossroad of converging glutamate inputs from limbic, thalamic and cortical regions, encoding components of drug-associated stimuli and environment, and dopamine that mediates reward prediction error and incentive values. These signals are integrated by medium-sized spiny neurons (MSN), which receive glutamate and dopamine axons converging onto their dendritic spines. MSN primarily form two mostly distinct populations based on the expression of either DA-D1 (D1R) or DA-D2 (D2R) receptors. While a classical view is that the two MSN populations act in parallel, playing antagonistic functional roles, the picture seems much more complex. Herein, we review recent studies, based on the use of cell-type-specific manipulations, demonstrating that dopamine differentially modulates dendritic spine density and synapse formation, as well as glutamate transmission, at specific inputs projecting onto D1R-MSN and D2R-MSN to shape persistent pathological behavioral in response to drugs of abuse. We also discuss the identification of distinct molecular events underlying the detrimental interplay between dopamine and glutamate signaling in D1R-MSN and D2R-MSN and highlight the relevance of such cell-type-specific molecular studies for the development of innovative strategies with potential therapeutic value for addiction. Because drug addiction is highly prevalent in patients with other psychiatric disorders when compared to the general population, we last discuss the hypothesis that shared cellular and molecular adaptations within common circuits could explain the co-occurrence of addiction and depression. We will therefore conclude this review by examining how the nucleus accumbens (NAc) could constitute a key interface between addiction and depression.
Collapse
Affiliation(s)
- Marie-Charlotte Allichon
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Vanesa Ortiz
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Paula Pousinha
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Andry Andrianarivelo
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Anna Petitbon
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Nicolas Heck
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Pierre Trifilieff
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Jacques Barik
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Peter Vanhoutte
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| |
Collapse
|
47
|
Jones-Tabah J, Martin RD, Chen JJ, Tanny JC, Clarke PBS, Hébert TE. A role for BET proteins in regulating basal, dopamine-induced and cAMP/PKA-dependent transcription in rat striatal neurons. Cell Signal 2021; 91:110226. [PMID: 34974082 DOI: 10.1016/j.cellsig.2021.110226] [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: 10/28/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 01/01/2023]
Abstract
The activity of striatal medium-spiny projection neurons is regulated by D1 and D2 dopamine receptors. The D1 receptor (D1R) is a Gαs/olf-coupled GPCR which activates a cAMP/PKA/DARPP-32 signalling cascade that increases excitability and facilitates plasticity, partly through the regulation of transcription. Upon activation via D1R, PKA can translocate to the nucleus to regulate transcription through the phosphorylation of various targets. One candidate effector of PKA-dependent transcriptional regulation is the BET protein Brd4. It is known that when Brd4 is activated by phosphorylation, it binds more readily to acetylated histones at promoters and enhancers; moreover, in non-neuronal cells, PKA signalling has been shown to increase recruitment of Brd4 to chromatin. However, it is unknown whether BET proteins, or Brd4 specifically, are involved in transcriptional activation by cAMP/PKA in neurons. Here, we demonstrate that in adult rats, inhibition of BET proteins with the bromodomain inhibitor JQ1 suppressed the expression of ~25% of D1R-upregulated genes, while also increasing the expression of a subset of immediate-early genes. We further found that cAMP/PKA signalling promotes Brd4 recruitment to dopamine-induced genes in striatal neurons, and that knockdown of Brd4 attenuates D1R-induced gene expression. Finally, we report that JQ1 treatment downregulated expression of many GPCRs and also impaired ERK1/2 signalling in striatal neurons. Our findings identify the BET protein family, and Brd4 in particular, as novel regulators of basal and D1R-dependent transcription in rat striatal neurons, and delineate complex bi-directional effects of bromodomain inhibitors on neuronal transcription.
Collapse
Affiliation(s)
- Jace Jones-Tabah
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Ryan D Martin
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Jennifer J Chen
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Jason C Tanny
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Paul B S Clarke
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| |
Collapse
|
48
|
Andrianarivelo A, Saint-Jour E, Pousinha P, Fernandez SP, Petitbon A, De Smedt-Peyrusse V, Heck N, Ortiz V, Allichon MC, Kappès V, Betuing S, Walle R, Zhu Y, Joséphine C, Bemelmans AP, Turecki G, Mechawar N, Javitch JA, Caboche J, Trifilieff P, Barik J, Vanhoutte P. Disrupting D1-NMDA or D2-NMDA receptor heteromerization prevents cocaine's rewarding effects but preserves natural reward processing. SCIENCE ADVANCES 2021; 7:eabg5970. [PMID: 34669474 PMCID: PMC8528421 DOI: 10.1126/sciadv.abg5970] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Addictive drugs increase dopamine in the nucleus accumbens (NAc), where it persistently shapes excitatory glutamate transmission and hijacks natural reward processing. Here, we provide evidence, from mice to humans, that an underlying mechanism relies on drug-evoked heteromerization of glutamate N-methyl-d-aspartate receptors (NMDAR) with dopamine receptor 1 (D1R) or 2 (D2R). Using temporally controlled inhibition of D1R-NMDAR heteromerization, we unraveled their selective implication in early phases of cocaine-mediated synaptic, morphological, and behavioral responses. In contrast, preventing D2R-NMDAR heteromerization blocked the persistence of these adaptations. Interfering with these heteromers spared natural reward processing. Notably, we established that D2R-NMDAR complexes exist in human samples and showed that, despite a decreased D2R protein expression in the NAc, individuals with psychostimulant use disorder display a higher proportion of D2R forming heteromers with NMDAR. These findings contribute to a better understanding of molecular mechanisms underlying addiction and uncover D2R-NMDAR heteromers as targets with potential therapeutic value.
Collapse
Affiliation(s)
- Andry Andrianarivelo
- CNRS, UMR 8246, Neuroscience Paris Seine, F-75005 Paris, France
- INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, F-75005 Paris, France
- Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, F-75005 Paris, France
| | - Estefani Saint-Jour
- CNRS, UMR 8246, Neuroscience Paris Seine, F-75005 Paris, France
- INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, F-75005 Paris, France
- Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, F-75005 Paris, France
| | - Paula Pousinha
- Université Côte d’Azur, Nice, France
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR7275, Valbonne, France
| | - Sebastian P. Fernandez
- Université Côte d’Azur, Nice, France
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR7275, Valbonne, France
| | - Anna Petitbon
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, 33000 Bordeaux, France
| | | | - Nicolas Heck
- CNRS, UMR 8246, Neuroscience Paris Seine, F-75005 Paris, France
- INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, F-75005 Paris, France
- Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, F-75005 Paris, France
| | - Vanesa Ortiz
- Université Côte d’Azur, Nice, France
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR7275, Valbonne, France
| | - Marie-Charlotte Allichon
- CNRS, UMR 8246, Neuroscience Paris Seine, F-75005 Paris, France
- INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, F-75005 Paris, France
- Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, F-75005 Paris, France
| | - Vincent Kappès
- CNRS, UMR 8246, Neuroscience Paris Seine, F-75005 Paris, France
- INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, F-75005 Paris, France
- Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, F-75005 Paris, France
| | - Sandrine Betuing
- CNRS, UMR 8246, Neuroscience Paris Seine, F-75005 Paris, France
- INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, F-75005 Paris, France
- Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, F-75005 Paris, France
| | - Roman Walle
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, 33000 Bordeaux, France
| | - Ying Zhu
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
| | - Charlène Joséphine
- Commissariat à l’Énergie Atomique et aux Énergies Alternatives (CEA), Département de la Recherche Fondamentale, Institut de biologie François Jacob, MIRCen, and CNRS UMR 9199, Université Paris-Sud, Université Paris-Saclay, Neurodegenerative Diseases Laboratory, Fontenay-aux-Roses, France
| | - Alexis-Pierre Bemelmans
- Commissariat à l’Énergie Atomique et aux Énergies Alternatives (CEA), Département de la Recherche Fondamentale, Institut de biologie François Jacob, MIRCen, and CNRS UMR 9199, Université Paris-Sud, Université Paris-Saclay, Neurodegenerative Diseases Laboratory, Fontenay-aux-Roses, France
| | - Gustavo Turecki
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Naguib Mechawar
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Jonathan A. Javitch
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
- Department of Pharmacology, Columbia University, New York, NY 10032, USA
| | - Jocelyne Caboche
- CNRS, UMR 8246, Neuroscience Paris Seine, F-75005 Paris, France
- INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, F-75005 Paris, France
- Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, F-75005 Paris, France
| | - Pierre Trifilieff
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, 33000 Bordeaux, France
| | - Jacques Barik
- Université Côte d’Azur, Nice, France
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR7275, Valbonne, France
| | - Peter Vanhoutte
- CNRS, UMR 8246, Neuroscience Paris Seine, F-75005 Paris, France
- INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, F-75005 Paris, France
- Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, F-75005 Paris, France
- Corresponding author.
| |
Collapse
|
49
|
van Zessen R, Li Y, Marion-Poll L, Hulo N, Flakowski J, Lüscher C. Dynamic dichotomy of accumbal population activity underlies cocaine sensitization. eLife 2021; 10:e66048. [PMID: 34608866 PMCID: PMC8523149 DOI: 10.7554/elife.66048] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 09/28/2021] [Indexed: 11/28/2022] Open
Abstract
Locomotor sensitization (LS) is an early behavioral adaptation to addictive drugs, driven by the increase of dopamine in the Nucleus Accumbens (NAc). However, the effect on accumbal population activity remains elusive. Here, we used single-cell calcium imaging in mice to record the activity of dopamine-1-receptor (D1R) and dopamine-2-receptor (D2R) expressing spiny projection neurons (SPNs) during cocaine LS. Acute exposure to cocaine elevated D1R SPN activity and reduced D2R SPN activity, albeit with high variability between neurons. During LS, the number of D1R and D2R neurons responding in opposite directions increased. Moreover, preventing LS by inhibition of the ERK signaling pathway decreased the number of cocaine responsive D1R SPNs, but had little effect on D2R SPNs. These results indicate that accumbal population dichotomy is dynamic and contains a subgroup of D1R SPNs that eventually drives LS. Insights into the drug-related activity dynamics provides a foundation for understanding the circuit-level addiction pathogenesis.
Collapse
Affiliation(s)
- Ruud van Zessen
- Department of Basic Neurosciences, Faculty of Medicine, University of GenevaGenevaSwitzerland
| | - Yue Li
- Department of Basic Neurosciences, Faculty of Medicine, University of GenevaGenevaSwitzerland
| | - Lucile Marion-Poll
- Department of Basic Neurosciences, Faculty of Medicine, University of GenevaGenevaSwitzerland
| | - Nicolas Hulo
- Institute of Genetics and Genomics of Geneva (IGE3), University of GenevaGenevaSwitzerland
| | - Jérôme Flakowski
- Department of Basic Neurosciences, Faculty of Medicine, University of GenevaGenevaSwitzerland
| | - Christian Lüscher
- Department of Basic Neurosciences, Faculty of Medicine, University of GenevaGenevaSwitzerland
- Clinic of Neurology, Dept. of Clinical Neurosciences, Geneva University HospitalGenevaSwitzerland
| |
Collapse
|
50
|
Balleine BW, Peak J, Matamales M, Bertran-Gonzalez J, Hart G. The dorsomedial striatum: an optimal cellular environment for encoding and updating goal-directed learning. Curr Opin Behav Sci 2021. [DOI: 10.1016/j.cobeha.2021.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|