1
|
Liang X, Wu Q, Liu W, Zhou Y, Tan C, Yin H, Sun C. Intrinsic plasticity coding improved spiking actor network for reinforcement learning. Neural Netw 2025; 184:107054. [PMID: 39732066 DOI: 10.1016/j.neunet.2024.107054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/01/2024] [Accepted: 12/12/2024] [Indexed: 12/30/2024]
Abstract
Deep reinforcement learning (DRL) exploits the powerful representational capabilities of deep neural networks (DNNs) and has achieved significant success. However, compared to DNNs, spiking neural networks (SNNs), which operate on binary signals, more closely resemble the biological characteristics of efficient learning observed in the brain. In SNNs, spiking neurons exhibit complex dynamic characteristics and learn based on principles of biological plasticity. Inspired by the brain's efficient computational mechanisms, information encoding plays a critical role in these networks. We propose an intrinsic plasticity coding improved spiking actor network (IP-SAN) for RL to achieve effective decision-making. The IP-SAN integrates adaptive population coding at the network level with dynamic spiking neuron coding at the neuron level, improving spatiotemporal state representation and promoting more accurate biological simulation. Experimental results show that our IP-SAN outperforms several state-of-the-art methods in five continuous control tasks.
Collapse
Affiliation(s)
- Xingyue Liang
- School of Artificial Intelligence, Anhui University, Hefei, 230601, Anhui, China; Engineering Research Center of Autonomous Unmanned System Technology, Ministry of Education, Hefei, 230601, Anhui, China; Anhui Provincial Engineering Research Center for Unmanned Systems and Intelligent Technology, Hefei, 230601, Anhui, China.
| | - Qiaoyun Wu
- School of Artificial Intelligence, Anhui University, Hefei, 230601, Anhui, China; Engineering Research Center of Autonomous Unmanned System Technology, Ministry of Education, Hefei, 230601, Anhui, China; Anhui Provincial Engineering Research Center for Unmanned Systems and Intelligent Technology, Hefei, 230601, Anhui, China.
| | - Wenzhang Liu
- School of Artificial Intelligence, Anhui University, Hefei, 230601, Anhui, China; Engineering Research Center of Autonomous Unmanned System Technology, Ministry of Education, Hefei, 230601, Anhui, China; Anhui Provincial Engineering Research Center for Unmanned Systems and Intelligent Technology, Hefei, 230601, Anhui, China.
| | - Yun Zhou
- School of Artificial Intelligence, Anhui University, Hefei, 230601, Anhui, China; Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, 230601, Anhui, China.
| | - Chunyu Tan
- School of Artificial Intelligence, Anhui University, Hefei, 230601, Anhui, China; Engineering Research Center of Autonomous Unmanned System Technology, Ministry of Education, Hefei, 230601, Anhui, China; Anhui Provincial Engineering Research Center for Unmanned Systems and Intelligent Technology, Hefei, 230601, Anhui, China.
| | - Hongfu Yin
- School of Artificial Intelligence, Anhui University, Hefei, 230601, Anhui, China; Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, 230601, Anhui, China.
| | - Changyin Sun
- School of Artificial Intelligence, Anhui University, Hefei, 230601, Anhui, China; Engineering Research Center of Autonomous Unmanned System Technology, Ministry of Education, Hefei, 230601, Anhui, China; Anhui Provincial Engineering Research Center for Unmanned Systems and Intelligent Technology, Hefei, 230601, Anhui, China; School of Automation, Southeast University, Nanjing, 211189, Jiangsu, China.
| |
Collapse
|
2
|
Wu X, Kobeissi AM, Phillips HL, Dai H, Yao WD. A Prefrontal Cortex-Nucleus Accumbens Circuit Attenuates Cocaine-conditioned Place Preference Memories. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.21.644656. [PMID: 40196555 PMCID: PMC11974754 DOI: 10.1101/2025.03.21.644656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
The infralimbic (IL) subregion of the prefrontal cortex (PFC), via its descending projection to the nucleus accumbens (NAc), inhibits cue-induced drug seeking and reinstatement, but the underlying mechanisms are not fully understood. Here we show that the intrinsic membrane excitability of IL layer 5 pyramidal neurons projecting to the NAc shell (IL-NAcSh neurons) suppresses cocaine-associated memories. Following repeated cocaine exposures in a conditioned place preference paradigm, IL-NAcSh neurons anatomically traced by fluorescent retrobeads undergo prolonged decrease of membrane excitability, lasting for at least 15 days after cocaine withdrawal. This persistent IL-NAcSh neuron hypoexcitability was accompanied by an increase in the rheobase, an increase in the afterhyperpolarization potential, and a decrease in the membrane input resistance. This cocaine induced neuroadapation in intrinsic excitability was not observed in prelimibic cortex neurons projecting to the NAc core (PL-NAcCo neurons), a separate descending circuit thought to promote cue-triggered drug seeking. Chemogenetic restoration of IL-NAcSh neuron activity extinguishes both the acquisition and retention of cocaine conditioned place preference memories. Our results provide direct support for the notion that the IL-NAcSh circuit serves to extinct drug associated memories and restoring the drug impaired excitability of IL-NAcSh neurons has the potential to mitigate drug-cue association memories and drug seeking.
Collapse
Affiliation(s)
- Xiaobo Wu
- Departments of Psychiatry & Behavioral Sciences and of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY 13210
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Jiangsu, China, 226019
| | - Aya M. Kobeissi
- Departments of Psychiatry & Behavioral Sciences and of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY 13210
| | - Hannah L. Phillips
- Departments of Psychiatry & Behavioral Sciences and of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY 13210
| | - Huihui Dai
- Departments of Psychiatry & Behavioral Sciences and of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY 13210
| | - Wei-Dong Yao
- Departments of Psychiatry & Behavioral Sciences and of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
3
|
Berry EA, Huhulea EN, Ishibashi M, McGregor R, Siegel JM, Leonard CS. Chronic but not acute morphine exposure reversibly impairs spike generation and repetitive firing in a functionally distinct subpopulation of orexin neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644444. [PMID: 40196653 PMCID: PMC11974729 DOI: 10.1101/2025.03.20.644444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Orexin (hypocretin) neuropeptides regulate numerous essential functions including sleep/wake state stability and reward processing. Orexin synthesizing neurons respond to drug cues and undergo structural changes following persistent drug exposure. Post-mortem brains from opioid users, and opioid-treated rodents have orexin somata that become ~20 % smaller and ~50% more numerous and are postulated to promote hyper-motivation for drug-seeking though increased orexin release. Biophysical considerations suggest that decreased soma size should increase cellular excitability, however the impact of chronic opioids on firing ability, which drives peptide release, has not been explored. To test this, we assessed the intrinsic electrophysiological properties of orexin neurons by whole-cell recordings in slices from male orexin-EGFP mice treated by daily morphine or saline injections for two weeks. Paradoxically, we found that while daily morphine decreased average soma size, it impaired excitability in a subpopulation of orexin neurons identified by electrophysiological criteria as "H-type", while entirely sparing "D-type" neurons. This impairment was manifest by smaller, broader action potentials, variable firing and a downscaling of firing gain. These adaptations required more than a single morphine dose and recovered, along with soma size, after four weeks of passive withdrawal. Taken together, these observations indicate that daily opioid exposure differentially impacts H-type orexin neurons and predicts that the ability of these neurons to encode synaptic inputs into spike trains and to release neuropeptides becomes impaired in conjunction with opioid dependence.
Collapse
Affiliation(s)
| | - Ellen N. Huhulea
- Department of Physiology, New York Medical College, Valhalla, NY, USA
| | - Masaru Ishibashi
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Ronald McGregor
- Neuropsychiatric Institute, University of California, Los Angeles, CA and Veterans Administration, Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Jerome M. Siegel
- Neuropsychiatric Institute, University of California, Los Angeles, CA and Veterans Administration, Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | | |
Collapse
|
4
|
Anderson EM, Tsvetkov E, Wood D, Akiki RM, Al Hasanieh K, McCue LM, Taniguchi M, Lavin A, Cowan CW. Heroin Regulates the Voltage-Gated Sodium Channel Auxiliary Subunit, SCN1b, to Modulate Nucleus Accumbens Medium Spiny Neuron Intrinsic Excitability and Cue-Induced Heroin Seeking. eNeuro 2025; 12:ENEURO.0017-25.2025. [PMID: 39947903 PMCID: PMC11913320 DOI: 10.1523/eneuro.0017-25.2025] [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: 01/13/2025] [Accepted: 01/28/2025] [Indexed: 03/16/2025] Open
Abstract
Self-administration of addictive substances like heroin can couple the rewarding/euphoric effects of the drug with drug-associated cues, and opioid cue reactivity contributes to relapse vulnerability in abstinent individuals recovering from an opioid use disorder (OUD). Opioids are reported to alter the intrinsic excitability of medium spiny neurons (MSNs) in the nucleus accumbens (NAc), a key brain reward region linked to drug seeking, but how opioids alter NAc MSN neuronal excitability and the impact of altered MSN excitability on relapse-like opioid seeking remain unclear. Here, we discovered that self-administered, but not experimenter-administered, heroin reduced NAc protein levels of the voltage-gated sodium channel auxiliary subunit, SCN1b, in male and female rats. Viral-mediated reduction of NAc SCN1b increased the intrinsic excitability of MSNs, but without altering glutamatergic and GABAergic synaptic transmission. While reducing NAc SCN1b levels had no effect on acquisition of heroin self-administration or extinction learning, we observed a significant increase in cue-reinstated heroin seeking, suggesting that NAc SCN1b normally limits cue-reinstated heroin seeking. We also observed that NAc SCN1b protein levels returned to baseline following heroin self-administration, home-cage abstinence, and extinction training, suggesting that the noted reduction of NAc SCN1b during acquisition of heroin self-administration likely enhances MSN excitability and the strength of heroin-cue associations formed during active heroin use. As such, enhancing NAc SCN1b function might mitigate opioid cue reactivity and a return to active drug use in individuals suffering from OUD.
Collapse
Affiliation(s)
- Ethan M Anderson
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
- Department of Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, Louisiana 70803
| | - Evgeny Tsvetkov
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Daniel Wood
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Rose Marie Akiki
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Karim Al Hasanieh
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Lauren M McCue
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Makoto Taniguchi
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Antonieta Lavin
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Christopher W Cowan
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| |
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
|
Huai Z, Huang B, He G, Li H, Liu Y, Le Q, Wang F, Ma L, Liu X. Accumulation of NMDA receptors in accumbal neuronal ensembles mediates increased conditioned place preference for cocaine after prolonged withdrawal. Prog Neurobiol 2024; 234:102573. [PMID: 38401668 DOI: 10.1016/j.pneurobio.2024.102573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 01/05/2024] [Accepted: 01/16/2024] [Indexed: 02/26/2024]
Abstract
Cue-induced cocaine craving gradually intensifies following abstinence, a phenomenon known as the incubation of drug craving. Neuronal ensembles activated by initial cocaine use, are critically involved in this process. However, the mechanisms by which neuronal changes occurring in the ensembles after withdrawal contribute to incubation remain largely unknown. Here we labeled neuronal ensembles in the shell of nucleus accumbens (NAcSh) activated by cocaine conditioned place preference (CPP) training. NAcSh ensembles showed an increasing activity induced by CPP test after 21-day withdrawal. Inhibiting synaptic transmission of NAcSh ensembles suppressed the preference for cocaine paired-side after 21-day withdrawal, demonstrating a critical role of NAcSh ensembles in increased preference for cocaine. The density of dendritic spines in dopamine D1 receptor expressing ensembles was increased after 21-day withdrawal. Moreover, the expression of Grin1, a subunit of the N-methyl-D-aspartate (NMDA) receptor, specifically increased in the NAcSh ensembles after cocaine withdrawal in both CPP and self-administration (SA) mouse models. Targeted knockdown or dysfunction of Grin1 in NAcSh ensembles significantly suppressed craving for cocaine. Our results suggest that the accumulation of NMDA receptors in NAcSh ensembles mediates increased craving for cocaine after prolonged withdrawal, thereby providing potential molecular targets for treatment of drug addiction.
Collapse
Affiliation(s)
- Ziqing Huai
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Pharmacology Research Center, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Bing Huang
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Pharmacology Research Center, Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Guanhong He
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Pharmacology Research Center, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Haibo Li
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Pharmacology Research Center, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Yonghui Liu
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Pharmacology Research Center, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Qiumin Le
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Pharmacology Research Center, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Feifei Wang
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Pharmacology Research Center, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Lan Ma
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Pharmacology Research Center, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China.
| | - Xing Liu
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Pharmacology Research Center, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China.
| |
Collapse
|
7
|
Wu J, Zhao Y. Single cocaine exposure attenuates the intrinsic excitability of CRH neurons in the ventral BNST via Sigma-1 receptors. Transl Neurosci 2024; 15:20220339. [PMID: 38681523 PMCID: PMC11047800 DOI: 10.1515/tnsci-2022-0339] [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: 12/31/2023] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 05/01/2024] Open
Abstract
The ventral bed nucleus of the stria terminalis (vBNST) plays a key role in cocaine addiction, especially relapse. However, the direct effects of cocaine on corticotropin-releasing hormone (CRH) neurons in the vBNST remain unclear. Here, we identify that cocaine exposure can remarkably attenuate the intrinsic excitability of CRH neurons in the vBNST in vitro. Accumulating studies reveal the crucial role of Sigma-1 receptors (Sig-1Rs) in modulating cocaine addiction. However, to the authors' best knowledge no investigations have explored the role of Sig-1Rs in the vBNST, let alone CRH neurons. Given that cocaine acts as a type of Sig-1Rs agonist, and the dramatic role of Sig-1Rs played in intrinsic excitability of neurons as well as cocaine addiction, we employ BD1063 a canonical Sig-1Rs antagonist to block the effects of cocaine, and significantly recover the excitability of CRH neurons. Together, we suggest that cocaine exposure leads to the firing rate depression of CRH neurons in the vBNST via binding to Sig-1Rs.
Collapse
Affiliation(s)
- Jintao Wu
- School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Yue Zhao
- School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou, China
| |
Collapse
|
8
|
Mulholland PJ, Padula AE, Wilhelm LJ, Park B, Grant KA, Ferguson BM, Cervera-Juanes R. Cross-species epigenetic regulation of nucleus accumbens KCNN3 transcripts by excessive ethanol drinking. Transl Psychiatry 2023; 13:364. [PMID: 38012158 PMCID: PMC10682415 DOI: 10.1038/s41398-023-02676-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 11/29/2023] Open
Abstract
The underlying genetic and epigenetic mechanisms driving functional adaptations in neuronal excitability and excessive alcohol intake are poorly understood. Small-conductance Ca2+-activated K+ (KCa2 or SK) channels encoded by the KCNN family of genes have emerged from preclinical studies as a key contributor to alcohol-induced functional neuroadaptations in alcohol-drinking monkeys and alcohol-dependent mice. Here, this cross-species analysis focused on KCNN3 DNA methylation, gene expression, and single nucleotide polymorphisms, including alternative promoters in KCNN3, that could influence surface trafficking and function of KCa2 channels. Bisulfite sequencing analysis of the nucleus accumbens tissue from alcohol-drinking monkeys and alcohol-dependent mice revealed a differentially methylated region in exon 1A of KCNN3 that overlaps with a predicted promoter sequence. The hypermethylation of KCNN3 in the accumbens paralleled an increase in the expression of alternative transcripts that encode apamin-insensitive and dominant-negative KCa2 channel isoforms. A polymorphic repeat in macaque KCNN3 encoded by exon 1 did not correlate with alcohol drinking. At the protein level, KCa2.3 channel expression in the accumbens was significantly reduced in very heavy-drinking monkeys. Together, our cross-species findings on epigenetic dysregulation of KCNN3 represent a complex mechanism that utilizes alternative promoters to potentially impact the firing of accumbens neurons. Thus, these results provide support for hypermethylation of KCNN3 as a possible key molecular mechanism underlying harmful alcohol intake and alcohol use disorder.
Collapse
Affiliation(s)
- Patrick J Mulholland
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Audrey E Padula
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Larry J Wilhelm
- Department of Translational Neuroscience, Atrium Health Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Byung Park
- Department of Public Health and Preventive Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Kathleen A Grant
- Department of Neurosciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Betsy M Ferguson
- Department of Neurosciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Rita Cervera-Juanes
- Department of Translational Neuroscience, Atrium Health Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
- Center for Precision Medicine, Atrium Health Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
9
|
Juanes RC, Mulholland P, Padula A, Wilhelm L, Park B, Grant K, Ferguson B. Cross-species epigenetic regulation of nucleus accumbens KCNN3 transcripts by excessive ethanol drinking. RESEARCH SQUARE 2023:rs.3.rs-3315122. [PMID: 37790552 PMCID: PMC10543433 DOI: 10.21203/rs.3.rs-3315122/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
The underlying genetic and epigenetic mechanisms driving functional adaptations in neuronal excitability and excessive alcohol intake are poorly understood. Small-conductance Ca2+-activated K+ (KCa2 or SK) channels encoded by the KCNN family of genes have emerged from preclinical studies as a key contributor to alcohol-induced functional neuroadaptations in alcohol-drinking monkeys and alcohol dependent mice. Here, this cross-species analysis focused on KCNN3 DNA methylation, gene expression, and single nucleotide polymorphisms including alternative promoters in KCNN3 that could influence surface trafficking and function of KCa2 channels. Bisulfite sequencing analysis of the nucleus accumbens tissue from alcohol-drinking monkeys and alcohol dependent mice revealed a differentially methylated region in exon 1A of KCNN3 that overlaps with a predicted promoter sequence. The hypermethylation of KCNN3 in the accumbens paralleled an increase in expression of alternative transcripts that encode apamin-insensitive and dominant-negative KCa2 channel isoforms. A polymorphic repeat in macaque KCNN3 encoded by exon 1 did not correlate with alcohol drinking. At the protein level, KCa2.3 channel expression in the accumbens was significantly reduced in very heavy drinking monkeys. Together, our cross-species findings on epigenetic dysregulation of KCNN3 represent a complex mechanism that utilizes alternative promoters to impact firing of accumbens neurons. Thus, these results provide support for hypermethylation of KCNN3 as a possible key molecular mechanism underlying harmful alcohol intake and alcohol use disorder.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Betsy Ferguson
- Oregon Health & Sciences University/Oregon National Primate Research Center
| |
Collapse
|
10
|
He Y, Wang J, Li KL, Wang YQ, Freyberg Z, Dong Y. Membrane excitability of nucleus accumbens neurons gates the incubation of cocaine craving. Neuropsychopharmacology 2023; 48:1318-1327. [PMID: 37041207 PMCID: PMC10354025 DOI: 10.1038/s41386-023-01580-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/15/2023] [Accepted: 03/28/2023] [Indexed: 04/13/2023]
Abstract
After drug withdrawal, a key factor triggering relapse is progressively intensified cue-associated drug craving, termed incubation of drug craving. After withdrawal from cocaine self-administration, incubation of cocaine craving develops more reliably in rats compared to mice. This species difference provides an opportunity to determine rat-specific cellular adaptations, which may constitute the critical mechanisms that contribute to incubated cocaine craving in humans. Expression of incubated cocaine seeking is mediated, in part, by cocaine-induced cellular adaptations in medium spiny neurons (MSNs) within the nucleus accumbens (NAc). In rats, decreased membrane excitability in NAc MSNs is a prominent cellular adaptation, which is induced after cocaine self-administration and lasts throughout prolonged drug withdrawal. Here, we show that, similar to rats, mice exhibit decreased membrane excitability of dopamine D1 receptor (D1)-, but not D2 (D2)-, expressing MSNs within the NAc shell (NAcSh) after 1 d withdrawal from cocaine self-administration. However, in contrast to rats, this membrane adaptation does not persist in mice, diminishing after 45-d withdrawal. We also find that restoring the membrane excitability of NAcSh MSNs after cocaine withdrawal decreases cocaine seeking in rats. This suggests that drug-induced membrane adaptations are essential for behavioral expression of incubated cocaine craving. In mice, however, experimentally inducing hypoactivity of D1 NAcSh MSNs after cocaine withdrawal does not alter cocaine seeking, suggesting that MSN hypo-excitability alone is insufficient to increase cocaine seeking. Together, our results demonstrate an overall permissive role of cocaine-induced hypoactivity of NAcSh MSNs in gating increased cocaine seeking after prolonged cocaine withdrawal.
Collapse
Affiliation(s)
- Yi He
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Junshi Wang
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - King-Lun Li
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Yao Q Wang
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15260, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA.
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
11
|
Salin A, Dugast E, Lardeux V, Solinas M, Belujon P. The amygdala-ventral pallidum pathway contributes to a hypodopaminergic state in the ventral tegmental area during protracted abstinence from chronic cocaine. Br J Pharmacol 2023; 180:1819-1831. [PMID: 36645812 DOI: 10.1111/bph.16034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/01/2022] [Accepted: 01/06/2023] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND AND PURPOSE Incubation of craving, the progressive increase in drug seeking over the first weeks of abstinence, is associated with temporal changes during abstinence in the activity of several structures involved in drug-seeking behaviour. Decreases of dopamine (DA) release and DA neuronal activity (hypodopaminergic state) have been reported in the ventral tegmental area (VTA) during cocaine abstinence, but the mechanisms underlying these neuroadaptations are not well understood. We investigated the potential involvement of a VTA inhibiting circuit (basolateral amygdala [BLA]-ventral pallidum [VP] pathway) in the hypodopaminergic state associated with abstinence from chronic cocaine. EXPERIMENTAL APPROACH In a model of cocaine self-administration, we performed in vivo electrophysiological recordings of DA VTA neurons and BLA neurons from anaesthetised rats during early and protracted abstinence and evaluated the involvement of the BLA-VP pathway using a pharmacological approach. KEY RESULTS We found significant decreases in VTA DA population activity and significant increases in BLA activity after protracted but not after short-term abstinence from chronic cocaine. The decrease in VTA DA activity was restored by pharmacological inhibition of the activity of either the BLA or the VP, suggesting that these regions exert a negative influence on DA activity. CONCLUSION AND IMPLICATIONS Our study sheds new lights on neuroadaptations occurring during incubation of craving leading to relapse. In particular, we describe the involvement of the BLA-VP pathway in cocaine-induced decreases of DA activity in the VTA. This study adds important information about the specific brain network dysfunctions underlying hypodopaminergic activity during abstinence.
Collapse
Affiliation(s)
- Adélie Salin
- Université de Poitiers, INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Poitiers, France
- Université de Rennes, Institut Numecan INRAE, INSERM, Rennes, France
| | - Emilie Dugast
- Université de Poitiers, INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Poitiers, France
- CHU de Poitiers, Poitiers, France
| | - Virginie Lardeux
- Université de Poitiers, INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Marcello Solinas
- Université de Poitiers, INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Pauline Belujon
- Université de Poitiers, INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Poitiers, France
| |
Collapse
|
12
|
Kokane SS, Cole RD, Bordieanu B, Ray CM, Haque IA, Otis JM, McGinty JF. Increased Excitability and Synaptic Plasticity of Drd1- and Drd2-Expressing Prelimbic Neurons Projecting to Nucleus Accumbens after Heroin Abstinence Are Reversed by Cue-Induced Relapse and Protein Kinase A Inhibition. J Neurosci 2023; 43:4019-4032. [PMID: 37094933 PMCID: PMC10255008 DOI: 10.1523/jneurosci.0108-23.2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/23/2023] [Accepted: 04/03/2023] [Indexed: 04/26/2023] Open
Abstract
Dysregulation of the input from the prefrontal cortex (PFC) to the nucleus accumbens (NAc) contributes to cue-induced opioid seeking but the heterogeneity in, and regulation of, prelimbic (PL)-PFC to NAc (PL->NAc) neurons that are altered has not been comprehensively explored. Recently, baseline and opiate withdrawal-induced differences in intrinsic excitability of Drd1+ (D1+) versus Drd2+ (D2+) PFC neurons have been demonstrated. Thus, here we investigated physiological adaptations of PL->NAc D1+ versus D2+ neurons after heroin abstinence and cue-induced relapse. Drd1-Cre+ and Drd2-Cre+ transgenic male Long-Evans rats with virally labeled PL->NAc neurons were trained to self-administer heroin followed by 1 week of forced abstinence. Heroin abstinence significantly increased intrinsic excitability in D1+ and D2+ PL->NAc neurons and increased postsynaptic strength selectively in D1+ neurons. These changes were normalized by cue-induced relapse to heroin seeking. Based on protein kinase A (PKA)-dependent changes in the phosphorylation of plasticity-related proteins in the PL cortex during abstinence and cue-induced relapse to cocaine seeking, we assessed whether the electrophysiological changes in D1+ and D2+ PL->NAc neurons during heroin abstinence were regulated by PKA. In heroin-abstinent PL slices, application of the PKA antagonist (R)-adenosine, cyclic 3',5'-(hydrogenphosphorothioate) triethylammonium (RP-cAMPs) reversed intrinsic excitability in both D1+ and D2+ neurons and postsynaptic strength in only D1+ neurons. Additionally, in vivo bilateral intra-PL infusion of RP-cAMPs after abstinence from heroin inhibited cue-induced relapse to heroin seeking. These data reveal that PKA activity in D1+ and D2+ PL->NAc neurons is not only required for abstinence-induced physiological adaptations but is also required for cue-induced relapse to heroin seeking.SIGNIFICANCE STATEMENT Neuronal plasticity in the medial prefrontal cortex is thought to underlie relapse to drug seeking, yet the subpopulation of neurons that express this plasticity to functionally guide relapse is unclear. Here we show cell type-specific adaptations in Drd1-expressing versus Drd2-expressing prelimbic pyramidal neurons with efferent projections to nucleus accumbens. These adaptations are bidirectionally regulated during abstinence versus relapse and involve protein kinase A (PKA) activation. Furthermore, we show that disruption of the abstinence-associated adaptations via site-specific PKA inhibition abolishes relapse. These data reveal the promising therapeutic potential of PKA inhibition for preventing relapse to heroin seeking and suggest that cell type-specific pharmacologies that target subpopulations of prefrontal neurons would be ideal for future therapeutic developments.
Collapse
Affiliation(s)
- Saurabh S Kokane
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Robert D Cole
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Bogdan Bordieanu
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Chevin M Ray
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Ishraq A Haque
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - James M Otis
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Jacqueline F McGinty
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| |
Collapse
|
13
|
Nimitvilai-Roberts S, Gioia D, Lopez MF, Glaser CM, Woodward JJ. Chronic intermittent ethanol exposure differentially alters the excitability of neurons in the orbitofrontal cortex and basolateral amygdala that project to the dorsal striatum. Neuropharmacology 2023; 228:109463. [PMID: 36792030 PMCID: PMC10006395 DOI: 10.1016/j.neuropharm.2023.109463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023]
Abstract
Alcohol use disorder is associated with altered neuron function including those in orbitofrontal cortex (OFC) and basolateral amygdala (BLA) that send glutamatergic inputs to areas of the dorsal striatum (DS) that mediate goal and habit directed actions. Previous studies reported that chronic intermittent (CIE) exposure to ethanol alters the electrophysiological properties of OFC and BLA neurons, although projection targets for these neurons were not identified. In this study, we used male and female mice and recorded current-evoked spiking of retrobead labeled DS-projecting OFC and BLA neurons in the same animals following air or CIE treatment. DS-projecting OFC neurons were hyperexcitable 3- and 7-days following CIE exposure and spiking returned to control levels after 14 days of withdrawal. In contrast, firing was decreased in DS-projecting BLA neurons at 3-days withdrawal, increased at 7- and 14-days and returned to baseline at 28 days post-CIE. CIE exposure enhanced the amplitude and frequency of spontaneous excitatory postsynaptic currents (sEPSCs) of DS-projecting OFC neurons but had no effect on inhibitory postsynaptic currents (sIPSCs). In DS-projecting BLA neurons, the amplitude and frequency of sIPSCs was enhanced 3 days post-CIE with no change in sEPSCs while at 7-days post-withdrawal, sEPSC amplitude and frequency were increased and sIPSCs had returned to normal. Finally, in CIE-treated mice, acute ethanol no longer inhibited spike firing of DS-projecting OFC and BLA neurons. Overall, these results suggest that CIE-induced changes in the excitability of DS-projecting OFC and BLA neurons could underlie deficits in behavioral control often observed in alcohol-dependent individuals.
Collapse
Affiliation(s)
| | - Dominic Gioia
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Marcelo F Lopez
- Department of Psychiatry and Behavioral Sciences, Addiction Sciences Division, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Christina M Glaser
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - John J Woodward
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA; Department of Psychiatry and Behavioral Sciences, Addiction Sciences Division, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
14
|
Chen F, Xu Y, Shi K, Zhang Z, Xie Z, Wu H, Ma Y, Zhou Y, Chen C, Yang J, Wang Y, Robbins TW, Wang K, Yu J. Multi-omics study reveals associations among neurotransmitter, extracellular vesicle-derived microRNA and psychiatric comorbidities during heroin and methamphetamine withdrawal. Biomed Pharmacother 2022; 155:113685. [PMID: 36137407 DOI: 10.1016/j.biopha.2022.113685] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 08/31/2022] [Accepted: 09/07/2022] [Indexed: 11/02/2022] Open
Abstract
Despite decades of research in the field of substance withdrawal, molecular biomarkers and related mechanistic study have generally been lacking. In addition to known neurotransmitters, circulating miRNAs are found in small vesicles known as exosomes within blood that have diagnostic potential and are known to contribute to psychiatric disorders. The aim of this work was to characterize the changes in neurotransmitter and exosomal miRNA profiles during heroin and methamphetamine withdrawal using a cross-sectional study design, and to determine their associations to psychiatric comorbidities in a large group of patients with substance use disorders (SUDs). Using weighted gene co-expression network analysis, a series of known, conserved, and novel exosomal miRNAs were identified as being associated with the severity of anxiety and depression, as well as the concentrations of neurotransmitters GABA, choline, and serotonin. Bioinformatics analyses established that the differences in the miRNA profile target signaling pathways are significantly associated with developmental and intellectual abnormalities. Notably, a set of dysregulated miRNA signatures including hsa-mia-451a and hsa-mir-21a resulted in an AUC of 0.966 and 0.861, respectively, for predicting the patients with SUDs. Furthermore, hsa-miR-744a-5p was positively correlated with serotonin, and its important role in maintaining neuronal development and function was revealed using an in vitro human induced pluripotent stem cells derived neuronal model. Our results suggest that the miRNA content of circulating exosomes represent a biomolecular "fingerprint" of the progression of substance withdrawal and may uncover the putative mechanism of how these exosomal miRNAs contribute to psychiatric symptoms.
Collapse
Affiliation(s)
- Fengrong Chen
- NHC Key Laboratory of Drug Addiction Medicine (Kunming Medical University), First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China; School of Medicine, Kunming University of Science and Technology
| | - Yu Xu
- NHC Key Laboratory of Drug Addiction Medicine (Kunming Medical University), First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Kai Shi
- College of Science, Guilin University of Technology, Guilin 541004, China
| | - Zunyue Zhang
- NHC Key Laboratory of Drug Addiction Medicine (Kunming Medical University), First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China; School of Medicine, Yunnan University, Yunnan, China
| | - Zhenrong Xie
- NHC Key Laboratory of Drug Addiction Medicine (Kunming Medical University), First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China; Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Hongjin Wu
- NHC Key Laboratory of Drug Addiction Medicine (Kunming Medical University), First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China; Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Yuru Ma
- NHC Key Laboratory of Drug Addiction Medicine (Kunming Medical University), First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China; Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Yong Zhou
- NHC Key Laboratory of Drug Addiction Medicine (Kunming Medical University), First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China; Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Cheng Chen
- NHC Key Laboratory of Drug Addiction Medicine (Kunming Medical University), First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China; Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Jiqing Yang
- NHC Key Laboratory of Drug Addiction Medicine (Kunming Medical University), First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China; Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Yuan Wang
- Department of R&D, Echo Biotech Co., Ltd, Beijing, China
| | - Trevor W Robbins
- Department of Psychology and the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom; Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Kunhua Wang
- NHC Key Laboratory of Drug Addiction Medicine (Kunming Medical University), First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China; School of Medicine, Yunnan University, Yunnan, China; Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China.
| | - Juehua Yu
- NHC Key Laboratory of Drug Addiction Medicine (Kunming Medical University), First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China; Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China.
| |
Collapse
|
15
|
Dvořáček J, Kodrík D. Drug effect and addiction research with insects - From Drosophila to collective reward in honeybees. Neurosci Biobehav Rev 2022; 140:104816. [PMID: 35940307 DOI: 10.1016/j.neubiorev.2022.104816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 10/16/2022]
Abstract
Animals and humans share similar reactions to the effects of addictive substances, including those of their brain networks to drugs. Our review focuses on simple invertebrate models, particularly the honeybee (Apis mellifera), and on the effects of drugs on bee behaviour and brain functions. The drug effects in bees are very similar to those described in humans. Furthermore, the honeybee community is a superorganism in which many collective functions outperform the simple sum of individual functions. The distribution of reward functions in this superorganism is unique - although sublimated at the individual level, community reward functions are of higher quality. This phenomenon of collective reward may be extrapolated to other animal species living in close and strictly organised societies, i.e. humans. The relationship between sociality and reward, based on use of similar parts of the neural network (social decision-making network in mammals, mushroom body in bees), suggests a functional continuum of reward and sociality in animals.
Collapse
Affiliation(s)
- Jiří Dvořáček
- Institute of Entomology, Biology Centre, Czech Academy of Sciences, Branišovská 31, 370 05, České Budĕjovice, Czech Republic; Faculty of Science, University of South Bohemia, Branišovská 31, 370 05, České Budĕjovice, Czech Republic.
| | - Dalibor Kodrík
- Institute of Entomology, Biology Centre, Czech Academy of Sciences, Branišovská 31, 370 05, České Budĕjovice, Czech Republic; Faculty of Science, University of South Bohemia, Branišovská 31, 370 05, České Budĕjovice, Czech Republic
| |
Collapse
|
16
|
Marrero-Cristobal G, Gelpi-Dominguez U, Morales-Silva R, Alvarado-Torres J, Perez-Torres J, Perez-Perez Y, Sepulveda-Orengo M. Aerobic exercise as a promising nonpharmacological therapy for the treatment of substance use disorders. J Neurosci Res 2022; 100:1602-1642. [PMID: 34850988 PMCID: PMC9156662 DOI: 10.1002/jnr.24990] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 10/06/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022]
Abstract
Despite the prevalence and public health impact of substance use disorders (SUDs), effective long-term treatments remain elusive. Aerobic exercise is a promising, nonpharmacological treatment currently under investigation as a strategy for preventing drug relapse. Aerobic exercise could be incorporated into the comprehensive treatment regimens for people with substance abuse disorders. Preclinical studies of SUD with animal models have shown that aerobic exercise diminishes drug-seeking behavior, which leads to relapse, in both male and female rats. Nevertheless, little is known regarding the effects of substance abuse-induced cellular and physiological adaptations believed to be responsible for drug-seeking behavior. Accordingly, the overall goal of this review is to provide a summary and an assessment of findings to date, highlighting evidence of the molecular and neurological effects of exercise on adaptations associated with SUD.
Collapse
Affiliation(s)
| | - Ursula Gelpi-Dominguez
- School of Behavioral and Brain Sciences, Ponce Health Sciences University, Ponce, PR, USA
| | - Roberto Morales-Silva
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR, USA
| | - John Alvarado-Torres
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR, USA
| | - Joshua Perez-Torres
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR, USA
| | - Yobet Perez-Perez
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR, USA
| | - Marian Sepulveda-Orengo
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR, USA
| |
Collapse
|
17
|
Glycogen Synthase Kinase 3: Ion Channels, Plasticity, and Diseases. Int J Mol Sci 2022; 23:ijms23084413. [PMID: 35457230 PMCID: PMC9028019 DOI: 10.3390/ijms23084413] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 12/15/2022] Open
Abstract
Glycogen synthase kinase 3β (GSK3) is a multifaceted serine/threonine (S/T) kinase expressed in all eukaryotic cells. GSK3β is highly enriched in neurons in the central nervous system where it acts as a central hub for intracellular signaling downstream of receptors critical for neuronal function. Unlike other kinases, GSK3β is constitutively active, and its modulation mainly involves inhibition via upstream regulatory pathways rather than increased activation. Through an intricate converging signaling system, a fine-tuned balance of active and inactive GSK3β acts as a central point for the phosphorylation of numerous primed and unprimed substrates. Although the full range of molecular targets is still unknown, recent results show that voltage-gated ion channels are among the downstream targets of GSK3β. Here, we discuss the direct and indirect mechanisms by which GSK3β phosphorylates voltage-gated Na+ channels (Nav1.2 and Nav1.6) and voltage-gated K+ channels (Kv4 and Kv7) and their physiological effects on intrinsic excitability, neuronal plasticity, and behavior. We also present evidence for how unbalanced GSK3β activity can lead to maladaptive plasticity that ultimately renders neuronal circuitry more vulnerable, increasing the risk for developing neuropsychiatric disorders. In conclusion, GSK3β-dependent modulation of voltage-gated ion channels may serve as an important pharmacological target for neurotherapeutic development.
Collapse
|
18
|
Klemz A, Wildner F, Tütüncü E, Gerevich Z. Regulation of Hippocampal Gamma Oscillations by Modulation of Intrinsic Neuronal Excitability. Front Neural Circuits 2022; 15:778022. [PMID: 35177966 PMCID: PMC8845518 DOI: 10.3389/fncir.2021.778022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022] Open
Abstract
Ion channels activated around the subthreshold membrane potential determine the likelihood of neuronal firing in response to synaptic inputs, a process described as intrinsic neuronal excitability. Long-term plasticity of chemical synaptic transmission is traditionally considered the main cellular mechanism of information storage in the brain; however, voltage- and calcium-activated channels modulating the inputs or outputs of neurons are also subjects of plastic changes and play a major role in learning and memory formation. Gamma oscillations are associated with numerous higher cognitive functions such as learning and memory, but our knowledge of their dependence on intrinsic plasticity is by far limited. Here we investigated the roles of potassium and calcium channels activated at near subthreshold membrane potentials in cholinergically induced persistent gamma oscillations measured in the CA3 area of rat hippocampal slices. Among potassium channels, which are responsible for the afterhyperpolarization in CA3 pyramidal cells, we found that blockers of SK (KCa2) and KV7.2/7.3 (KCNQ2/3), but not the BK (KCa1.1) and IK (KCa3.1) channels, increased the power of gamma oscillations. On the contrary, activators of these channels had an attenuating effect without affecting the frequency. Pharmacological blockade of the low voltage-activated T-type calcium channels (CaV3.1–3.3) reduced gamma power and increased the oscillation peak frequency. Enhancement of these channels also inhibited the peak power without altering the frequency of the oscillations. The presented data suggest that voltage- and calcium-activated ion channels involved in intrinsic excitability strongly regulate the power of hippocampal gamma oscillations. Targeting these channels could represent a valuable pharmacological strategy against cognitive impairment.
Collapse
|
19
|
Viet NM, Wang T, Tran-Anh K, Sugihara I. Heterogeneity of intrinsic plasticity in cerebellar Purkinje cells linked with cortical molecular zones. iScience 2022; 25:103705. [PMID: 35059609 PMCID: PMC8760437 DOI: 10.1016/j.isci.2021.103705] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/26/2021] [Accepted: 12/23/2021] [Indexed: 11/28/2022] Open
Abstract
In the cerebellar cortex, heterogeneous populations of Purkinje cells (PCs), classified into zebrin (aldolase C)-positive (Z+) and -negative (Z-) types, are arranged into separate longitudinal zones. They have different topographic neuronal connections and show different patterns of activity in behavior tasks. However, whether the zebrin type of PCs directly links with the physiological properties of the PC has not been well clarified. Therefore, we applied in vitro whole-cell patch-clamp recording in Z+ and Z- PCs in vermal and hemispheric neighboring zebrin zones in zebrin-visualized mice. Intrinsic excitability is significantly higher in Z- PCs than in Z+ PCs. Furthermore, intrinsic plasticity and synaptic long-term potentiation are enhanced more in Z- PCs than in Z+ PCs. The difference was mediated by different modulation of SK channel activities between Z+ and Z- PCs. The results indicate that cellular physiology differentially tunes to the functional compartmentalization of heterogeneous PCs.
Collapse
Affiliation(s)
- Nguyen-Minh Viet
- Department of Systems Neurophysiology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tianzhuo Wang
- Department of Systems Neurophysiology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Khoa Tran-Anh
- Department of Systems Neurophysiology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Izumi Sugihara
- Department of Systems Neurophysiology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
20
|
Zinsmaier AK, Dong Y, Huang YH. Cocaine-induced projection-specific and cell type-specific adaptations in the nucleus accumbens. Mol Psychiatry 2022; 27:669-686. [PMID: 33963288 PMCID: PMC8691189 DOI: 10.1038/s41380-021-01112-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 03/29/2021] [Accepted: 04/09/2021] [Indexed: 02/03/2023]
Abstract
Cocaine craving, seeking, and relapse are mediated, in part, by cocaine-induced adaptive changes in the brain reward circuits. The nucleus accumbens (NAc) integrates and prioritizes different emotional and motivational inputs to the reward system by processing convergent glutamatergic projections from the medial prefrontal cortex, basolateral amygdala, ventral hippocampus, and other limbic and paralimbic brain regions. Medium spiny neurons (MSNs) are the principal projection neurons in the NAc, which can be divided into two major subpopulations, namely dopamine receptor D1- versus D2-expressing MSNs, with complementing roles in reward-associated behaviors. After cocaine experience, NAc MSNs exhibit complex and differential adaptations dependent on cocaine regimen, withdrawal time, cell type, location (NAc core versus shell), and related input and output projections, or any combination of these factors. Detailed characterization of these cellular adaptations has been greatly facilitated by the recent development of optogenetic/chemogenetic techniques combined with transgenic tools. In this review, we discuss such cell type- and projection-specific adaptations induced by cocaine experience. Specifically, (1) D1 and D2 NAc MSNs frequently exhibit differential adaptations in spinogenesis, glutamatergic receptor trafficking, and intrinsic membrane excitability, (2) cocaine experience differentially changes the synaptic transmission at different afferent projections onto NAc MSNs, (3) cocaine-induced NAc adaptations exhibit output specificity, e.g., being different at NAc-ventral pallidum versus NAc-ventral tegmental area synapses, and (4) the input, output, subregion, and D1/D2 cell type may together determine cocaine-induced circuit plasticity in the NAc. In light of the projection- and cell-type specificity, we also briefly discuss ensemble and circuit mechanisms contributing to cocaine craving and relapse after drug withdrawal.
Collapse
Affiliation(s)
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15219,Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219
| | - Yanhua H. Huang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219
| |
Collapse
|
21
|
Kiguchi N, Ko MC. Potential therapeutic targets for the treatment of opioid abuse and pain. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 93:335-371. [PMID: 35341570 PMCID: PMC10948018 DOI: 10.1016/bs.apha.2021.09.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Although μ-opioid peptide (MOP) receptor agonists are effective analgesics available in clinical settings, their serious adverse effects put limits on their use. The marked increase in abuse and misuse of prescription opioids for pain relief and opioid overdose mortality in the past decade has seriously impacted society. Therefore, safe analgesics that produce potent analgesic effects without causing MOP receptor-related adverse effects are needed. This review highlights the potential therapeutic targets for the treatment of opioid abuse and pain based on available evidence generated through preclinical studies and clinical trials. To ameliorate the abuse-related effects of opioids, orexin-1 receptor antagonists and mixed nociceptin/MOP partial agonists have shown promising results in translational aspects of animal models. There are several promising non-opioid targets for selectively inhibiting pain-related responses, including nerve growth factor inhibitors, voltage-gated sodium channel inhibitors, and cannabinoid- and nociceptin-related ligands. We have also discussed several emerging and novel targets. The current medications for opioid abuse are opioid receptor-based ligands. Although neurobiological studies in rodents have discovered several non-opioid targets, there is a translational gap between rodents and primates. Given that the neuroanatomical aspects underlying opioid abuse and pain are different between rodents and primates, it is pivotal to investigate the functional profiles of these non-opioid compounds compared to those of clinically used drugs in non-human primate models before initiating clinical trials. More pharmacological studies of the functional efficacy, selectivity, and tolerability of these newly discovered compounds in non-human primates will accelerate the development of effective medications for opioid abuse and pain.
Collapse
Affiliation(s)
- Norikazu Kiguchi
- Department of Physiological Sciences, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Japan.
| | - Mei-Chuan Ko
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
22
|
Metz VG, da Rosa JLO, Rossato DR, Milanesi LH, Burger ME, Pase CS. Cannabidiol prevents amphetamine relapse and modulates D1- and D2-receptor levels in mesocorticolimbic brain areas of rats. Eur Neuropsychopharmacol 2021; 50:23-33. [PMID: 33951588 DOI: 10.1016/j.euroneuro.2021.04.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 03/01/2021] [Accepted: 04/11/2021] [Indexed: 12/16/2022]
Abstract
Amphetamine (AMPH) is an addictive psychostimulant highly used worldwide and its consumption is related to neurotoxic effects. Currently, there is no pharmacotherapy approved for treating AMPH or other psychostimulant drug addiction. Different studies have shown promising properties of cannabidiol (CBD) for treating many neurological and psychiatric diseases, and recently, CBD is being considered a potential strategy for the treatment of drug addiction disorders. Thus, we investigated possible CBD beneficial effects on relapse symptoms following AMPH re-exposure considering drug relapse is the most difficult clinical factor to control during addiction treatment. Rats received d,l-AMPH (4 mg/kg, i.p.) or vehicle in the conditioned place preference (CPP) paradigm (8 days), when each experimental group was re-assigned to receive CBD at two different doses (5 or 10 mg/kg, i.p) or control, for 5 days. Subsequently, animals were re-exposed to AMPH-CPP (4 mg/kg, i.p.) for 3 additional days to assess relapse behavior. Besides locomotor and anxiety-like behaviors, dopaminergic molecular parameters were quantified in both prefrontal cortex and ventral striatum. Regarding molecular levels, CBD modulated at basal levels the dopaminergic targets (D1R, D2R, DAT, and TH) in the assessed brain areas, preventing AMPH relapse and decreasing anxiety-like behavior per se and in AMPH-CPP animals. The current findings give evidence about CBD-induced AMPH-relapse prevention, which may be linked to dopaminergic mesocorticolimbic system modulation. Although future and clinical studies are needed, our outcomes show that CBD may be a useful alternative to prevent AMPH relapse.
Collapse
Affiliation(s)
- Vinícia Garzella Metz
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | | | - Domenika Rubert Rossato
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Laura Hautrive Milanesi
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Marilise Escobar Burger
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil.
| | - Camila Simonetti Pase
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil; Universidade Federal do Pampa, Campus Uruguaiana, 97500-970 Uruguaiana, RS, Brazil.
| |
Collapse
|
23
|
Zhang J, Zhang C, Chen X, Wang B, Ma W, Yang Y, Zheng R, Huang Z. PKA-RIIβ autophosphorylation modulates PKA activity and seizure phenotypes in mice. Commun Biol 2021; 4:263. [PMID: 33649504 PMCID: PMC7921646 DOI: 10.1038/s42003-021-01748-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 12/22/2020] [Indexed: 11/20/2022] Open
Abstract
Temporal lobe epilepsy (TLE) is one of the most common and intractable neurological disorders in adults. Dysfunctional PKA signaling is causally linked to the TLE. However, the mechanism underlying PKA involves in epileptogenesis is still poorly understood. In the present study, we found the autophosphorylation level at serine 114 site (serine 112 site in mice) of PKA-RIIβ subunit was robustly decreased in the epileptic foci obtained from both surgical specimens of TLE patients and seizure model mice. The p-RIIβ level was negatively correlated with the activities of PKA. Notably, by using a P-site mutant that cannot be autophosphorylated and thus results in the released catalytic subunit to exert persistent phosphorylation, an increase in PKA activities through transduction with AAV-RIIβ-S112A in hippocampal DG granule cells decreased mIPSC frequency but not mEPSC, enhanced neuronal intrinsic excitability and seizure susceptibility. In contrast, a reduction of PKA activities by RIIβ knockout led to an increased mIPSC frequency, a reduction in neuronal excitability, and mice less prone to experimental seizure onset. Collectively, our data demonstrated that the autophosphorylation of RIIβ subunit plays a critical role in controlling neuronal and network excitabilities by regulating the activities of PKA, providing a potential therapeutic target for TLE.
Collapse
Affiliation(s)
- Jingliang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Chenyu Zhang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xiaoling Chen
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Bingwei Wang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Weining Ma
- Department of Neurology, Shengjing Hospital Affiliated to China Medical University, Shenyang, China
| | - Yang Yang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University College of Pharmacy, West Lafayette, IN, USA
| | - Ruimao Zheng
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
- Neuroscience Research Institute, Peking University, Beijing, China.
- Key Laboratory for Neuroscience, Ministry of Education, Beijing, China.
- Key Laboratory for Neuroscience of National Health Commission, Beijing, China.
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China.
- Key Laboratory for Neuroscience, Ministry of Education, Beijing, China.
- Key Laboratory for Neuroscience of National Health Commission, Beijing, China.
| |
Collapse
|
24
|
Chapp AD, Mermelstein PG, Thomas MJ. The ethanol metabolite acetic acid activates mouse nucleus accumbens shell medium spiny neurons. J Neurophysiol 2021; 125:620-627. [PMID: 33405999 DOI: 10.1152/jn.00659.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Although ethanol consumption leads to an array of neurophysiological alterations involving the neural circuits for reward, the underlying mechanisms remain unclear. Acetic acid is a major metabolite of ethanol with high bioactivity and potentially significant pharmacological importance in regulating brain function. Yet, the impact of acetic acid on reward circuit function has not been well explored. Given the rewarding properties associated with ethanol consumption, we investigated the acute effects of ethanol and/or acetic acid on the neurophysiological function of medium spiny neurons of the nucleus accumbens shell, a key node in the mammalian reward circuit. We find that acetic acid, but not ethanol, provided a rapid and robust boost in neuronal excitability at physiologically relevant concentrations, whereas both compounds enhanced glutamatergic synaptic activity. These effects were consistent across both sexes in C57BL/6J mice. Overall, our data suggest acetic acid is a promising candidate mediator for ethanol effects on mood and motivation that deserves further investigation.NEW & NOTEWORTHY Ethanol consumption disrupts many neurophysiological processes leading to alterations in behavior and physiological function. The possible involvement of acetic acid, produced via ethanol metabolism, has been insufficiently explored. Here, we demonstrate that acetic acid contributes to rapid neurophysiological alterations in the accumbens shell. These findings raise the interesting possibility that ethanol may serve as a prodrug-generating acetic acid as a metabolite-that may influence ethanol consumption-associated behaviors and physiological responses by altering neurophysiological function.
Collapse
Affiliation(s)
- Andrew D Chapp
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota.,Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
| | - Paul G Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota.,Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
| | - Mark J Thomas
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota.,Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
25
|
Zhou Y, Yan E, Cheng D, Zhu H, Liu Z, Chen X, Ma L, Liu X. The Projection From Ventral CA1, Not Prefrontal Cortex, to Nucleus Accumbens Core Mediates Recent Memory Retrieval of Cocaine-Conditioned Place Preference. Front Behav Neurosci 2020; 14:558074. [PMID: 33304246 PMCID: PMC7701212 DOI: 10.3389/fnbeh.2020.558074] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 10/15/2020] [Indexed: 01/15/2023] Open
Abstract
Drug-paired cues inducing memory retrieval by expressing drug-seeking behaviors present a major challenge to drug abstinence. How neural circuits coordinate for drug memory retrieval remains unclear. Here, we report that exposure of the training chamber where cocaine-conditioned place preference (CPP) was performed increased neuronal activity in the core of nucleus accumbens (AcbC), ventral CA1 (vCA1), and medial prefrontal cortex (mPFC), as shown by elevated pERK and c-Fos levels. Chemogenetic inhibition of neuronal activity in the vCA1 and AcbC, but not mPFC, reduced the time spent in the cocaine-paired compartment, suggesting that the vCA1 and AcbC are required for the retrieval of cocaine-CPP memory and are key nodes recruited for cocaine memory storage. Furthermore, chemogenetic inhibition of the AcbC-projecting vCA1 neurons, but not the AcbC-projecting mPFC neurons, decreased the expression of cocaine-CPP. Optogenetic inhibition of the vCA1–AcbC projection, but not the mPFC–AcbC projection, also reduced the preference for the cocaine-paired compartment. Taken together, the cue-induced natural recall of cocaine memory depends on vCA1–AcbC circuits. The connectivity from the vCA1 to the AcbC may store the information of the cue–cocaine reward association critically required for memory retrieval. These data thus provide insights into the neural circuit basis of retrieval of drug-related memory.
Collapse
Affiliation(s)
- Yiming Zhou
- The State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences and the Institutes of Brain Science, Fudan University, Shanghai, China
| | - Enhui Yan
- The State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences and the Institutes of Brain Science, Fudan University, Shanghai, China
| | - Deqin Cheng
- The State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences and the Institutes of Brain Science, Fudan University, Shanghai, China
| | - Huiwen Zhu
- The State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences and the Institutes of Brain Science, Fudan University, Shanghai, China
| | - Zhiyuan Liu
- The State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences and the Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xi Chen
- The State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences and the Institutes of Brain Science, Fudan University, Shanghai, China
| | - Lan Ma
- The State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences and the Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xing Liu
- The State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences and the Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Jaudon F, Thalhammer A, Cingolani LA. Integrin adhesion in brain assembly: From molecular structure to neuropsychiatric disorders. Eur J Neurosci 2020; 53:3831-3850. [PMID: 32531845 DOI: 10.1111/ejn.14859] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/21/2020] [Accepted: 06/02/2020] [Indexed: 02/07/2023]
Abstract
Integrins are extracellular matrix receptors that mediate biochemical and mechanical bi-directional signals between the extracellular and intracellular environment of a cell thanks to allosteric conformational changes. In the brain, they are found in both neurons and glial cells, where they play essential roles in several aspects of brain development and function, such as cell migration, axon guidance, synaptogenesis, synaptic plasticity and neuro-inflammation. Although there are many successful examples of how regulating integrin adhesion and signaling can be used for therapeutic purposes, for example for halting tumor progression, this is not the case for the brain, where the growing evidence of the importance of integrins for brain pathophysiology has not translated yet into medical applications. Here, we review recent literature showing how alterations in integrin structure, expression and signaling may be involved in the etiology of autism spectrum disorder, epilepsy, schizophrenia, addiction, depression and Alzheimer's disease. We focus on common mechanisms and recurrent signaling pathways, trying to bridge studies on the genetics and molecular structure of integrins with those on synaptic physiology and brain pathology. Further, we discuss integrin-targeting strategies and their potential benefits for therapeutic purposes in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Fanny Jaudon
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Agnes Thalhammer
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Lorenzo A Cingolani
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Genoa, Italy.,Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
27
|
Cannady R, Nimitvilai-Roberts S, Jennings SD, Woodward JJ, Mulholland PJ. Distinct Region- and Time-Dependent Functional Cortical Adaptations in C57BL/6J Mice after Short and Prolonged Alcohol Drinking. eNeuro 2020; 7:ENEURO.0077-20.2020. [PMID: 32439714 PMCID: PMC7307629 DOI: 10.1523/eneuro.0077-20.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/03/2020] [Accepted: 05/15/2020] [Indexed: 12/27/2022] Open
Abstract
Alcohol (ethanol) use disorder is associated with changes in frontal cortical areas including the anterior cingulate cortex (ACC) and orbitofrontal cortex (OFC) that contribute to cognitive deficits, uncontrolled drinking, and relapse. Acute ethanol exposure reduces intrinsic excitability of lateral OFC (lOFC) neurons, while chronic exposure and long-term drinking influence plasticity of intrinsic excitability and function of glutamatergic synapses. However, the time course that these adaptations occur across a history of ethanol drinking is unknown. The current study examined whether short-term and long-term voluntary ethanol consumption using an intermittent access paradigm would alter the biophysical properties of deep-layer pyramidal neurons in the ACC and lOFC. Neuronal spiking varied in the ACC with an initial increase in evoked firing after 1 d of drinking followed by a decrease in firing in mice that consumed ethanol for one week. No difference in lOFC spike number was observed between water controls and 1-d ethanol drinking mice, but mice that consumed ethanol for one week or more showed a significant increase in evoked firing. Voluntary ethanol drinking for 4 weeks also produced a total loss of ethanol inhibition of lOFC neurons. There was no effect of drinking on excitatory or inhibitory synaptic events in ACC or lOFC neurons across all time points in this model. Overall, these results demonstrate that voluntary drinking alters neuronal excitability in the ACC and lOFC in distinct ways and on a different time scale that may contribute to the impairment of prefrontal cortex-dependent behaviors observed in individuals with alcohol use disorder (AUD).
Collapse
Affiliation(s)
| | | | - Sarah D Jennings
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425
| | - John J Woodward
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425
| | - Patrick J Mulholland
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425
| |
Collapse
|
28
|
Chouhan D, Uniyal A, Gadepalli A, Akhilesh, Tiwari V, Agrawal S, Roy TK, Shaw S, Purohit N, Tiwari V. Probing the Manipulated Neurochemical Drive in Alcohol Addiction and Novel Therapeutic Advancements. ACS Chem Neurosci 2020; 11:1210-1217. [PMID: 32243128 DOI: 10.1021/acschemneuro.0c00073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Alcohol addiction is one of the highly prevalent neurological disorders and a major threat to public health in the 21st century. Alcohol addiction affects people from all age groups and often leads to other serious comorbidities. The pathophysiology of alcohol addiction involves imbalance between the excitatory and inhibitory neurotransmitters in the brain. These changes occur in various regions of the brain including reward circuit such as the ventral tegmental area (VTA), nucleus accumbens (NAc), and prefrontal cortex. In this review, we have discussed several neurochemical circuitries which get manipulated and maladapted during alcohol addiction. To date there is no effective therapeutic intervention in clinics devoid of side effects that can successfully treat the patients suffering from alcohol addiction. Understanding the neurobiological intricacies of alcohol addiction is critical for the development of novel anti-addiction therapeutics. Apart from this, we have also discussed the recent therapeutic milestones for the management of alcohol addiction including vasopressin receptors, corticotrophin-releasing factor, GABA receptors, glucocorticoid receptors, brain stimulation and mindfulness-oriented recovery enhancement.
Collapse
Affiliation(s)
- Deepak Chouhan
- Neuroscience and Pain Research Lab, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Ankit Uniyal
- Neuroscience and Pain Research Lab, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Anagha Gadepalli
- Neuroscience and Pain Research Lab, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Akhilesh
- Neuroscience and Pain Research Lab, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Vineeta Tiwari
- Neuroscience and Pain Research Lab, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Somesh Agrawal
- Neuroscience and Pain Research Lab, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Tapas Kumar Roy
- Neuroscience and Pain Research Lab, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Sneha Shaw
- Neuroscience and Pain Research Lab, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Narendra Purohit
- Neuroscience and Pain Research Lab, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Vinod Tiwari
- Neuroscience and Pain Research Lab, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| |
Collapse
|
29
|
Matovic S, Ichiyama A, Igarashi H, Salter EW, Sunstrum JK, Wang XF, Henry M, Kuebler ES, Vernoux N, Martinez-Trujillo J, Tremblay ME, Inoue W. Neuronal hypertrophy dampens neuronal intrinsic excitability and stress responsiveness during chronic stress. J Physiol 2020; 598:2757-2773. [PMID: 32347541 DOI: 10.1113/jp279666] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/17/2020] [Indexed: 01/16/2023] Open
Abstract
KEY POINTS The hypothalamic-pituitary-adrenal (HPA) axis habituates to repeated stress exposure. We studied hypothalamic corticotropin-releasing hormone (CRH) neurons that form the apex of the HPA axis in a mouse model of stress habituation using repeated restraint. The intrinsic excitability of CRH neurons decreased after repeated stress in a time course that coincided with the development of HPA axis habituation. This intrinsic excitability plasticity co-developed with an expansion of surface membrane area, which increased a passive electric load and dampened membrane depolarization in response to the influx of positive charge. We report a novel structure-function relationship for intrinsic excitability plasticity as a neural correlate for HPA axis habituation. ABSTRACT Encountering a stressor immediately activates the hypothalamic-pituitary-adrenal (HPA) axis, but this stereotypic stress response also undergoes experience-dependent adaptation. Despite the biological and clinical importance, how the brain adjusts stress responsiveness in the long term remains poorly understood. We studied hypothalamic corticotropin-releasing hormone neurons that form the apex of the HPA axis in a mouse model of stress habituation using repeated restraint. Using patch-clamp electrophysiology in acute slices, we found that the intrinsic excitability of these neurons substantially decreased after daily repeated stress in a time course that coincided with their loss of stress responsiveness in vivo. This intrinsic excitability plasticity co-developed with an expansion of surface membrane area, which increased a passive electric load, and dampened membrane depolarization in response to the influx of positive charge. Multiphoton imaging and electron microscopy revealed that repeated stress augmented ruffling of the plasma membrane, suggesting an ultrastructural plasticity that may efficiently accommodate the membrane area expansion. Overall, we report a novel structure-function relationship for intrinsic excitability plasticity as a neural correlate for adaptation of the neuroendocrine stress response.
Collapse
Affiliation(s)
- Sara Matovic
- Robarts Research Institute, University of Western Ontario.,Neuroscience Program, University of Western Ontario
| | - Aoi Ichiyama
- Neuroscience Program, University of Western Ontario
| | | | - Eric W Salter
- Robarts Research Institute, University of Western Ontario.,Current address: University of Toronto
| | | | - Xue Fan Wang
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario
| | - Mathilde Henry
- Axe Neurosciences, CRCHU de Quebec-Université Laval.,Current address: INRAE, Univ. Bordeaux, Bordeaux INP, Nutrineuro, UMR 1286, Bordeaux, F-33000, France
| | - Eric S Kuebler
- Robarts Research Institute, University of Western Ontario
| | | | - Julio Martinez-Trujillo
- Robarts Research Institute, University of Western Ontario.,Neuroscience Program, University of Western Ontario.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario
| | - Marie-Eve Tremblay
- Axe Neurosciences, CRCHU de Quebec-Université Laval.,Département de médecine moléculaire, Université Laval.,Division of Medical Sciences, University of Victoria
| | - Wataru Inoue
- Robarts Research Institute, University of Western Ontario.,Neuroscience Program, University of Western Ontario.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario
| |
Collapse
|
30
|
Mitf Links Neuronal Activity and Long-Term Homeostatic Intrinsic Plasticity. eNeuro 2020; 7:ENEURO.0412-19.2020. [PMID: 32193365 PMCID: PMC7174873 DOI: 10.1523/eneuro.0412-19.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 02/07/2020] [Accepted: 03/02/2020] [Indexed: 12/25/2022] Open
Abstract
Neuroplasticity forms the basis for neuronal circuit complexity and differences between otherwise similar circuits. We show that the microphthalmia-associated transcription factor (Mitf) plays a central role in intrinsic plasticity of olfactory bulb (OB) projection neurons. Mitral and tufted (M/T) neurons from Mitf mutant mice are hyperexcitable, have a reduced A-type potassium current (IA) and exhibit reduced expression of Kcnd3, which encodes a potassium voltage-gated channel subunit (Kv4.3) important for generating the IA Furthermore, expression of the Mitf and Kcnd3 genes is activity dependent in OB projection neurons and the MITF protein activates expression from Kcnd3 regulatory elements. Moreover, Mitf mutant mice have changes in olfactory habituation and have increased habituation for an odorant following long-term exposure, indicating that regulation of Kcnd3 is pivotal for long-term olfactory adaptation. Our findings show that Mitf acts as a direct regulator of intrinsic homeostatic feedback and links neuronal activity, transcriptional changes and neuronal function.
Collapse
|
31
|
Brebner LS, Ziminski JJ, Margetts-Smith G, Sieburg MC, Hall CN, Heintz TG, Lagnado L, Hirrlinger J, Crombag HS, Koya E. Extinction of cue-evoked food-seeking recruits a GABAergic interneuron ensemble in the dorsal medial prefrontal cortex of mice. Eur J Neurosci 2020; 52:3723-3737. [PMID: 32307758 DOI: 10.1111/ejn.14754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/25/2020] [Accepted: 04/10/2020] [Indexed: 11/27/2022]
Abstract
Animals must quickly adapt food-seeking strategies to locate nutrient sources in dynamically changing environments. Learned associations between food and environmental cues that predict its availability promote food-seeking behaviors. However, when such cues cease to predict food availability, animals undergo "extinction" learning, resulting in the inhibition of food-seeking responses. Repeatedly activated sets of neurons, or "neuronal ensembles," in the dorsal medial prefrontal cortex (dmPFC) are recruited following appetitive conditioning and undergo physiological adaptations thought to encode cue-reward associations. However, little is known about how the recruitment and intrinsic excitability of such dmPFC ensembles are modulated by extinction learning. Here, we used in vivo 2-Photon imaging in male Fos-GFP mice that express green fluorescent protein (GFP) in recently behaviorally activated neurons to determine the recruitment of activated pyramidal and GABAergic interneuron dmPFC ensembles during extinction. During extinction, we revealed a persistent activation of a subset of interneurons which emerged from a wider population of interneurons activated during the initial extinction session. This activation pattern was not observed in pyramidal cells, and extinction learning did not modulate the excitability properties of activated pyramidal cells. Moreover, extinction learning reduced the likelihood of reactivation of pyramidal cells activated during the initial extinction session. Our findings illuminate novel neuronal activation patterns in the dmPFC underlying extinction of food-seeking, and in particular, highlight an important role for interneuron ensembles in this inhibitory form of learning.
Collapse
Affiliation(s)
- Leonie S Brebner
- Sussex Neuroscience, School of Psychology, University of Sussex, Falmer, UK
| | - Joseph J Ziminski
- Sussex Neuroscience, School of Psychology, University of Sussex, Falmer, UK
| | | | - Meike C Sieburg
- Sussex Neuroscience, School of Psychology, University of Sussex, Falmer, UK
| | - Catherine N Hall
- Sussex Neuroscience, School of Psychology, University of Sussex, Falmer, UK
| | - Tristan G Heintz
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Falmer, UK
| | - Leon Lagnado
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Falmer, UK
| | - Johannes Hirrlinger
- Carl-Ludwig-Institute for Physiology, University of Leipzig, Leipzig, Germany.,Department of Neurogenetics, Max-Planck-Institute for Experimental Medicine, Göttingen, Germany
| | - Hans S Crombag
- Sussex Neuroscience, School of Psychology, University of Sussex, Falmer, UK
| | - Eisuke Koya
- Sussex Neuroscience, School of Psychology, University of Sussex, Falmer, UK
| |
Collapse
|
32
|
Lu H, Xiao W, Deng S, Cheng X, Zheng H, Chen J, Wang F. Activation of AMPK-dependent autophagy in the nucleus accumbens opposes cocaine-induced behaviors of mice. Addict Biol 2020; 25:e12736. [PMID: 30788886 DOI: 10.1111/adb.12736] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 01/26/2019] [Accepted: 01/28/2019] [Indexed: 12/25/2022]
Abstract
Cocaine is a strong central nervous system stimulant, which can induce drug addiction. Previous studies have reported that cocaine-induced autophagy is involved in neuroinflammation and cell death. However, the role of autophagy in psychomotor sensitivity to cocaine has not been explored. Here, we reported that D1 receptor -CaMKII-AMPK-FoxO3a signaling pathway was involved in acute cocaine application-induced autophagy in the nucleus accumbens (NAc) both in vitro and in vivo. Furthermore, we found that knockdown of the ATG5 gene in the NAc augmented behavioral response to cocaine, and induction of autophagy in the NAc with rapamycin attenuated cocaine-induced behavioral response, which was coincident with the alterations of dendritic spine density in neurons of NAc. These results suggest that cocaine exposure leads to the induction of autophagy, which is a protective mechanism against behavioral response to cocaine of male mice.
Collapse
Affiliation(s)
- Hai‐Feng Lu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan 430030 China
| | - Wen Xiao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan 430030 China
| | - Si‐Long Deng
- Department of Pharmacology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan 430030 China
| | - Xiao‐Ling Cheng
- Department of Pharmacology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan 430030 China
| | - Hui‐Ling Zheng
- Department of Pharmacology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan 430030 China
| | - Jian‐Guo Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan 430030 China
- The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China Wuhan 430030 China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation (HUST) Wuhan 430030 China
- Laboratory of Neuropsychiatric Diseases, The Institute of Brain ResearchHuazhong University of Science and Technology Wuhan 430030 China
- The Collaborative‐Innovation Center for Brain Science Wuhan 430030 China
| | - Fang Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan 430030 China
- The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China Wuhan 430030 China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation (HUST) Wuhan 430030 China
- Laboratory of Neuropsychiatric Diseases, The Institute of Brain ResearchHuazhong University of Science and Technology Wuhan 430030 China
- The Collaborative‐Innovation Center for Brain Science Wuhan 430030 China
| |
Collapse
|
33
|
Delint-Ramirez I, Garcia-Oscos F, Segev A, Kourrich S. Cocaine engages a non-canonical, dopamine-independent, mechanism that controls neuronal excitability in the nucleus accumbens. Mol Psychiatry 2020; 25:680-691. [PMID: 29880884 PMCID: PMC7042730 DOI: 10.1038/s41380-018-0092-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 04/03/2018] [Accepted: 04/13/2018] [Indexed: 11/25/2022]
Abstract
Drug-induced enhanced dopamine (DA) signaling in the brain is a canonical mechanism that initiates addiction processes. However, indirect evidence suggests that cocaine also triggers non-canonical, DA-independent, mechanisms that contribute to behavioral responses to cocaine, including psychomotor sensitization and cocaine self-administration. Identifying these mechanisms and determining how they are initiated is fundamental to further our understanding of addiction processes. Using physiologically relevant in vitro tractable models, we found that cocaine-induced hypoactivity of nucleus accumbens shell (NAcSh) medium spiny neurons (MSNs), one hallmark of cocaine addiction, is independent of DA signaling. Combining brain slice studies and site-directed mutagenesis in HEK293T cells, we found that cocaine binding to intracellular sigma-1 receptor (σ1) initiates this mechanism. Subsequently, σ1 binds to Kv1.2 potassium channels, followed by accumulation of Kv1.2 in the plasma membrane, thereby depressing NAcSh MSNs firing. This mechanism is specific to D1 receptor-expressing MSNs. Our study uncovers a mechanism for cocaine that bypasses DA signaling and leads to addiction-relevant neuroadaptations, thereby providing combinatorial strategies for treating stimulant abuse.
Collapse
Affiliation(s)
- Ilse Delint-Ramirez
- 0000 0000 9482 7121grid.267313.2Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Francisco Garcia-Oscos
- 0000 0000 9482 7121grid.267313.2Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Amir Segev
- 0000 0000 9482 7121grid.267313.2Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Saïd Kourrich
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
34
|
McDevitt DS, Jonik B, Graziane NM. Morphine Differentially Alters the Synaptic and Intrinsic Properties of D1R- and D2R-Expressing Medium Spiny Neurons in the Nucleus Accumbens. Front Synaptic Neurosci 2019; 11:35. [PMID: 31920618 PMCID: PMC6932971 DOI: 10.3389/fnsyn.2019.00035] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 12/06/2019] [Indexed: 12/31/2022] Open
Abstract
Exposure to opioids reshapes future reward and motivated behaviors partially by altering the functional output of medium spiny neurons (MSNs) in the nucleus accumbens shell. Here, we investigated how morphine, a highly addictive opioid, alters synaptic transmission and intrinsic excitability on dopamine D1-receptor (D1R) expressing and dopamine D2-receptor (D2R) expressing MSNs, the two main output neurons in the nucleus accumbens shell. Using whole-cell electrophysiology recordings, we show, that 24 h abstinence following repeated non-contingent administration of morphine (10 mg/kg, i.p.) in mice reduces the miniature excitatory postsynaptic current (mEPSC) frequency and miniature inhibitory postsynaptic current (mIPSC) frequency on D2R-MSNs, with concomitant increases in D2R-MSN intrinsic membrane excitability. We did not observe any changes in synaptic or intrinsic changes on D1R-MSNs. Last, in an attempt to determine the integrated effect of the synaptic and intrinsic alterations on the overall functional output of D2R-MSNs, we measured the input-output efficacy by measuring synaptically-driven action potential firing. We found that both D1R-MSN and D2R-MSN output was unchanged following morphine treatment.
Collapse
Affiliation(s)
- Dillon S McDevitt
- Departments of Anesthesiology and Perioperative Medicine, and Pharmacology, Penn State College of Medicine, Hershey, PA, United States.,Neuroscience Graduate Program, Penn State College of Medicine, Hershey, PA, United States
| | - Benjamin Jonik
- Medical Student Research Program, Penn State College of Medicine, Hershey, PA, United States
| | - Nicholas M Graziane
- Departments of Anesthesiology and Perioperative Medicine, and Pharmacology, Penn State College of Medicine, Hershey, PA, United States
| |
Collapse
|
35
|
Soriani O, Kourrich S. The Sigma-1 Receptor: When Adaptive Regulation of Cell Electrical Activity Contributes to Stimulant Addiction and Cancer. Front Neurosci 2019; 13:1186. [PMID: 31780884 PMCID: PMC6861184 DOI: 10.3389/fnins.2019.01186] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 10/21/2019] [Indexed: 12/17/2022] Open
Abstract
The sigma-1 receptor (σ1R) is an endoplasmic reticulum (ER)-resident chaperone protein that acts like an inter-organelle signaling modulator. Among its several functions such as ER lipid metabolisms/transports and indirect regulation of genes transcription, one of its most intriguing feature is the ability to regulate the function and trafficking of a variety of functional proteins. To date, and directly relevant to the present review, σ1R has been found to regulate both voltage-gated ion channels (VGICs) belonging to distinct superfamilies (i.e., sodium, Na+; potassium, K+; and calcium, Ca2+ channels) and non-voltage-gated ion channels. This regulatory function endows σ1R with a powerful capability to fine tune cells’ electrical activity and calcium homeostasis—a regulatory power that appears to favor cell survival in pathological contexts such as stroke or neurodegenerative diseases. In this review, we present the current state of knowledge on σ1R’s role in the regulation of cellular electrical activity, and how this seemingly adaptive function can shift cell homeostasis and contribute to the development of very distinct chronic pathologies such as psychostimulant abuse and tumor cell growth in cancers.
Collapse
Affiliation(s)
| | - Saïd Kourrich
- Département des Sciences Biologiques, Université du Québec à Montréal, Montréal, QC, Canada.,Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois, Université du Québec à Montréal, Montréal, QC, Canada.,Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
36
|
Reward Devaluation Attenuates Cue-Evoked Sucrose Seeking and Is Associated with the Elimination of Excitability Differences between Ensemble and Non-ensemble Neurons in the Nucleus Accumbens. eNeuro 2019; 6:ENEURO.0338-19.2019. [PMID: 31699890 PMCID: PMC6905639 DOI: 10.1523/eneuro.0338-19.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 11/21/2022] Open
Abstract
Animals must learn relationships between foods and the environmental cues that predict their availability for survival. Such cue–food associations are encoded in sparse sets of neurons or “neuronal ensembles” in the nucleus accumbens (NAc). For these ensemble-encoded, cue-controlled appetitive responses to remain adaptive, they must allow for their dynamic updating depending on acute changes in internal states such as physiological hunger or the perceived desirability of food. However, how these neuronal ensembles are recruited and physiologically modified following the update of such learned associations is unclear. To investigate this, we examined the effects of devaluation on ensemble plasticity at the levels of recruitment, intrinsic excitability, and synaptic physiology in sucrose-conditioned Fos-GFP mice that express green fluorescent protein (GFP) in recently activated neurons. Neuronal ensemble activation patterns and their physiology were examined using immunohistochemistry and slice electrophysiology, respectively. Reward-specific devaluation following 4 d of ad libitum sucrose consumption, but not general caloric devaluation, attenuated cue-evoked sucrose seeking. This suggests that changes in the hedonic and/or incentive value of sucrose, and not caloric need, drove this behavior. Moreover, devaluation attenuated the size of the neuronal ensemble recruited by the cue in the NAc shell. Finally, it eliminated the relative enhanced excitability of ensemble (GFP+) neurons against non-ensemble (GFP−) neurons observed under non-devalued conditions, and did not induce any ensemble-specific changes in excitatory synaptic physiology. Our findings provide new insights into neuronal ensemble mechanisms that underlie the changes in the incentive and/or hedonic impact of cues that support adaptive food seeking.
Collapse
|
37
|
Klorig DC, Alberto GE, Smith T, Godwin DW. Optogenetically-Induced Population Discharge Threshold as a Sensitive Measure of Network Excitability. eNeuro 2019; 6:ENEURO.0229-18.2019. [PMID: 31619450 PMCID: PMC6838688 DOI: 10.1523/eneuro.0229-18.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/06/2019] [Accepted: 09/27/2019] [Indexed: 12/21/2022] Open
Abstract
Network excitability is governed by synaptic efficacy, intrinsic excitability, and the circuitry in which these factors are expressed. The complex interplay between these factors determines how circuits function and, at the extreme, their susceptibility to seizure. We have developed a sensitive, quantitative estimate of network excitability in freely behaving mice using a novel optogenetic intensity-response procedure. Synchronous activation of deep sublayer CA1 pyramidal cells produces abnormal network-wide epileptiform population discharges (PDs) that are nearly indistinguishable from spontaneously-occurring interictal spikes (IISs). By systematically varying light intensity, and therefore the magnitude of the optogenetically-mediated current, we generated intensity-response curves using the probability of PD as the dependent variable. Manipulations known to increase excitability, such as sub-convulsive doses (20 mg/kg) of the chemoconvulsant pentylenetetrazol (PTZ), produced a leftward shift in the curve compared to baseline. The anti-epileptic drug levetiracetam (LEV; 40 mk/kg), in combination with PTZ, produced a rightward shift. Optogenetically-induced PD threshold (oPDT) baselines were stable over time, suggesting the metric is appropriate for within-subject experimental designs with multiple pharmacological manipulations.
Collapse
Affiliation(s)
- D C Klorig
- Department of Neurobiology and Anatomy
- Neuroscience Program
| | - G E Alberto
- Department of Neurobiology and Anatomy
- Neuroscience Program
| | - T Smith
- Department of Neurobiology and Anatomy
| | - D W Godwin
- Department of Neurobiology and Anatomy
- Neuroscience Program
- Department of Physiology and Pharmacology, Wake Forest University, Winston-Salem, NC
| |
Collapse
|
38
|
Bimpisidis Z, Wallén-Mackenzie Å. Neurocircuitry of Reward and Addiction: Potential Impact of Dopamine-Glutamate Co-release as Future Target in Substance Use Disorder. J Clin Med 2019; 8:E1887. [PMID: 31698743 PMCID: PMC6912639 DOI: 10.3390/jcm8111887] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 10/29/2019] [Accepted: 11/01/2019] [Indexed: 12/21/2022] Open
Abstract
Dopamine-glutamate co-release is a unique property of midbrain neurons primarily located in the ventral tegmental area (VTA). Dopamine neurons of the VTA are important for behavioral regulation in response to rewarding substances, including natural rewards and addictive drugs. The impact of glutamate co-release on behaviors regulated by VTA dopamine neurons has been challenging to probe due to lack of selective methodology. However, several studies implementing conditional knockout and optogenetics technologies in transgenic mice have during the past decade pointed towards a role for glutamate co-release in multiple physiological and behavioral processes of importance to substance use and abuse. In this review, we discuss these studies to highlight findings that may be critical when considering mechanisms of importance for prevention and treatment of substance abuse.
Collapse
|
39
|
Ethanol-induced conditioned place preference and aversion differentially alter plasticity in the bed nucleus of stria terminalis. Neuropsychopharmacology 2019; 44:1843-1854. [PMID: 30795004 PMCID: PMC6785142 DOI: 10.1038/s41386-019-0349-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 02/14/2019] [Accepted: 02/15/2019] [Indexed: 01/11/2023]
Abstract
Contextual cues associated with drugs of abuse, such as ethanol, can trigger craving and drug-seeking behavior. Pavlovian procedures, such as place conditioning, have been widely used to study the rewarding/aversive properties of drugs and the association between environmental cues and drug seeking. Previous research has shown that ethanol as an unconditioned stimulus can induce a strong conditioned place preference (CPP) or aversion (CPA) in rodents. However, the neural mechanisms underlying ethanol-induced reward and aversion have not been thoroughly investigated. The bed nucleus of the stria terminalis (BNST), an integral part of the extended amygdala, is engaged by both rewarding and aversive stimuli and plays a role in ethanol-seeking behavior. Here, we used ex-vivo slice physiology to probe learning-induced synaptic plasticity in the BNST following ethanol-induced CPP and CPA. Male DBA/2 J mice (2-3 months old) were conditioned using previously reported ethanol-induced CPP/CPA procedures. Ethanol-induced CPP was associated with increased neuronal excitability in the ventral BNST (vBNST). Conversely, ethanol-induced CPA resulted in a significant decrease in spontaneous glutamatergic transmission without alterations in GABAergic signaling. Ethanol-CPA also led to a significant increase in the paired-pulse ratio at excitatory synapses, suggestive of a decrease in presynaptic glutamate release. Collectively, these data demonstrate that the vBNST is involved in the modulation of contextual learning associated with both the rewarding and the aversive properties of ethanol in mice.
Collapse
|
40
|
Oginsky MF, Ferrario CR. Eating "junk food" has opposite effects on intrinsic excitability of nucleus accumbens core neurons in obesity-susceptible versus -resistant rats. J Neurophysiol 2019; 122:1264-1273. [PMID: 31365322 DOI: 10.1152/jn.00361.2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The nucleus accumbens (NAc) plays critical roles in motivated behaviors, including food seeking and feeding. Differences in NAc function contribute to overeating that drives obesity, but the underlying mechanisms are poorly understood. In addition, there is a fair degree of variation in individual susceptibility versus resistance to obesity that is due in part to differences in NAc function. For example, using selectively bred obesity-prone and obesity-resistant rats, we have found that excitability of medium spiny neurons (MSNs) within the NAc core is enhanced in obesity-prone versus -resistant populations, before any diet manipulation. However, it is unknown whether consumption of sugary, fatty "junk food" alters MSN excitability. Here whole cell patch-clamp recordings were conducted to examine MSN intrinsic excitability in adult male obesity-prone and obesity-resistant rats with and without exposure to a sugary, fatty junk food diet. We replicated our initial finding that basal excitability is enhanced in obesity-prone versus obesity-resistant rats and determined that this is due to a lower fast transient potassium current (IA) in prone versus resistant groups. In addition, the junk food diet had opposite effects on excitability in obesity-prone versus obesity-resistant rats. Specifically, junk food enhanced excitability in MSNs of obesity-resistant rats; this was mediated by a reduction in IA. In contrast, junk food reduced excitability in MSNs from obesity-prone rats; this was mediated by an increase in inward-rectifying potassium current. Thus individual differences in obesity susceptibility influence both basal excitability and how MSN excitability adapts to junk food consumption.NEW & NOTEWORTHY Medium spiny neurons (MSNs) in the nucleus accumbens of obesity-prone rats are hyperexcitable compared with MSNs from obesity-resistant rats. We found that 10 days of "junk food" exposure reduces MSN excitability in obesity-prone rats by increasing inward-rectifying potassium current and increases MSN excitability in obesity-resistant rats by decreasing fast transient potassium current. These data show that there are basal and junk food diet-induced differences in MSN excitability in obesity-prone and obesity-resistant individuals; this may contribute to previously observed differences in incentive motivation.
Collapse
Affiliation(s)
- Max F Oginsky
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| | - Carrie R Ferrario
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
41
|
Teal LB, Gould RW, Felts AS, Jones CK. Selective allosteric modulation of muscarinic acetylcholine receptors for the treatment of schizophrenia and substance use disorders. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2019; 86:153-196. [PMID: 31378251 DOI: 10.1016/bs.apha.2019.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Muscarinic acetylcholine receptor (mAChRs) subtypes represent exciting new targets for the treatment of schizophrenia and substance use disorder (SUD). Recent advances in the development of subtype-selective allosteric modulators have revealed promising effects in preclinical models targeting the different symptoms observed in schizophrenia and SUD. M1 PAMs display potential for addressing the negative and cognitive symptoms of schizophrenia, while M4 PAMs exhibit promise in treating preclinical models predictive of antipsychotic-like activity. In SUD, there is increasing support for modulation of mesocorticolimbic dopaminergic circuitry involved in SUD with selective M4 mAChR PAMs or M5 mAChR NAMs. Allosteric modulators of these mAChR subtypes have demonstrated efficacy in rodent models of cocaine and ethanol seeking, with indications that these ligand may also be useful for other substances of abuse, as well as in various stages in the cycle of addiction. Importantly, allosteric modulators of the different mAChR subtypes may provide viable treatment options, while conferring greater subtype specificity and corresponding enhanced therapeutic index than orthosteric muscarinic ligands and maintaining endogenous temporo-spatial ACh signaling. Overall, subtype specific mAChR allosteric modulators represent important novel therapeutic mechanisms for schizophrenia and SUD.
Collapse
Affiliation(s)
- Laura B Teal
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States
| | - Robert W Gould
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States
| | - Andrew S Felts
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States
| | - Carrie K Jones
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States.
| |
Collapse
|
42
|
Santin JM, Schulz DJ. Membrane Voltage Is a Direct Feedback Signal That Influences Correlated Ion Channel Expression in Neurons. Curr Biol 2019; 29:1683-1688.e2. [PMID: 31080077 PMCID: PMC6677403 DOI: 10.1016/j.cub.2019.04.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/03/2019] [Accepted: 04/03/2019] [Indexed: 11/18/2022]
Abstract
The number and type of ion channels present in the membrane determines the electrophysiological function of every neuron. In many species, stereotyped output of neurons often persists for years [1], and ion channel dysregulation can change these properties to cause severe neurological disorders [2-4]. Thus, a fundamental question is how do neurons coordinate channel expression to uphold their firing patterns over long timescales [1, 5]? One major hypothesis purports that neurons homeostatically regulate their ongoing activity through mechanisms that link membrane voltage to expression relationships among ion channels [6-10]. However, experimentally establishing this feedback loop for the control of expression relationships has been a challenge: manipulations that aim to test for voltage feedback invariably disrupt trophic signaling from synaptic transmission and neuromodulation in addition to activity [9, 11, 12]. Further, neuronal activity often relies critically on these chemical mediators, obscuring the contribution of voltage activity of the membrane per se in forming the channel relationships that determine neuronal output [6, 13]. To resolve this, we isolated an identifiable neuron in crustaceans and then either kept this neuron silent or used a long-term voltage clamp protocol to artificially maintain activity. We found that physiological voltage activity-independent of all known forms of synaptic and neuromodulatory feedback-maintains most channel mRNA relationships, while metabotropic influences may play a relatively smaller role. Thus, ion channel relationships likely needed to maintain neuronal identity are actively and continually regulated at least in part at the level of channel mRNAs through feedback by membrane voltage.
Collapse
Affiliation(s)
- Joseph M Santin
- University of Missouri, Columbia, Division of Biological Sciences, Columbia, MO 65211, USA; The University of North Carolina at Greensboro, Department of Biology, Greensboro, NC 27402, USA
| | - David J Schulz
- University of Missouri, Columbia, Division of Biological Sciences, Columbia, MO 65211, USA.
| |
Collapse
|
43
|
Chronic unpredictable stress promotes cell-specific plasticity in prefrontal cortex D1 and D2 pyramidal neurons. Neurobiol Stress 2019; 10:100152. [PMID: 30937357 PMCID: PMC6430618 DOI: 10.1016/j.ynstr.2019.100152] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 03/04/2019] [Accepted: 03/04/2019] [Indexed: 11/24/2022] Open
Abstract
Exposure to unpredictable environmental stress is widely recognized as a major determinant for risk and severity in neuropsychiatric disorders such as major depressive disorder, anxiety, schizophrenia, and PTSD. The ability of ostensibly unrelated disorders to give rise to seemingly similar psychiatric phenotypes highlights a need to identify circuit-level concepts that could unify diverse factors under a common pathophysiology. Although difficult to disentangle a causative effect of stress from other factors on medial prefrontal cortex (PFC) dysfunction, a wealth of data from humans and rodents demonstrates that the PFC is a key target of stress. The present study sought to identify a model of chronic unpredictable stress (CUS) which induces affective behaviors in C57BL6J mice and once established, measure stress-related alterations in intrinsic excitability and synaptic regulation of mPFC layer 5/6 pyramidal neurons. Adult male mice received 2 weeks of 'less intense' stress or 2 or 4 weeks of 'more intense' CUS followed by sucrose preference for assessment of anhedonia, elevated plus maze for assessment of anxiety and forced swim test for assessment of depressive-like behaviors. Our findings indicate that more intense CUS exposure results in increased anhedonia, anxiety, and depressive behaviors, while the less intense stress results in no measured behavioral phenotypes. Once a behavioral model was established, mice were euthanized approximately 21 days post-stress for whole-cell patch clamp recordings from layer 5/6 pyramidal neurons in the prelimbic (PrL) and infralimbic (IL) cortices. No significant differences were initially observed in intrinsic cell excitability in either region. However, post-hoc analysis and subsequent confirmation using transgenic mice expressing tdtomato or eGFP under control of dopamine D1-or D2-type receptor showed that D1-expressing pyramidal neurons (D1-PYR) in the PrL exhibit reduced thresholds to fire an action potential (increased excitability) but impaired firing capacity at more depolarized potentials, whereas D2-expressing pyramidal neurons (D2-PYR) showed an overall reduction in excitability and spike firing frequency. Examination of synaptic transmission showed that D1-and D2-PYR exhibit differences in basal excitatory and inhibitory signaling under naïve conditions. In CUS mice, D1-PYR showed increased frequency of both miniature excitatory and inhibitory postsynaptic currents, whereas D2-PYR only showed a reduction in excitatory currents. These findings demonstrate that D1-and D2-PYR subpopulations differentially undergo stress-induced intrinsic and synaptic plasticity that may have functional implications for stress-related pathology, and that these adaptations may reflect unique differences in basal properties regulating output of these cells.
Collapse
|
44
|
Influence of pharmacological and epigenetic factors to suppress neurotrophic factors and enhance neural plasticity in stress and mood disorders. Cogn Neurodyn 2019; 13:219-237. [PMID: 31168328 DOI: 10.1007/s11571-019-09522-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 12/17/2018] [Accepted: 01/16/2019] [Indexed: 02/08/2023] Open
Abstract
Stress-induced major depression and mood disorders are characterized by behavioural abnormalities and psychiatric illness, leading to disability and immature mortality worldwide. Neurobiological mechanisms of stress and mood disorders are discussed considering recent findings, and challenges to enhance pharmacological effects of antidepressant, and mood stabilizers. Pharmacological enhancement of ketamine and scopolamine regulates depression at the molecular level, increasing synaptic plasticity in prefrontal regions. Blood-derived neurotrophic factors facilitate mood-deficit symptoms. Epigenetic factors maintain stress-resilience in hippocampal region. Regulation of neurotrophic factors blockades stress, and enhances neuronal survival though it paralyzes limbic regions. Molecular agents and neurotrophic factors also control behavioral and synaptic plasticity in addiction and stress disorders. Future research on neuronal dynamics and cellular actions can be directed to obtain the etiology of synaptic dysregulation in mood disorder and stress. For the first time, the current review contributes to the literature of synaptic plasticity representing the role of epigenetic mechanisms and glucocorticoid receptors to predict depression and anxiety in clinical conditions.
Collapse
|
45
|
Repetitive transcranial magnetic stimulation: Re-wiring the alcoholic human brain. Alcohol 2019; 74:113-124. [PMID: 30420113 DOI: 10.1016/j.alcohol.2018.05.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 05/15/2018] [Accepted: 05/28/2018] [Indexed: 12/28/2022]
Abstract
Alcohol use disorders (AUDs) are one of the leading causes of mortality and morbidity worldwide. In spite of significant advances in understanding the neural underpinnings of AUDs, therapeutic options remain limited. Recent studies have highlighted the potential of repetitive transcranial magnetic stimulation (rTMS) as an innovative, safe, and cost-effective treatment for AUDs. Here, we summarize the fundamental principles of rTMS and its putative mechanisms of action via neurocircuitries related to alcohol addiction. We will also discuss advantages and limitations of rTMS, and argue that Hebbian plasticity and connectivity changes, as well as state-dependency, play a role in shaping some of the long-term effects of rTMS. Visual imaging studies will be linked to recent clinical pilot studies describing the effect of rTMS on alcohol craving and intake, pinpointing new advances, and highlighting conceptual gaps to be filled by future controlled studies.
Collapse
|
46
|
Exposure to conditions of uncertainty promotes the pursuit of amphetamine. Neuropsychopharmacology 2019; 44:274-280. [PMID: 29875447 PMCID: PMC6300556 DOI: 10.1038/s41386-018-0099-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 04/19/2018] [Accepted: 05/16/2018] [Indexed: 12/11/2022]
Abstract
Prior exposure to abused drugs leads to long-lasting neuroadaptations culminating in excessive drug intake. Given the comorbidity between substance use and gambling disorders, surprisingly little is known about the effects of exposure to reinforcement contingencies experienced during games of chance. As it is a central feature of these games, we characterized the effects of exposure to uncertainty on biochemical and behavioral effects normally observed in rats exposed to amphetamine. Rats in different groups were trained to nose-poke for saccharin under certain [fixed-ratio (FR)] or uncertain conditions [variable-ratio (VR)] for 55 1-h sessions. Ratios were escalated on successive sessions and rats maintained on the last ratio (FR/VR 20) for 20-25 days. Two to three weeks later, rats were tested for their locomotor or nucleus accumbens dopamine (NAcc DA) response to amphetamine or self-administration of the drug using a lever press operant. NAcc DA overflow was also assessed in additional rats during the saccharin sessions. Rats exposed to uncertainty subsequently showed a higher locomotor and NAcc DA response to amphetamine and self-administered more drug infusions relative to rats exposed to predictable reinforcement. NAcc DA levels during the saccharin sessions tracked the variance of the scheduled ratios (a measure of uncertainty). VR rats showed escalating DA overflow with increasing ratios. Exposure to uncertainty triggered neuroadaptations similar to those produced by exposure to abused drugs. As these were produced in drug naive rats both during and after exposure to uncertainty, they provide a novel common pathway to drug and behavioral addictions.
Collapse
|
47
|
Beayno A, El Hayek S, Noufi P, Tarabay Y, Shamseddeen W. The Role of Epigenetics in Addiction: Clinical Overview and Recent Updates. Methods Mol Biol 2019; 2011:609-631. [PMID: 31273724 DOI: 10.1007/978-1-4939-9554-7_35] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Addiction is an international public health problem. It is a polygenic disorder best understood by accounting for the interplay between genetic and environmental factors. A recent way of perceiving this interaction is through epigenetics, which help grasp the neurobiological changes that occur in addiction and explain its relapsing-remitting nature. It is now known that every cell has a different way of expressing its phenotype, despite a universal DNA sequence. This is particularly true in the central nervous system where environmental factors influence this expression. Three major epigenetic processes have been found to participate in the perpetuation of addiction by changing the state of the chromatin and the degree of gene transcription: histone acetylation and methylation, DNA methylation, and noncoding RNAs. In the animal model literature, substantial evidence exists about the role of these epigenetic changes in the different phases of substance use disorders. This book chapter is a non-systematic literature review of the recent publications tackling the topic of epigenetics in addiction. Even though this evidence remains scarce and relatively poorly systematized, it is a promising foundation for future research of molecules that target specific brain regions and their functions to address core behavioral changes seen in addiction.
Collapse
Affiliation(s)
- Antoine Beayno
- Department of Psychiatry, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Samer El Hayek
- Department of Psychiatry, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Paul Noufi
- Department of Psychiatry, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Yara Tarabay
- Faculty of Pedagogy, Lebanese University, New Rawda, Lebanon.,Faculty of Natural and Applied Sciences, Notre Dame University, Louaize, Lebanon
| | - Wael Shamseddeen
- Department of Psychiatry, Faculty of Medicine, American University of Beirut, Beirut, Lebanon. .,Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
48
|
Lin J, Guha S, Ramanathan S. Vanadium Dioxide Circuits Emulate Neurological Disorders. Front Neurosci 2018; 12:856. [PMID: 30555289 PMCID: PMC6284030 DOI: 10.3389/fnins.2018.00856] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 11/01/2018] [Indexed: 12/12/2022] Open
Abstract
Information in the central nervous system (CNS) is conducted via electrical signals known as action potentials and is encoded in time. Several neurological disorders including depression, Attention Deficit Hyperactivity Disorder (ADHD), originate in faulty brain signaling frequencies. Here, we present a Hodgkin-Huxley model analog for a strongly correlated VO2 artificial neuron system that undergoes an electrically-driven insulator-metal transition. We demonstrate that tuning of the insulating phase resistance in VO2 threshold switch circuits can enable direct mimicry of neuronal origins of disorders in the CNS. The results introduce use of circuits based on quantum materials as complementary to model animal studies for neuroscience, especially when precise measurements of local electrical properties or competing parallel paths for conduction in complex neural circuits can be a challenge to identify onset of breakdown or diagnose early symptoms of disease.
Collapse
Affiliation(s)
- Jianqiang Lin
- Center for Nanoscale Materials, Argonne National Laboratory, Lemont, IL, United States.,Institute for Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Supratik Guha
- Center for Nanoscale Materials, Argonne National Laboratory, Lemont, IL, United States.,Institute for Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Shriram Ramanathan
- School of Materials Engineering, Purdue University, West Lafayette, IN, United States.,School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
49
|
Hearing M. Prefrontal-accumbens opioid plasticity: Implications for relapse and dependence. Pharmacol Res 2018; 139:158-165. [PMID: 30465850 DOI: 10.1016/j.phrs.2018.11.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 11/05/2018] [Accepted: 11/07/2018] [Indexed: 01/12/2023]
Abstract
In addiction, an individual's ability to inhibit drug seeking and drug taking is thought to reflect a pathological strengthening of drug-seeking behaviors or impairments in the capacity to control maladaptive behavior. These processes are not mutually exclusive and reflect drug-induced modifications within prefrontal cortical and nucleus accumbens circuits, however unlike psychostimulants such as cocaine, far less is known about the temporal, anatomical, and cellular dynamics of these changes. We discuss what is known regarding opioid-induced adaptations in intrinsic membrane physiology and pre-/postsynaptic neurotransmission in principle pyramidal and medium spiny neurons in the medial prefrontal cortex and nucleus accumbens from electrophysiological studies and explore how circuit specific adaptations may contribute to unique facets of opioid addiction.
Collapse
Affiliation(s)
- Matthew Hearing
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, 53233, USA.
| |
Collapse
|
50
|
Persistent "Sag" in Prefrontal Cortex Function following Adolescent Binge Drinking. J Neurosci 2018; 38:9615-9617. [PMID: 30404940 DOI: 10.1523/jneurosci.1755-18.2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 09/07/2018] [Accepted: 09/12/2018] [Indexed: 11/21/2022] Open
|