1
|
Meyers AM, Gnazzo FG, Barrera ED, Nabatian T, Chan L, Beeler JA. Dietary regulation of silent synapses in the dorsolateral striatum. Neuroscience 2024; 563:43-50. [PMID: 39510440 DOI: 10.1016/j.neuroscience.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 09/29/2024] [Accepted: 11/04/2024] [Indexed: 11/15/2024]
Abstract
Obesity and drugs of abuse share overlapping neural circuits and behaviors. Silent synapses are transient synapses that are important for remodeling brain circuits. They are prevalent during early development but largely disappear by adulthood. Drugs of abuse increase silent synapses during adulthood and may facilitate reorganizing brain circuits around drug-related experience, facilitating addiction and contributing to relapse during treatment and abstinence. Whether obesity causes alterations in the expression of silent synapses in a manner similar to drugs of abuse has not been examined. Using a dietary-induced obesity paradigm, mice that chronically consumed high fat diet (HFD) exhibited increased silent synapses in both direct and indirect pathway medium spiny neurons in the dorsolateral striatum. Both the time of onset of increased silent synapses and their normalization upon discontinuation of HFD occurs on an extended time scale compared to drugs of abuse. These data demonstrate that chronic consumption of HFD, like drugs of abuse, can alter mechanisms of circuit plasticity likely facilitating neural reorganization analogous to drugs of abuse.
Collapse
Affiliation(s)
- Allison M Meyers
- Psychology Program, The Graduate Center, City University of New York, New York, NY, USA
| | - Federico G Gnazzo
- Department of Psychology, Queens College, City University of New York, Flushing, NY, USA
| | - Eddy D Barrera
- Biology Program, The Graduate Center, City University of New York, New York, NY, USA
| | - Tikva Nabatian
- Cognitive Neuroscience MS Program, Graduate Center, City University of New York, New York, NY, USA
| | - Larry Chan
- Department of Psychology, Queens College, City University of New York, Flushing, NY, USA
| | - Jeff A Beeler
- Department of Psychology, Queens College, City University of New York, Flushing, NY, USA; Psychology Program, The Graduate Center, City University of New York, New York, NY, USA; Biology Program, The Graduate Center, City University of New York, New York, NY, USA; Cognitive Neuroscience MS Program, Graduate Center, City University of New York, New York, NY, USA.
| |
Collapse
|
2
|
Li Q, Yu ZP, Li YG, Tang ZH, Hu YF, Wang MJ, Shen HW. Single-nucleus RNA-sequencing of orbitofrontal cortex in rat model of methamphetamine-induced sensitization. Neurosci Lett 2024; 841:137953. [PMID: 39214331 DOI: 10.1016/j.neulet.2024.137953] [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: 06/26/2024] [Revised: 08/15/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
The behavioral sensitization, characterized by escalated behavioral responses triggered by recurrent exposure to psychostimulants, involves neurobiological mechanisms that are brain-region and cell-type specific. Enduring neuroadaptive changes have been observed in response to methamphetamine (METH) within the orbitofrontal cortex (OFC), the cell-type specific transcriptional alterations in response to METH sensitization remain understudied. In this study, we utilized Single-nucleus RNA-sequencing (snRNA-seq) to profile the gene expression changes in the OFC of a rat METH sensitization model. The analyses of differentially expressed genes (DEGs) unveiled cell-type specific transcriptional reactions associated with METH sensitization, with the most significant alterations documented in microglial cells. Bioinformatic investigations revealed that distinct functional and signaling pathways enriched in microglia-specific DEGs majorly involved in macroautophagy processes and the activation of N-methyl-D-aspartate ionotropic glutamate receptors (NMDAR). To validate the translational relevance of our findings, we analyzed our snRNA-seq data in conjunction with a transcriptomic study of individuals with opioid use disorder (OUD) and a large-scale Genome-Wide Association Studies (GWAS) from multiple externalizing phenotypes related to drug addiction. The validation analysis confirmed the consistent expression changes of key microglial DEGs in human METH addiction. Moreover, the integration with GWAS data revealed associations between addiction risk genes and the DEGs observed in specific cell types, particularly microglia and excitatory neurons. Our study highlights the importance of cell-type specific transcriptional alterations in the OFC in the context of METH sensitization and their potential translational relevance to human drug addiction.
Collapse
Affiliation(s)
- Qiong Li
- Department of Pharmacology, Health Science Center, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang 315211, China
| | - Zhi-Peng Yu
- Department of Pharmacology, Health Science Center, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang 315211, China; Faculty of Electrical Engineering and Computer Science, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang 315211, China
| | - Yan-Guo Li
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Zi-Hang Tang
- Department of Pharmacology, Health Science Center, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang 315211, China
| | - Yong-Feng Hu
- Department of Pharmacology, Health Science Center, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang 315211, China
| | - Ma-Jie Wang
- Department of psychiatry, Affiliated Kangning Hospital of Ningbo University, Ningbo, Zhejiang 315201, China
| | - Hao-Wei Shen
- Department of Pharmacology, Health Science Center, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang 315211, China; Department of psychiatry, Affiliated Kangning Hospital of Ningbo University, Ningbo, Zhejiang 315201, China.
| |
Collapse
|
3
|
Meyers AM, Gnazzo FG, Barrera ED, Nabatian T, Chan L, Beeler JA. DIETARY REGULATION OF SILENT SYNAPSES IN THE DORSOLATERAL STRIATUM. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.24.586457. [PMID: 38585967 PMCID: PMC10996560 DOI: 10.1101/2024.03.24.586457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Obesity and drugs of abuse share overlapping neural circuits and behaviors. Silent synapses are transient synapses that are important for remodeling brain circuits. They are prevalent during early development but largely disappear by adulthood. Drugs of abuse increase silent synapses during adulthood and may facilitate reorganizing brain circuits around drug-related experience, facilitating addiction and contributing to relapse during treatment and abstinence. Whether obesity causes alterations in the expression of silent synapses in a manner similar to drugs of abuse has not been examined. Using a dietary-induced obesity paradigm, mice that chronically consumed high fat diet (HFD) exhibited increased silent synapses in both direct and indirect pathway medium spiny neurons in the dorsolateral striatum. Both the time of onset of increased silent synapses and their normalization upon discontinuation of HFD occurs on an extended time scale compared to drugs of abuse. These data demonstrate that chronic consumption of HFD, like drugs of abuse, can alter mechanisms of circuit plasticity likely facilitating neural reorganization analogous to drugs of abuse.
Collapse
Affiliation(s)
- Allison M Meyers
- Psychology Program, The Graduate Center, City University of New York, New York, NY, USA
| | - Federico G Gnazzo
- Department of Psychology, Queens College, City University of New York, Flushing, NY, USA
| | - Eddy D Barrera
- Biology Program, The Graduate Center, City University of New York, New York, NY, USA
| | - Tikva Nabatian
- Cognitive Neuroscience MS program, Graduate Center, City University of New York, New York, NY, USA
| | - Larry Chan
- Department of Psychology, Queens College, City University of New York, Flushing, NY, USA
| | - Jeff A Beeler
- Department of Psychology, Queens College, City University of New York, Flushing, NY, USA
- Psychology Program, The Graduate Center, City University of New York, New York, NY, USA
- Biology Program, The Graduate Center, City University of New York, New York, NY, USA
- Cognitive Neuroscience MS program, Graduate Center, City University of New York, New York, NY, USA
| |
Collapse
|
4
|
Sagarkar S, Bhat N, Rotti D, Subhedar NK. AMPA and NMDA receptors in dentate gyrus mediate memory for sucrose in two port discrimination task. Hippocampus 2024; 34:342-356. [PMID: 38780087 DOI: 10.1002/hipo.23609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 03/30/2024] [Accepted: 04/17/2024] [Indexed: 05/25/2024]
Abstract
Although the phenomenon of memory formation and recall associated with the use of psychotropic drugs has been extensively studied, mechanisms underlying memories for natural reward have not been clarified. Herein, we test the hypothesis that glutamatergic receptors in the dentate gyrus play a role in memories associated with sucrose. We used pellet self-administration protocol to generate memories in two-port nose-poke discrimination task using male Wistar rats. During non-rewarded probe trial, the conditioned animals readily discriminated the active port versus inactive port and showed massive increase in mRNA expression of AMPA receptor subunit genes (gria2, gria3) as well as c-Fos protein in the DG. Access to sweet pellet further enhanced c-Fos expression in the DG. However, animals pre-treated with AMPA receptor antagonist CNQX (intra-DG), on exposure to operant chamber (no pellet), showed decreased discrimination as well as c-Fos expression. We suggest that AMPA receptors in DG mediate recall and consolidation of memories associated with sucrose consumption. CNQX pre-treated animals, if presented with sweet pellet on nose poke, exhibited high discrimination index coupled with increased c-Fos expression. In these CNQX treated rats, the DI was again decreased following administration of NMDA receptor antagonist AP5. We suggest that, although AMPA receptors are blocked, the access to sweet pellet may induce surge of glutamate in the DG, which in turn may reinstate memories via activation of erstwhile silent synapses in NMDA dependant manner.
Collapse
MESH Headings
- Animals
- Male
- Rats
- 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology
- Conditioning, Operant/drug effects
- Conditioning, Operant/physiology
- Dentate Gyrus/drug effects
- Dentate Gyrus/metabolism
- Discrimination Learning/drug effects
- Discrimination Learning/physiology
- Discrimination, Psychological/drug effects
- Discrimination, Psychological/physiology
- Excitatory Amino Acid Antagonists/pharmacology
- Memory/physiology
- Memory/drug effects
- Proto-Oncogene Proteins c-fos/metabolism
- Rats, Wistar
- Receptors, AMPA/metabolism
- Receptors, AMPA/antagonists & inhibitors
- Receptors, N-Methyl-D-Aspartate/metabolism
- Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
- RNA, Messenger/metabolism
- Self Administration
- Sucrose/administration & dosage
Collapse
Affiliation(s)
- Sneha Sagarkar
- Department of Zoology, Savitribai Phule Pune University, Pune, India
| | - Nagashree Bhat
- Department of Zoology, Savitribai Phule Pune University, Pune, India
| | - Deepa Rotti
- Department of Zoology, Savitribai Phule Pune University, Pune, India
| | | |
Collapse
|
5
|
Brida KL, Jorgensen ET, Phillips RA, Newman CE, Tuscher JJ, Morring EK, Zipperly ME, Ianov L, Montgomery KD, Tippani M, Hyde TM, Maynard KR, Martinowich K, Day JJ. Reelin marks cocaine-activated striatal ensembles, promotes neuronal excitability, and regulates cocaine reward. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.599348. [PMID: 38948801 PMCID: PMC11212904 DOI: 10.1101/2024.06.17.599348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Drugs of abuse activate defined neuronal ensembles in brain reward structures such as the nucleus accumbens (NAc), which are thought to promote the enduring synaptic, circuit, and behavioral consequences of drug exposure. While the molecular and cellular effects arising from experience with drugs like cocaine are increasingly well understood, the mechanisms that sculpt NAc ensemble participation are largely unknown. Here, we leveraged unbiased single-nucleus transcriptional profiling to identify expression of the secreted glycoprotein Reelin (encoded by the Reln gene) as a marker of cocaine-activated neuronal ensembles within the rat NAc. Multiplexed in situ detection confirmed selective expression of the immediate early gene Fos in Reln+ neurons after cocaine experience, and also revealed enrichment of Reln mRNA in Drd1 + medium spiny neurons (MSNs) in both the rat and human brain. Using a novel CRISPR interference strategy enabling selective Reln knockdown in the adult NAc, we observed altered expression of genes linked to calcium signaling, emergence of a transcriptional trajectory consistent with loss of cocaine sensitivity, and a striking decrease in MSN intrinsic excitability. At the behavioral level, loss of Reln prevented cocaine locomotor sensitization, abolished cocaine place preference memory, and decreased cocaine self-administration behavior. Together, these results identify Reelin as a critical mechanistic link between ensemble participation and cocaine-induced behavioral adaptations.
Collapse
|
6
|
Pelletier OB, Brunori G, Wang Y, Robishaw JD. Post-transcriptional regulation and subcellular localization of G-protein γ7 subunit: implications for striatal function and behavioral responses to cocaine. Front Neuroanat 2024; 18:1394659. [PMID: 38764487 PMCID: PMC11100332 DOI: 10.3389/fnana.2024.1394659] [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: 03/01/2024] [Accepted: 04/17/2024] [Indexed: 05/21/2024] Open
Abstract
The striatal D1 dopamine receptor (D1R) and A2a adenosine receptor (A2aR) signaling pathways play important roles in drug-related behaviors. These receptors activate the Golf protein comprised of a specific combination of αolfβ2γ7 subunits. During assembly, the γ7 subunit sets the cellular level of the Golf protein. In turn, the amount of Golf protein determines the collective output from both D1R and A2aR signaling pathways. This study shows the Gng7 gene encodes multiple γ7 transcripts differing only in their non-coding regions. In striatum, Transcript 1 is the predominant isoform. Preferentially expressed in the neuropil, Transcript 1 is localized in dendrites where it undergoes post-transcriptional regulation mediated by regulatory elements in its 3' untranslated region that contribute to translational suppression of the γ7 protein. Earlier studies on gene-targeted mice demonstrated loss of γ7 protein disrupts assembly of the Golf protein. In the current study, morphological analysis reveals the loss of the Golf protein is associated with altered dendritic morphology of medium spiny neurons. Finally, behavioral analysis of conditional knockout mice with cell-specific deletion of the γ7 protein in distinct populations of medium spiny neurons reveals differential roles of the Golf protein in mediating behavioral responses to cocaine. Altogether, these findings provide a better understanding of the regulation of γ7 protein expression, its impact on Golf function, and point to a new potential target and mechanisms for treating addiction and related disorders.
Collapse
Affiliation(s)
- Oliver B. Pelletier
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| | - Gloria Brunori
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| | - Yingcai Wang
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| | - Janet D. Robishaw
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
7
|
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
|
8
|
Davidson CJ, Mascarin AT, Yahya MA, Rubio FJ, Gheidi A. Approaches and considerations of studying neuronal ensembles: a brief review. Front Cell Neurosci 2023; 17:1310724. [PMID: 38155864 PMCID: PMC10752959 DOI: 10.3389/fncel.2023.1310724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/27/2023] [Indexed: 12/30/2023] Open
Abstract
First theorized by Hebb, neuronal ensembles have provided a framework for understanding how the mammalian brain operates, especially regarding learning and memory. Neuronal ensembles are discrete, sparsely distributed groups of neurons that become activated in response to a specific stimulus and are thought to provide an internal representation of the world. Beyond the study of region-wide or projection-wide activation, the study of ensembles offers increased specificity and resolution to identify and target specific memories or associations. Neuroscientists interested in the neurobiology of learning, memory, and motivated behavior have used electrophysiological-, calcium-, and protein-based proxies of neuronal activity in preclinical models to better understand the neurobiology of learned and motivated behaviors. Although these three approaches may be used to pursue the same general goal of studying neuronal ensembles, technical differences lead to inconsistencies in the output and interpretation of data. This mini-review highlights some of the methodologies used in electrophysiological-, calcium-, and protein-based studies of neuronal ensembles and discusses their strengths and weaknesses.
Collapse
Affiliation(s)
- Cameron J. Davidson
- William Beaumont School of Medicine, Oakland University, Rochester, MI, United States
| | - Alixandria T. Mascarin
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Majd A. Yahya
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - F. Javier Rubio
- Neuronal Ensembles in Addiction Section, Behavioral Neuroscience Research Branch, Intramural Research Program/National Institute on Drug Abuse/National Institutes of Health, Bethesda, MD, United States
| | - Ali Gheidi
- Department of Biomedical Sciences, Mercer University, Macon, GA, United States
| |
Collapse
|
9
|
Liu X, Wang F, Le Q, Ma L. Cellular and molecular basis of drug addiction: The role of neuronal ensembles in addiction. Curr Opin Neurobiol 2023; 83:102813. [PMID: 37972536 DOI: 10.1016/j.conb.2023.102813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/25/2023] [Accepted: 10/25/2023] [Indexed: 11/19/2023]
Abstract
Addiction has been conceptualized as a disease of learning and memory. Learned associations between environmental cues and unconditioned rewards induced by drug administration, which play a critical role in addiction, have been shown to be encoded in sparsely distributed populations of neurons called neuronal ensembles. This review aims to highlight how synaptic remodeling and alterations in signaling pathways that occur specifically in neuronal ensembles contribute to the pathogenesis of addiction. Furthermore, a causal link between transcriptional and epigenetic modifications in neuronal ensembles and the development of the addictive state is proposed. Translational studies of molecular and cellular changes in neuronal ensembles that contribute to drug-seeking behavior, will allow the identification of molecular and circuit targets and interventions for substance use disorders.
Collapse
Affiliation(s)
- Xing Liu
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, China
| | - Feifei Wang
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, China.
| | - Qiumin Le
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, China
| | - Lan Ma
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, China
| |
Collapse
|
10
|
Dysregulation of AMPA Receptor Trafficking and Intracellular Vesicular Sorting in the Prefrontal Cortex of Dopamine Transporter Knock-Out Rats. Biomolecules 2023; 13:biom13030516. [PMID: 36979451 PMCID: PMC10046215 DOI: 10.3390/biom13030516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/23/2023] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
Dopamine (DA) and glutamate interact, influencing neural excitability and promoting synaptic plasticity. However, little is known regarding the molecular mechanisms underlying this crosstalk. Since perturbation of DA-AMPA receptor interaction might sustain pathological conditions, the major aim of our work was to evaluate the effect of the hyperactive DA system on the AMPA subunit composition, trafficking, and membrane localization in the prefrontal cortex (PFC). Taking advantage of dopamine transporter knock-out (DAT−/−) rats, we found that DA overactivity reduced the translation of cortical AMPA receptors and their localization at both synaptic and extra-synaptic sites through, at least in part, altered intracellular vesicular sorting. Moreover, the reduced expression of AMPA receptor-specific anchoring proteins and structural markers, such as Neuroligin-1 and nCadherin, likely indicate a pattern of synaptic instability. Overall, these data reveal that a condition of hyperdopaminergia markedly alters the homeostatic plasticity of AMPA receptors, suggesting a general destabilization and depotentiation of the AMPA-mediated glutamatergic neurotransmission in the PFC. This effect might be functionally relevant for disorders characterized by elevated dopaminergic activity.
Collapse
|
11
|
miR-34a regulates silent synapse and synaptic plasticity in mature hippocampus. Prog Neurobiol 2023; 222:102404. [PMID: 36642095 DOI: 10.1016/j.pneurobio.2023.102404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/26/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
AMPAR-lacking silent synapses are prevailed and essential for synaptic refinement and synaptic plasticity in developing brains. In mature brain, they are sparse but could be induced under several pathological conditions. How they are regulated molecularly is far from clear. miR-34a is a highly conserved and brain-enriched microRNA with age-dependent upregulated expression profile. Its neuronal function in mature brain remains to be revealed. Here by analyzing synaptic properties of the heterozygous miR-34a knock out mice (34a_ht), we have discovered that mature but not juvenile 34a_ht mice have more silent synapses in the hippocampus accompanied with enhanced synaptic NMDAR but not AMPAR function and increased spine density. As a result, 34a_ht mice display enhanced long-term potentiation (LTP) in the Schaffer collateral synapses and better spatial learning and memory. We further found that Creb1 is a direct target of miR-34a, whose upregulation and activation may mediate the silent synapse increment in 34a_ht mice. Hence, we reveal a novel physiological role of miR-34a in mature brains and provide a molecular mechanism underlying silent synapse regulation.
Collapse
|
12
|
Bijoch Ł, Klos J, Pawłowska M, Wiśniewska J, Legutko D, Szachowicz U, Kaczmarek L, Beroun A. Whole-brain tracking of cocaine and sugar rewards processing. Transl Psychiatry 2023; 13:20. [PMID: 36683039 PMCID: PMC9868126 DOI: 10.1038/s41398-023-02318-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/07/2023] [Accepted: 01/10/2023] [Indexed: 01/24/2023] Open
Abstract
Natural rewards, such as food, and sex are appetitive stimuli available for animals in their natural environment. Similarly, addictive rewards such as drugs of abuse possess strong, positive valence, but their action relies on their pharmacological properties. Nevertheless, it is believed that both of these kinds of rewards activate similar brain circuitry. The present study aimed to discover which parts of the brain process the experience of natural and addictive rewards. To holistically address this question, we used a single-cell whole-brain imaging approach to find patterns of activation for acute and prolonged sucrose and cocaine exposure. We analyzed almost 400 brain structures and created a brain-wide map of specific, c-Fos-positive neurons engaged by these rewards. Acute but not prolonged sucrose exposure triggered a massive c-Fos expression throughout the brain. Cocaine exposure on the other hand potentiated c-Fos expression with prolonged use, engaging more structures than sucrose treatment. The functional connectivity analysis unraveled an increase in brain modularity after the initial exposure to both types of rewards. This modularity was increased after repeated cocaine, but not sucrose, intake. To check whether discrepancies between the processing of both types of rewards can be found on a cellular level, we further studied the nucleus accumbens, one of the most strongly activated brain structures by both sucrose and cocaine experience. We found a high overlap between natural and addictive rewards on the level of c-Fos expression. Electrophysiological measurements of cellular correlates of synaptic plasticity revealed that natural and addictive rewards alike induce the accumulation of silent synapses. These results strengthen the hypothesis that in the nucleus accumbens drugs of abuse cause maladaptive neuronal plasticity in the circuitry that typically processes natural rewards.
Collapse
Affiliation(s)
- Łukasz Bijoch
- grid.419305.a0000 0001 1943 2944Laboratory of Neuronal Plasticity, Nencki-EMBL Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Joanna Klos
- grid.419305.a0000 0001 1943 2944Laboratory of Neuronal Plasticity, Nencki-EMBL Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Monika Pawłowska
- grid.419305.a0000 0001 1943 2944Laboratory of Neurobiology, Nencki-EMBL Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland ,grid.12847.380000 0004 1937 1290Institute of Experimental Physics, Faculty of Physics, University of Warsaw, Warsaw, Poland
| | - Justyna Wiśniewska
- grid.419305.a0000 0001 1943 2944Laboratory of Neuronal Plasticity, Nencki-EMBL Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Diana Legutko
- grid.419305.a0000 0001 1943 2944Laboratory of Neurobiology, Nencki-EMBL Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Urszula Szachowicz
- grid.419305.a0000 0001 1943 2944Laboratory of Neuronal Plasticity, Nencki-EMBL Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Leszek Kaczmarek
- grid.419305.a0000 0001 1943 2944Laboratory of Neurobiology, Nencki-EMBL Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Anna Beroun
- Laboratory of Neuronal Plasticity, Nencki-EMBL Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
13
|
Panopoulou M, Schlüter OM. Ca 2+-permeable AMPA receptors set the threshold for retrieval of drug memories. Mol Psychiatry 2022; 27:2868-2878. [PMID: 35296806 DOI: 10.1038/s41380-022-01505-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 02/13/2022] [Accepted: 02/22/2022] [Indexed: 11/09/2022]
Abstract
Frequent relapse prevents the successful treatment of substance use disorders and is triggered in part by retrieval of drug-associated memories. Drug-conditioned behaviours in rodents are reinstated upon drug memory retrieval following re-exposure to cues previously associated with the drug, or the drug itself. Therapies based on mechanistic insights from rodent studies have focused on amnesic procedures of cue-drug associations but with so far limited success. Conversely, more recent studies propose that inhibiting drug memory retrieval offers improved anti-relapse efficacy. However, mechanisms of memory retrieval are poorly understood. Here, we used a conditioned place preference (CPP) procedure in mice to investigate the cellular and molecular underpinnings of drug-induced memory retrieval. After extinction training of CPP, Ca2+-permeable AMPA receptors (CP-AMPARs) accumulated at drug-generated silent synapses of nucleus accumbens (NAc) medium spiny neurons. The NAc CP-AMPARs regulated the retrieval mechanism of drug memories after extinction. Specifically, we used different priming doses of cocaine, fentanyl, or a cue associated with drug exposure to reinstate CPP, providing different memory retrieval conditions. Although both high and low doses of these two drugs induced CPP reinstatement, compromising CP-AMPAR accumulation impaired CPP reinstatement, induced by low doses of each drug or the cue. This threshold effect was mediated by NAc CP-AMPARs as region specific knock-down of PSD-95 prevented low-dose cocaine-induced retrieval selectively. These results demonstrate the NAc as a brain region and CP-AMPARs as key synaptic substrates that govern the threshold for drug-induced retrieval and behavioural expression of drug memories.
Collapse
Affiliation(s)
- Myrto Panopoulou
- Department of Psychiatry and Psychotherapy, University Medical Center, D-37075, Göttingen, Germany.,International Max Planck Research School for Neurosciences, D-37077, Göttingen, Germany
| | - Oliver M Schlüter
- Department of Psychiatry and Psychotherapy, University Medical Center, D-37075, Göttingen, Germany. .,Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
14
|
Ortega-de San Luis C, Ryan TJ. Understanding the physical basis of memory: Molecular mechanisms of the engram. J Biol Chem 2022; 298:101866. [PMID: 35346687 PMCID: PMC9065729 DOI: 10.1016/j.jbc.2022.101866] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 03/08/2022] [Accepted: 03/11/2022] [Indexed: 12/18/2022] Open
Abstract
Memory, defined as the storage and use of learned information in the brain, is necessary to modulate behavior and critical for animals to adapt to their environments and survive. Despite being a cornerstone of brain function, questions surrounding the molecular and cellular mechanisms of how information is encoded, stored, and recalled remain largely unanswered. One widely held theory is that an engram is formed by a group of neurons that are active during learning, which undergoes biochemical and physical changes to store information in a stable state, and that are later reactivated during recall of the memory. In the past decade, the development of engram labeling methodologies has proven useful to investigate the biology of memory at the molecular and cellular levels. Engram technology allows the study of individual memories associated with particular experiences and their evolution over time, with enough experimental resolution to discriminate between different memory processes: learning (encoding), consolidation (the passage from short-term to long-term memories), and storage (the maintenance of memory in the brain). Here, we review the current understanding of memory formation at a molecular and cellular level by focusing on insights provided using engram technology.
Collapse
Affiliation(s)
- Clara Ortega-de San Luis
- School of Biochemistry and Immunology and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland.
| | - Tomás J Ryan
- School of Biochemistry and Immunology and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland; Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia; Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Ontario, Canada.
| |
Collapse
|
15
|
Allichon MC, Ortiz V, Pousinha P, Andrianarivelo A, Petitbon A, Heck N, Trifilieff P, Barik J, Vanhoutte P. Cell-Type-Specific Adaptions in Striatal Medium-Sized Spiny Neurons and Their Roles in Behavioral Responses to Drugs of Abuse. Front Synaptic Neurosci 2022; 13:799274. [PMID: 34970134 PMCID: PMC8712310 DOI: 10.3389/fnsyn.2021.799274] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/26/2021] [Indexed: 12/21/2022] Open
Abstract
Drug addiction is defined as a compulsive pattern of drug-seeking- and taking- behavior, with recurrent episodes of abstinence and relapse, and a loss of control despite negative consequences. Addictive drugs promote reinforcement by increasing dopamine in the mesocorticolimbic system, which alters excitatory glutamate transmission within the reward circuitry, thereby hijacking reward processing. Within the reward circuitry, the striatum is a key target structure of drugs of abuse since it is at the crossroad of converging glutamate inputs from limbic, thalamic and cortical regions, encoding components of drug-associated stimuli and environment, and dopamine that mediates reward prediction error and incentive values. These signals are integrated by medium-sized spiny neurons (MSN), which receive glutamate and dopamine axons converging onto their dendritic spines. MSN primarily form two mostly distinct populations based on the expression of either DA-D1 (D1R) or DA-D2 (D2R) receptors. While a classical view is that the two MSN populations act in parallel, playing antagonistic functional roles, the picture seems much more complex. Herein, we review recent studies, based on the use of cell-type-specific manipulations, demonstrating that dopamine differentially modulates dendritic spine density and synapse formation, as well as glutamate transmission, at specific inputs projecting onto D1R-MSN and D2R-MSN to shape persistent pathological behavioral in response to drugs of abuse. We also discuss the identification of distinct molecular events underlying the detrimental interplay between dopamine and glutamate signaling in D1R-MSN and D2R-MSN and highlight the relevance of such cell-type-specific molecular studies for the development of innovative strategies with potential therapeutic value for addiction. Because drug addiction is highly prevalent in patients with other psychiatric disorders when compared to the general population, we last discuss the hypothesis that shared cellular and molecular adaptations within common circuits could explain the co-occurrence of addiction and depression. We will therefore conclude this review by examining how the nucleus accumbens (NAc) could constitute a key interface between addiction and depression.
Collapse
Affiliation(s)
- Marie-Charlotte Allichon
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Vanesa Ortiz
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Paula Pousinha
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Andry Andrianarivelo
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Anna Petitbon
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Nicolas Heck
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Pierre Trifilieff
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Jacques Barik
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Peter Vanhoutte
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| |
Collapse
|
16
|
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: 40] [Impact Index Per Article: 20.0] [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
|
17
|
Silent Synapses in Cocaine-Associated Memory and Beyond. J Neurosci 2021; 41:9275-9285. [PMID: 34759051 DOI: 10.1523/jneurosci.1559-21.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/22/2021] [Accepted: 09/27/2021] [Indexed: 11/21/2022] Open
Abstract
Glutamatergic synapses are key cellular sites where cocaine experience creates memory traces that subsequently promote cocaine craving and seeking. In addition to making across-the-board synaptic adaptations, cocaine experience also generates a discrete population of new synapses that selectively encode cocaine memories. These new synapses are glutamatergic synapses that lack functionally stable AMPARs, often referred to as AMPAR-silent synapses or, simply, silent synapses. They are generated de novo in the NAc by cocaine experience. After drug withdrawal, some of these synapses mature by recruiting AMPARs, contributing to the consolidation of cocaine-associated memory. After cue-induced retrieval of cocaine memories, matured silent synapses alternate between two dynamic states (AMPAR-absent vs AMPAR-containing) that correspond with the behavioral manifestations of destabilization and reconsolidation of these memories. Here, we review the molecular mechanisms underlying silent synapse dynamics during behavior, discuss their contributions to circuit remodeling, and analyze their role in cocaine-memory-driven behaviors. We also propose several mechanisms through which silent synapses can form neuronal ensembles as well as cross-region circuit engrams for cocaine-specific behaviors. These perspectives lead to our hypothesis that cocaine-generated silent synapses stand as a distinct set of synaptic substrates encoding key aspects of cocaine memory that drive cocaine relapse.
Collapse
|
18
|
Liu Q, Yu J, Li X, Guo Y, Sun T, Luo L, Ren J, Jiang W, Zhang R, Yang P, Yang Q. Cannabinoid receptor GPR55 activation blocks nicotine use disorder by regulation of AMPAR phosphorylation. Psychopharmacology (Berl) 2021; 238:3335-3346. [PMID: 34648060 DOI: 10.1007/s00213-021-05949-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 08/03/2021] [Indexed: 11/28/2022]
Abstract
RATIONALE Nicotine use disorder can alter synaptic plasticity correlated with learning and memory process. G protein-coupled receptor 55 (GPR55), a novel cannabinoid receptor, which is highly expressed in the central nervous system, plays a prominent role in learning and memory. However, the role of GPR55 in nicotine use disorder remains unclear. METHODS In this study, we used the conditioned place preference (CPP) paradigm, a standard and well-established model for evaluating the rewarding effect of drug abuse, to investigate nicotine use disorder behavior in mice. After behavioral tests, the effect of GPR55 on nicotine response was evaluated using Western blotting, immunofluorescence staining, whole-cell patch-clamp recordings, and ELISA. RESULTS GPR55 activation significantly reduced nicotine-CPP behavior by decreasing the spontaneous excitatory postsynaptic currents frequency in the nucleus accumbens (NAc) and the release of dopamine in serum. Furthermore, we found that the inhibition effects of nicotine response were mediated by phosphorylation of AMPAR. The PI3K-Akt signaling was involved in nicotine-CPP via GPR55 activation. CONCLUSION Our findings showed that GPR55 in the NAc plays a specific role in blocking nicotine-CPP behavior and might be a potential target for the treatment of nicotine use disorder.
Collapse
Affiliation(s)
- Qingqing Liu
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Jiaoyan Yu
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Xi Li
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Yanyan Guo
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Ting Sun
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Li Luo
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Jing Ren
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Wei Jiang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Ruitao Zhang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Peng Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Qi Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China.
| |
Collapse
|
19
|
Rao-Ruiz P, Visser E, Mitrić M, Smit AB, van den Oever MC. A Synaptic Framework for the Persistence of Memory Engrams. Front Synaptic Neurosci 2021; 13:661476. [PMID: 33841124 PMCID: PMC8024575 DOI: 10.3389/fnsyn.2021.661476] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 02/26/2021] [Indexed: 12/31/2022] Open
Abstract
The ability to store and retrieve learned information over prolonged periods of time is an essential and intriguing property of the brain. Insight into the neurobiological mechanisms that underlie memory consolidation is of utmost importance for our understanding of memory persistence and how this is affected in memory disorders. Recent evidence indicates that a given memory is encoded by sparsely distributed neurons that become highly activated during learning, so-called engram cells. Research by us and others confirms the persistent nature of cortical engram cells by showing that these neurons are required for memory expression up to at least 1 month after they were activated during learning. Strengthened synaptic connectivity between engram cells is thought to ensure reactivation of the engram cell network during retrieval. However, given the continuous integration of new information into existing neuronal circuits and the relatively rapid turnover rate of synaptic proteins, it is unclear whether a lasting learning-induced increase in synaptic connectivity is mediated by stable synapses or by continuous dynamic turnover of synapses of the engram cell network. Here, we first discuss evidence for the persistence of engram cells and memory-relevant adaptations in synaptic plasticity, and then propose models of synaptic adaptations and molecular mechanisms that may support memory persistence through the maintenance of enhanced synaptic connectivity within an engram cell network.
Collapse
Affiliation(s)
- Priyanka Rao-Ruiz
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Esther Visser
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Miodrag Mitrić
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Michel C van den Oever
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
20
|
Salery M, Godino A, Nestler EJ. Drug-activated cells: From immediate early genes to neuronal ensembles in addiction. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 90:173-216. [PMID: 33706932 DOI: 10.1016/bs.apha.2020.09.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Beyond their rapid rewarding effects, drugs of abuse can durably alter an individual's response to their environment as illustrated by the compulsive drug seeking and risk of relapse triggered by drug-associated stimuli. The persistence of these associations even long after cessation of drug use demonstrates the enduring mark left by drugs on brain reward circuits. However, within these circuits, neuronal populations are differently affected by drug exposure and growing evidence indicates that relatively small subsets of neurons might be involved in the encoding and expression of drug-mediated associations. The identification of sparse neuronal populations recruited in response to drug exposure has benefited greatly from the study of immediate early genes (IEGs) whose induction is critical in initiating plasticity programs in recently activated neurons. In particular, the development of technologies to manipulate IEG-expressing cells has been fundamental to implicate broadly distributed neuronal ensembles coincidently activated by either drugs or drug-associated stimuli and to then causally establish their involvement in drug responses. In this review, we summarize the literature regarding IEG regulation in different learning paradigms and addiction models to highlight their role as a marker of activity and plasticity. As the exploration of neuronal ensembles in addiction improves our understanding of drug-associated memory encoding, it also raises several questions regarding the cellular and molecular characteristics of these discrete neuronal populations as they become incorporated in drug-associated neuronal ensembles. We review recent efforts towards this goal and discuss how they will offer a more comprehensive understanding of addiction pathophysiology.
Collapse
Affiliation(s)
- Marine Salery
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Arthur Godino
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
21
|
Bobadilla AC, Dereschewitz E, Vaccaro L, Heinsbroek JA, Scofield MD, Kalivas PW. Cocaine and sucrose rewards recruit different seeking ensembles in the nucleus accumbens core. Mol Psychiatry 2020; 25:3150-3163. [PMID: 32985600 PMCID: PMC8532193 DOI: 10.1038/s41380-020-00888-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 09/07/2020] [Accepted: 09/14/2020] [Indexed: 11/09/2022]
Abstract
Poorly regulated reward seeking is a central feature of substance use disorder. Recent research shows that rewarding drug-related experiences induce synchronous activation of a discrete number of neurons in the nucleus accumbens that are causally linked to reward-related contexts. Here we comprehensively characterize the specific ensemble of neurons built through experience that are linked to seeking behavior. We additionally address the question of whether or not addictive drugs usurp the neuronal networks recruited by natural rewards by evaluating cocaine- and sucrose-associated ensembles within the same mouse. We used FosCreERT2/+/Ai14 transgenic mice to tag cells activated by and potentially encoding cocaine and sucrose seeking. We tagged ~1% of neurons in the core subregion of the accumbens (NAcore) activated during cue-induced seeking for cocaine or sucrose. The majority of tagged cells in the seeking ensembles were D1-MSNs, and specifically activated during seeking, not during extinction or when mice remained in the home cage. To compare different reward-specific ensembles within the same mouse, we used a dual cocaine and sucrose self-administration protocol allowing reward-specific seeking. Using this model, we found ~70% distinction between the cells constituting the cocaine- compared to the sucrose-seeking ensemble. Establishing that cocaine recruits an ensemble of NAcore neurons largely distinct from neurons recruited into an ensemble coding for sucrose seeking suggest a finely tuned specificity of ensembles. The findings allow further exploration of the mechanisms that transform reward-based positive reinforcement into maladaptive drug seeking.
Collapse
Affiliation(s)
- Ana-Clara Bobadilla
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
- School of Pharmacy, University of Wyoming, Laramie, WY, USA.
| | - Eric Dereschewitz
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Lucio Vaccaro
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Jasper A Heinsbroek
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Michael D Scofield
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
22
|
Lynn MB, Lee KFH, Soares C, Naud R, Béïque JC. A Synthetic Likelihood Solution to the Silent Synapse Estimation Problem. Cell Rep 2020; 32:107916. [PMID: 32697998 DOI: 10.1016/j.celrep.2020.107916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/04/2020] [Accepted: 06/25/2020] [Indexed: 11/19/2022] Open
Abstract
Functional features of synaptic populations are typically inferred from random electrophysiological sampling of small subsets of synapses. Are these samples unbiased? Here, we develop a biophysically constrained statistical framework to address this question and apply it to assess the performance of a widely used method based on a failure-rate analysis to quantify the occurrence of silent (AMPAR-lacking) synapses. We simulate this method in silico and find that it is characterized by strong and systematic biases, poor reliability, and weak statistical power. Key conclusions are validated by whole-cell recordings from hippocampal neurons. To address these shortcomings, we develop a simulator of the experimental protocol and use it to compute a synthetic likelihood. By maximizing the likelihood, we infer silent synapse fraction with no bias, low variance, and superior statistical power over alternatives. Together, this generalizable approach highlights how a simulator of experimental methodologies can substantially improve the estimation of physiological properties.
Collapse
Affiliation(s)
- Michael B Lynn
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Kevin F H Lee
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Cary Soares
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Richard Naud
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Centre for Neural Dynamics, University of Ottawa, Ottawa, ON K1H 8M5, Canada; University of Ottawa's Brain and Mind Research Institute, Ottawa, ON K1H 8M5, Canada; Department of Physics, STEM Complex, Room 336, 150 Louis Pasteur Private, University of Ottawa, Ottawa, ON K1N 6N5, Canada.
| | - Jean-Claude Béïque
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Canadian Partnership for Stroke Recovery, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Centre for Neural Dynamics, University of Ottawa, Ottawa, ON K1H 8M5, Canada; University of Ottawa's Brain and Mind Research Institute, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
23
|
Simpson S, Kimbrough A, Boomhower B, McLellan R, Hughes M, Shankar K, de Guglielmo G, George O. Depletion of the Microbiome Alters the Recruitment of Neuronal Ensembles of Oxycodone Intoxication and Withdrawal. eNeuro 2020; 7:ENEURO.0312-19.2020. [PMID: 32341122 PMCID: PMC7242819 DOI: 10.1523/eneuro.0312-19.2020] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 02/26/2020] [Accepted: 03/24/2020] [Indexed: 12/13/2022] Open
Abstract
Substance use disorders have a complex etiology. Genetics, the environment, and behavior all play a role in the initiation, escalation, and relapse of drug use. Recently, opioid use disorder has become a national health crisis. One aspect of opioid addiction that has yet to be fully examined is the effects of alterations of the microbiome and gut-brain axis signaling on central nervous system activity during opioid intoxication and withdrawal. The effect of microbiome depletion on the activation of neuronal ensembles was measured by detecting Fos-positive (Fos+) neuron activation during intoxication and withdrawal using a rat model of oxycodone dependence. Daily oxycodone administration (2 mg/kg) increased pain thresholds and increased Fos+ neurons in the basolateral amygdala (BLA) during intoxication, with a decrease in pain thresholds and increase in Fos+ neurons in the periaqueductal gray (PAG), central nucleus of the amygdala (CeA), locus coeruleus (LC), paraventricular nucleus of the thalamus (PVT), agranular insular cortex (AI), bed nucleus of the stria terminalis (BNST), and lateral habenula medial parvocellular region during withdrawal. Microbiome depletion produced widespread but region- and state-specific changes in neuronal ensemble activation. Oxycodone intoxication and withdrawal also increased functional connectivity among brain regions. Microbiome depletion resulted in a decorrelation of this functional network. These data indicate that microbiome depletion by antibiotics produces widespread changes in the recruitment of neuronal ensembles that are activated by oxycodone intoxication and withdrawal, suggesting that the gut microbiome may play a role in opioid use and dependence. Future studies are needed to better understand the molecular, neurobiological, and behavioral effects of microbiome depletion on addiction-like behaviors.
Collapse
Affiliation(s)
- Sierra Simpson
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037
| | - Adam Kimbrough
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093
| | - Brent Boomhower
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093
| | - Rio McLellan
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093
| | - Marcella Hughes
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093
| | - Kokila Shankar
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037
| | | | - Olivier George
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093,
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
24
|
Hwang EK, Lupica CR. Altered Corticolimbic Control of the Nucleus Accumbens by Long-term Δ 9-Tetrahydrocannabinol Exposure. Biol Psychiatry 2020; 87:619-631. [PMID: 31543247 PMCID: PMC7002212 DOI: 10.1016/j.biopsych.2019.07.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/14/2019] [Accepted: 07/04/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND The decriminalization and legalization of cannabis and the expansion of availability of medical cannabis in North America have led to an increase in cannabis use and the availability of high-potency strains. Cannabis potency is determined by the concentration of Δ9-tetrahydrocannabinol (Δ9-THC), a psychoactive constituent that activates cannabinoid CB1 and CB2 receptors. The use of high-potency cannabis is associated with cannabis use disorder and increased susceptibility to psychiatric illness. The nucleus accumbens (NAc) is part of a brain reward circuit affected by Δ9-THC through modulation of glutamate afferents arising from corticolimbic brain areas implicated in drug addiction and psychiatric disorders. Moreover, brain imaging studies show alterations in corticolimbic and NAc properties in human cannabis users. METHODS Using in vitro electrophysiology and optogenetics, we examined how Δ9-THC alters corticolimbic input to the NAc in rats. RESULTS We found that long-term exposure to Δ9-THC weakens prefrontal cortex glutamate input to the NAc shell and strengthens input from basolateral amygdala and ventral hippocampus. Further, whereas long-term exposure to Δ9-THC had no effect on net strength of glutamatergic input to NAc shell arising from midbrain dopamine neurons, it alters fundamental properties of these synapses. CONCLUSIONS Long-term exposure to Δ9-THC shifts control of the NAc shell from cortical to limbic input, likely contributing to cognitive and psychiatric dysfunction that is associated with cannabis use.
Collapse
|
25
|
Gao S, Zhang H, He J, Zheng H, Zhang P, Xu J, Shen Z, Zhao H, Wang F, Hu Z, Chen J. Neuronal HMGB1 in nucleus accumbens regulates cocaine reward memory. Addict Biol 2020; 25:e12739. [PMID: 31056833 DOI: 10.1111/adb.12739] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 11/29/2022]
Abstract
Cocaine is a common abused drug that can induce abnormal synaptic and immune responses in the central nervous system (CNS). High mobility group box 1 (HMGB1) is one kind of inflammatory molecules that is expressed both on neurons and immune cells. Previous studies of HMGB1 in the CNS have largely focused on immune function, and the role of HMGB1 in neurons and cocaine addiction remains unknown. Here, we show that cocaine exposure induced the translocation and release of HMGB1 in the nucleus accumbens (NAc) neurons. Gain and loss of HMGB1 in the NAc bidirectionally regulate cocaine-induced conditioned place preference. From the nucleus to the cytosol, HMGB1 binds to glutamate receptor subunits (GluA2/GluN2B) on the membrane, which regulates cocaine-induced synaptic adaptation and the formation of cocaine-related memory. These data unveil the role of HMGB1 in neurons and provide the evidence for the HMGB1 involvement in drug addiction.
Collapse
Affiliation(s)
- Shuang‐Qi Gao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan China
| | - Hai Zhang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic EvaluationHuazhong University of Science and Technology Wuhan China
- Laboratory of Neuropsychiatric Diseases, The Institute of Brain ResearchHuazhong University of Science and Technology Wuhan China
- The Key Laboratory of Neurological DiseasesHuazhong University of Science and Technology, Ministry of Education of China Wuhan China
| | - Jin‐Gang He
- Department of Pharmacology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan China
| | - Hui‐Ling Zheng
- Department of Pharmacology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan China
| | - Pei‐Wei Zhang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan China
| | - Jun‐Feng Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan China
| | - Zu‐Cheng Shen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan China
| | - Huan‐Huan Zhao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan China
| | - Fang Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic EvaluationHuazhong University of Science and Technology Wuhan China
- Laboratory of Neuropsychiatric Diseases, The Institute of Brain ResearchHuazhong University of Science and Technology Wuhan China
- The Collaborative‐Innovation Center for Brain ScienceHuazhong University of Science and Technology Wuhan China
- The Key Laboratory of Neurological DiseasesHuazhong University of Science and Technology, Ministry of Education of China Wuhan China
| | - Zhuang‐Li Hu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic EvaluationHuazhong University of Science and Technology Wuhan China
- Laboratory of Neuropsychiatric Diseases, The Institute of Brain ResearchHuazhong University of Science and Technology Wuhan China
- The Key Laboratory of Neurological DiseasesHuazhong University of Science and Technology, Ministry of Education of China Wuhan China
| | - Jian‐Guo Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and Technology Wuhan China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic EvaluationHuazhong University of Science and Technology Wuhan China
- Laboratory of Neuropsychiatric Diseases, The Institute of Brain ResearchHuazhong University of Science and Technology Wuhan China
- The Collaborative‐Innovation Center for Brain ScienceHuazhong University of Science and Technology Wuhan China
- The Key Laboratory of Neurological DiseasesHuazhong University of Science and Technology, Ministry of Education of China Wuhan China
| |
Collapse
|
26
|
Wall NR, Neumann PA, Beier KT, Mokhtari AK, Luo L, Malenka RC. Complementary Genetic Targeting and Monosynaptic Input Mapping Reveal Recruitment and Refinement of Distributed Corticostriatal Ensembles by Cocaine. Neuron 2019; 104:916-930.e5. [PMID: 31759807 DOI: 10.1016/j.neuron.2019.10.032] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 09/24/2019] [Accepted: 10/24/2019] [Indexed: 10/25/2022]
Abstract
Drugs of abuse elicit powerful experiences that engage populations of neurons broadly distributed throughout the brain. To determine how synaptic connectivity is organized to enable robust communication between populations of drug-activated neurons, we developed a complementary targeting system for monosynaptic rabies virus (RV) tracing that identifies direct inputs to activated versus nonactivated neuronal populations. Analysis of over 100,000 synaptic input neurons demonstrated that cocaine-activated neurons comprise selectively connected but broadly distributed corticostriatal networks. Electrophysiological assays using optogenetics to stimulate activated versus nonactivated inputs revealed stronger synapses between coactivated cortical pyramidal neurons and neurons in the dorsal striatum (DS). Repeated cocaine exposure further enhanced the connectivity specifically between drug-activated neurons in the orbitofrontal cortex (OFC) and coactive DS neurons. Selective chemogenetic silencing of cocaine-activated OFC neurons or their terminals in the DS disrupted behavioral sensitization, demonstrating the utility of this methodology for identifying novel circuit elements that contribute to behavioral plasticity.
Collapse
Affiliation(s)
- Nicholas R Wall
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Peter A Neumann
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kevin T Beier
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Biology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ava K Mokhtari
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Liqun Luo
- Department of Biology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Robert C Malenka
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
27
|
The Emergence of a Stable Neuronal Ensemble from a Wider Pool of Activated Neurons in the Dorsal Medial Prefrontal Cortex during Appetitive Learning in Mice. J Neurosci 2019; 40:395-410. [PMID: 31727794 DOI: 10.1523/jneurosci.1496-19.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 11/04/2019] [Accepted: 11/06/2019] [Indexed: 11/21/2022] Open
Abstract
Animals selectively respond to environmental cues associated with food reward to optimize nutrient intake. Such appetitive conditioned stimulus-unconditioned stimulus (CS-US) associations are thought to be encoded in select, stable neuronal populations or neuronal ensembles, which undergo physiological modifications during appetitive conditioning. These ensembles in the medial prefrontal cortex (mPFC) control well-established, cue-evoked food seeking, but the mechanisms involved in the genesis of these ensembles are unclear. Here, we used male Fos-GFP mice that express green fluorescent protein (GFP) in recently behaviorally activated neurons, to reveal how dorsal mPFC neurons are recruited and modified to encode CS-US memory representations using an appetitive conditioning task. In the initial conditioning session, animals did not exhibit discriminated, cue-selective food seeking, but did so in later sessions indicating that a CS-US association was established. Using microprism-based in vivo 2-Photon imaging, we revealed that only a minority of neurons activated during the initial session was consistently activated throughout subsequent conditioning sessions and during cue-evoked memory recall. Notably, using ex vivo electrophysiology, we found that neurons activated following the initial session exhibited transient hyperexcitability. Chemogenetically enhancing the excitability of these neurons throughout subsequent conditioning sessions interfered with the development of reliable cue-selective food seeking, indicated by persistent, nondiscriminated performance. We demonstrate how appetitive learning consistently activates a subset of neurons to form a stable neuronal ensemble during the formation of a CS-US association. This ensemble may arise from a pool of hyperexcitable neurons activated during the initial conditioning session.SIGNIFICANCE STATEMENT Appetitive conditioning endows cues associated with food with the ability to guide food-seeking, through the formation of a food-cue association. Neuronal ensembles in the mPFC control established cue-evoked food-seeking. However, how neurons undergo physiological modifications and become part of an ensemble during conditioning remain unclear. We found that only a minority of dorsal mPFC neurons activated on the initial conditioning session became consistently activated during conditioning and memory recall. These initially activated neurons were also transiently hyperexcitable. We demonstrate the following: (1) how stable neuronal ensemble formation in the dorsal mPFC underlies appetitive conditioning; and (2) how this ensemble may arise from hyperexcitable neurons activated before the establishment of cue-evoked food seeking.
Collapse
|
28
|
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: 0.8] [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
|
29
|
Cameron CM, Murugan M, Choi JY, Engel EA, Witten IB. Increased Cocaine Motivation Is Associated with Degraded Spatial and Temporal Representations in IL-NAc Neurons. Neuron 2019; 103:80-91.e7. [PMID: 31101395 DOI: 10.1016/j.neuron.2019.04.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 02/07/2019] [Accepted: 04/05/2019] [Indexed: 01/03/2023]
Abstract
Craving for cocaine progressively increases in cocaine users during drug-free periods, contributing to relapse. The projection from the infralimbic cortex to the nucleus accumbens shell (IL-NAc) is thought to inhibit cocaine seeking. However, it is not known whether and how IL-NAc neurons contribute to the increased motivation associated with a drug-free period. We first performed cellular resolution imaging of IL-NAc neurons in rats during a drug-seeking test. This revealed neurons with spatial selectivity within the cocaine-associated context, a decrease in activity around the time of cocaine seeking, and an inverse relationship between cocaine-seeking activity and subsequent cocaine motivation. All these properties were reduced by a drug-free period. Next, we transiently activated this projection, which resulted in reduced drug seeking, regardless of the drug-free period. Taken together, this suggests that altered IL-NAc activity after a drug-free period may enhance cocaine motivation without fundamentally altering the projection's ability to inhibit drug seeking.
Collapse
Affiliation(s)
- Courtney M Cameron
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Malavika Murugan
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Jung Yoon Choi
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA; Department of Psychology, Princeton University, Princeton, NJ 08544, USA
| | - Esteban A Engel
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Ilana B Witten
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA; Department of Psychology, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
30
|
Bariselli S, Fobbs WC, Creed MC, Kravitz AV. A competitive model for striatal action selection. Brain Res 2018; 1713:70-79. [PMID: 30300636 DOI: 10.1016/j.brainres.2018.10.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 10/03/2018] [Accepted: 10/05/2018] [Indexed: 12/20/2022]
Abstract
The direct and indirect pathway striatal medium spiny neurons (dMSNs and iMSNs) have long been linked to action selection, but the precise roles of these neurons in this process remain unclear. Here, we review different models of striatal pathway function, focusing on the classic "go/no-go" model which posits that dMSNs facilitate movement while iMSNs inhibit movement, and the "complementary" model, which argues that dMSNs facilitate the selection of specific actions while iMSNs inhibit potentially conflicting actions. We discuss the merits and shortcomings of these models and propose a "competitive" model to explain the contribution of these two pathways to behavior. The "competitive" model argues that rather than inhibiting conflicting actions, iMSNs are tuned to the same actions that dMSNs facilitate, and the two populations "compete" to determine the animal's behavioral response. This model provides a theoretical explanation for how these pathways work together to select actions. In addition, it provides a link between action selection and behavioral reinforcement, via modulating synaptic strength at inputs onto dMSNs and iMSNs. Finally, this model makes predictions about how imbalances in the activity of these pathways may underlie behavioral traits associated with psychiatric disorders. Understanding the roles of these striatal pathways in action selection may help to clarify the neuronal mechanisms of decision-making under normal and pathological conditions.
Collapse
Affiliation(s)
- S Bariselli
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, United States
| | - W C Fobbs
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, United States
| | - M C Creed
- Washington University in St Louis, Department of Anesthesiology, St Louis, MO, United States
| | - A V Kravitz
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, United States; National Institute on Drug Abuse, Baltimore, MD, United States.
| |
Collapse
|
31
|
Generation of silent synapses in dentate gyrus correlates with development of alcohol addiction. Neuropsychopharmacology 2018; 43:1989-1999. [PMID: 29967367 PMCID: PMC6098144 DOI: 10.1038/s41386-018-0119-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 05/29/2018] [Accepted: 06/05/2018] [Indexed: 12/20/2022]
Abstract
The brain circuits and synaptic processes that underlie alcohol addiction are currently the subject of intensive research. Here we focus on hippocampal circuitry and show that chemogenetic inhibition of dentate gyrus (DG) during presentation of alcohol-associated cues has long-lasting effects on mice behavior. DG inhibition enhances alcohol seeking and drinking, suggesting that DG regulates addiction-related behaviors. To test this hypothesis, we perform whole-cell patch-clamp recordings from the granule cells of DG and look for electrophysiological correlates of alcohol addiction. We observe that presentation of alcohol-associated cue light that induces relapse to alcohol-seeking results in generation of silent synapses, that lack functional AMPA receptors. Furthermore, using human criteria of addiction, we differentiate mice controlling their alcohol consumption from those that undergo transition to addiction to discover that the levels of silent synapses induced by alcohol cues are specifically increased in the addicted mice. As the total level of dendritic spines that harbor synapses is constant at this time point, our data indicate that synapses of perforant path to DG are weakened during cue relapse. Finally we demonstrate that, acamprosate, a drug that limits alcohol drinking and seeking in addicts, prevents generation of silent synapses in DG upon presentation of alcohol-associated cues. Altogether, our data suggest that weakening of DG synapses upon cue relapse contributes to persistent alcohol addiction-related behaviors.
Collapse
|
32
|
Koya E, Dong Y. Sound of silent synapses from the addicted hippocampus. Neuropsychopharmacology 2018; 43:1981-1982. [PMID: 30030543 PMCID: PMC6098148 DOI: 10.1038/s41386-018-0142-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 06/28/2018] [Indexed: 11/09/2022]
Affiliation(s)
- Eisuke Koya
- Sussex Neuroscience, School of Psychology, University of Sussex, Falmer, BN1 9QG, UK.
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| |
Collapse
|
33
|
McDevitt DS, Graziane NM. Neuronal mechanisms mediating pathological reward-related behaviors: A focus on silent synapses in the nucleus accumbens. Pharmacol Res 2018; 136:90-96. [PMID: 30171902 DOI: 10.1016/j.phrs.2018.08.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 08/28/2018] [Indexed: 12/11/2022]
Abstract
The compulsive drive to seek drugs despite negative consequences relies heavily on drug-induced alterations that occur within the reward neurocircuit. These alterations include changes in neuromodulator and neurotransmitter systems that ultimately lock behaviors into an inflexible and permanent state. To provide clinicians with improved treatment options, researchers are trying to identify, as potential targets of therapeutic intervention, the neural mechanisms mediating an "addictive-like state". Here, we discuss how drug-induced generation of silent synapses in the nucleus accumbens may be a potential therapeutic target capable of reversing drug-related behaviors.
Collapse
Affiliation(s)
- Dillon S McDevitt
- Departments of Anesthesiology and Perioperative Medicine and Pharmacology, Penn State College of Medicine, Hershey, PA, 17033 USA; Neuroscience graduate program, Penn State College of Medicine, Hershey, PA, 17033 USA
| | - Nicholas M Graziane
- Departments of Anesthesiology and Perioperative Medicine and Pharmacology, Penn State College of Medicine, Hershey, PA, 17033 USA.
| |
Collapse
|
34
|
Whitaker LR, Hope BT. Chasing the addicted engram: identifying functional alterations in Fos-expressing neuronal ensembles that mediate drug-related learned behavior. ACTA ACUST UNITED AC 2018; 25:455-460. [PMID: 30115767 PMCID: PMC6097770 DOI: 10.1101/lm.046698.117] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 07/09/2018] [Indexed: 01/13/2023]
Abstract
Given that addiction has been characterized as a disorder of maladaptive learning and memory, one critical question is whether there are unique physical adaptations within neuronal ensembles that support addiction-related learned behavior. The search for the physical mechanisms of encoding these and other memories in the brain, often called the engram as a whole, continues despite decades of research. As we develop new technologies and tools that allow us to study cue- and behavior-activated Fos-expressing neuronal ensembles, the possibility of identifying the engrams of learning and memory is moving into the realm of reality rather than speculation. It has become clear from recent studies that there are specific functional, electrophysiological alterations unique to Fos-expressing ensemble neurons that may participate in encoding memories. The ultimate goal is to identify the addicted engram and reverse the physical changes that support this maladaptive form of learning.
Collapse
Affiliation(s)
- Leslie R Whitaker
- Office of the Scientific Director; Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Bruce T Hope
- Neuronal Ensembles in Addiction Section; Behavioral Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224, USA
| |
Collapse
|
35
|
Cahill ME, Walker DM, Gancarz AM, Wang ZJ, Lardner CK, Bagot RC, Neve RL, Dietz DM, Nestler EJ. The dendritic spine morphogenic effects of repeated cocaine use occur through the regulation of serum response factor signaling. Mol Psychiatry 2018; 23:1474-1486. [PMID: 28555077 PMCID: PMC5709273 DOI: 10.1038/mp.2017.116] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 03/23/2017] [Accepted: 04/12/2017] [Indexed: 12/28/2022]
Abstract
The nucleus accumbens (NAc) is a primary brain reward region composed predominantly of medium spiny neurons (MSNs). In response to early withdrawal from repeated cocaine administration, de novo dendritic spine formation occurs in NAc MSNs. Much evidence indicates that this new spine formation facilitates the rewarding properties of cocaine. Early withdrawal from repeated cocaine also produces dramatic alterations in the transcriptome of NAc MSNs, but how such alterations influence cocaine's effects on dendritic spine formation remain unclear. Studies in non-neuronal cells indicate that actin cytoskeletal regulatory pathways in nuclei have a direct role in the regulation of gene transcription in part by controlling the access of co-activators to their transcription factor partners. In particular, actin state dictates the interaction between the serum response factor (SRF) transcription factor and one of its principal co-activators, MAL. Here we show that cocaine induces alterations in nuclear F-actin signaling pathways in the NAc with associated changes in the nuclear subcellular localization of SRF and MAL. Using in vivo optogenetics, the brain region-specific inputs to the NAc that mediate these nuclear changes are investigated. Finally, we demonstrate that regulated SRF expression, in turn, is critical for the effects of cocaine on dendritic spine formation and for cocaine-mediated behavioral sensitization. Collectively, these findings reveal a mechanism by which nuclear-based changes influence the structure of NAc MSNs in response to cocaine.
Collapse
Affiliation(s)
- ME Cahill
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - DM Walker
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - AM Gancarz
- Department of Pharmacology and Toxicology, Research Institute on Addictions, Program in Neuroscience, State University at Buffalo, Buffalo, NY, USA
| | - ZJ Wang
- Department of Pharmacology and Toxicology, Research Institute on Addictions, Program in Neuroscience, State University at Buffalo, Buffalo, NY, USA
| | - CK Lardner
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - RC Bagot
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Department of Psychology, McGill University, Montréal, QC, Canada
| | - RL Neve
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - DM Dietz
- Department of Pharmacology and Toxicology, Research Institute on Addictions, Program in Neuroscience, State University at Buffalo, Buffalo, NY, USA
| | - EJ Nestler
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
36
|
Parrilla-Carrero J, Buchta WC, Goswamee P, Culver O, McKendrick G, Harlan B, Moutal A, Penrod R, Lauer A, Ramakrishnan V, Khanna R, Kalivas P, Riegel AC. Restoration of Kv7 Channel-Mediated Inhibition Reduces Cued-Reinstatement of Cocaine Seeking. J Neurosci 2018; 38:4212-4229. [PMID: 29636392 PMCID: PMC5963852 DOI: 10.1523/jneurosci.2767-17.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 03/27/2018] [Accepted: 03/29/2018] [Indexed: 12/16/2022] Open
Abstract
Cocaine addicts display increased sensitivity to drug-associated cues, due in part to changes in the prelimbic prefrontal cortex (PL-PFC). The cellular mechanisms underlying cue-induced reinstatement of cocaine seeking remain unknown. Reinforcement learning for addictive drugs may produce persistent maladaptations in intrinsic excitability within sparse subsets of PFC pyramidal neurons. Using a model of relapse in male rats, we sampled >600 neurons to examine spike frequency adaptation (SFA) and afterhyperpolarizations (AHPs), two systems that attenuate low-frequency inputs to regulate neuronal synchronization. We observed that training to self-administer cocaine or nondrug (sucrose) reinforcers decreased SFA and AHPs in a subpopulation of PL-PFC neurons. Only with cocaine did the resulting hyperexcitability persist through extinction training and increase during reinstatement. In neurons with intact SFA, dopamine enhanced excitability by inhibiting Kv7 potassium channels that mediate SFA. However, dopamine effects were occluded in neurons from cocaine-experienced rats, where SFA and AHPs were reduced. Pharmacological stabilization of Kv7 channels with retigabine restored SFA and Kv7 channel function in neuroadapted cells. When microinjected bilaterally into the PL-PFC 10 min before reinstatement testing, retigabine reduced cue-induced reinstatement of cocaine seeking. Last, using cFos-GFP transgenic rats, we found that the loss of SFA correlated with the expression of cFos-GFP following both extinction and re-exposure to drug-associated cues. Together, these data suggest that cocaine self-administration desensitizes inhibitory Kv7 channels in a subpopulation of PL-PFC neurons. This subpopulation of neurons may represent a persistent neural ensemble responsible for driving drug seeking in response to cues.SIGNIFICANCE STATEMENT Long after the cessation of drug use, cues associated with cocaine still elicit drug-seeking behavior, in part by activation of the prelimbic prefrontal cortex (PL-PFC). The underlying cellular mechanisms governing these activated neurons remain unclear. Using a rat model of relapse to cocaine seeking, we identified a population of PL-PFC neurons that become hyperexcitable following chronic cocaine self-administration. These neurons show persistent loss of spike frequency adaptation, reduced afterhyperpolarizations, decreased sensitivity to dopamine, and reduced Kv7 channel-mediated inhibition. Stabilization of Kv7 channel function with retigabine normalized neuronal excitability, restored Kv7 channel currents, and reduced drug-seeking behavior when administered into the PL-PFC before reinstatement. These data highlight a persistent adaptation in a subset of PL-PFC neurons that may contribute to relapse vulnerability.
Collapse
Affiliation(s)
- Jeffrey Parrilla-Carrero
- Department of Neuroscience
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - William C Buchta
- Department of Neuroscience
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Priyodarshan Goswamee
- Department of Neuroscience
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Oliver Culver
- Department of Neuroscience
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Greer McKendrick
- Department of Neuroscience
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Benjamin Harlan
- Department of Neuroscience
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Aubin Moutal
- Department of Pharmacology, University of Arizona, Tucson, Arizona 85724, and
| | - Rachel Penrod
- Department of Neuroscience
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Abigail Lauer
- Department of Public Health Sciences., Medical University of South Carolina, Charleston, SC 29425
| | - Viswanathan Ramakrishnan
- Department of Public Health Sciences., Medical University of South Carolina, Charleston, SC 29425
| | - Rajesh Khanna
- Department of Pharmacology, University of Arizona, Tucson, Arizona 85724, and
| | - Peter Kalivas
- Department of Neuroscience
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Arthur C Riegel
- Department of Neuroscience,
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, South Carolina 29425
| |
Collapse
|
37
|
|
38
|
Regional Differences in Striatal Neuronal Ensemble Excitability Following Cocaine and Extinction Memory Retrieval in Fos-GFP Mice. Neuropsychopharmacology 2018; 43:718-727. [PMID: 28540927 PMCID: PMC5809776 DOI: 10.1038/npp.2017.101] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/03/2017] [Accepted: 05/18/2017] [Indexed: 11/08/2022]
Abstract
Learned associations between drugs of abuse and the drug administration environment have an important role in addiction. In rodents, exposure to a drug-associated environment elicits conditioned psychomotor activation, which may be weakened following extinction (EXT) learning. Although widespread drug-induced changes in neuronal excitability have been observed, little is known about specific changes within neuronal ensembles activated during the recall of drug-environment associations. Using a cocaine-conditioned locomotion (CL) procedure, the present study assessed the excitability of neuronal ensembles in the nucleus accumbens core and shell (NAccore and NAcshell), and dorsal striatum (DS) following cocaine conditioning and EXT in Fos-GFP mice that express green fluorescent protein (GFP) in activated neurons (GFP+). During conditioning, mice received repeated cocaine injections (20 mg/kg) paired with a locomotor activity chamber (Paired) or home cage (Unpaired). Seven to 13 days later, both groups were re-exposed to the activity chamber under drug-free conditions and Paired, but not Unpaired, mice exhibited CL. In a separate group of mice, CL was extinguished by repeatedly exposing mice to the activity chamber under drug-free conditions. Following the expression and EXT of CL, GFP+ neurons in the NAccore (but not NAcshell and DS) displayed greater firing capacity compared to surrounding GFP- neurons. This difference in excitability was due to a generalized decrease in GFP- excitability following CL and a selective increase in GFP+ excitability following its EXT. These results suggest a role for both widespread and ensemble-specific changes in neuronal excitability following recall of drug-environment associations.
Collapse
|
39
|
Hearing M, Graziane N, Dong Y, Thomas MJ. Opioid and Psychostimulant Plasticity: Targeting Overlap in Nucleus Accumbens Glutamate Signaling. Trends Pharmacol Sci 2018; 39:276-294. [PMID: 29338873 DOI: 10.1016/j.tips.2017.12.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/11/2017] [Accepted: 12/13/2017] [Indexed: 12/11/2022]
Abstract
Commonalities in addictive behavior, such as craving, stimuli-driven drug seeking, and a high propensity for relapse following abstinence, have pushed for a unified theory of addiction that encompasses most abused substances. This unitary theory has recently been challenged - citing distinctions in structural neural plasticity, biochemical signaling, and neural circuitry to argue that addiction to opioids and psychostimulants is behaviorally and neurobiologically distinct. Recent more selective examination of drug-induced plasticity has highlighted that these two drug classes promote an overall reward circuitry signaling overlap through modifying excitatory synapses in the nucleus accumbens - a key constituent of the reward system. We discuss adaptations in presynaptic/postsynaptic and extrasynaptic glutamate signaling produced by opioids and psychostimulants, and their relevance to circuit remodeling and addiction-related behavior - arguing that these core neural adaptations are important targets for developing pharmacotherapies to treat addiction to multiple drugs.
Collapse
Affiliation(s)
- Matthew Hearing
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI 53233, USA.
| | - Nicholas Graziane
- Department of Anesthesiology and Perioperative Medicine, Penn State College of Medicine, Hershey, PA 17033, USA; Departments of Neuroscience and Psychiatry, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Yan Dong
- Departments of Neuroscience and Psychiatry, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Mark J Thomas
- Department of Psychology, University of Minnesota, Minneapolis, MN 55455, USA; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
40
|
Dos Santos M, Salery M, Forget B, Garcia Perez MA, Betuing S, Boudier T, Vanhoutte P, Caboche J, Heck N. Rapid Synaptogenesis in the Nucleus Accumbens Is Induced by a Single Cocaine Administration and Stabilized by Mitogen-Activated Protein Kinase Interacting Kinase-1 Activity. Biol Psychiatry 2017; 82:806-818. [PMID: 28545678 DOI: 10.1016/j.biopsych.2017.03.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 03/10/2017] [Accepted: 03/14/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Repeated cocaine exposure produces new spine formation in striatal projection neurons (SPNs) of the nucleus accumbens. However, an acute exposure to cocaine can trigger long-lasting synaptic plasticity in SPNs leading to behavioral alterations. This raises the intriguing question as to whether a single administration of cocaine could enduringly modify striatal connectivity. METHODS A three-dimensional morphometric analysis of presynaptic glutamatergic boutons and dendritic spines was performed on SPNs 1 hour and 1 week after a single cocaine administration. Time-lapse two-photon microscopy in adult slices was used to determine the precise molecular-events sequence responsible for the rapid spine formation. RESULTS A single injection triggered a rapid synaptogenesis and persistent increase in glutamatergic connectivity in SPNs from the shell part of the nucleus accumbens, specifically. Synapse formation occurred through clustered growth of active spines contacting pre-existing axonal boutons. Spine growth required extracellular signal-regulated kinase activation, while spine stabilization involved transcription-independent protein synthesis driven by mitogen-activated protein kinase interacting kinase-1, downstream from extracellular signal-regulated kinase. The maintenance of new spines driven by mitogen-activated protein kinase interacting kinase-1 was essential for long-term connectivity changes induced by cocaine in vivo. CONCLUSIONS Our study originally demonstrates that a single administration of cocaine is able to induce stable synaptic rewiring in the nucleus accumbens, which will likely influence responses to subsequent drug exposure. It also unravels a new functional role for cocaine-induced extracellular signal-regulated kinase pathway independently of nuclear targets. Finally, it reveals that mitogen-activated protein kinase interacting kinase-1 has a pivotal role in cocaine-induced connectivity.
Collapse
Affiliation(s)
- Marc Dos Santos
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Marine Salery
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Benoit Forget
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Maria Alexandra Garcia Perez
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Sandrine Betuing
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Thomas Boudier
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France; Bioinformatics Institute, Agency for Science, Technology, and Research, Singapore
| | - Peter Vanhoutte
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Jocelyne Caboche
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France.
| | - Nicolas Heck
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France
| |
Collapse
|
41
|
Xia J, Meyers AM, Beeler JA. Chronic Nicotine Alters Corticostriatal Plasticity in the Striatopallidal Pathway Mediated By NR2B-Containing Silent Synapses. Neuropsychopharmacology 2017; 42:2314-2324. [PMID: 28462940 PMCID: PMC5645753 DOI: 10.1038/npp.2017.87] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 04/22/2017] [Accepted: 04/25/2017] [Indexed: 12/28/2022]
Abstract
Smoking is the leading cause of preventable death in the United States and success rates for quitting remain low. High relapse rates are attributed to pervasive nicotine-reinforced associative learning of incentive cues that is highly resistant to extinction. Why such learning is so persistent is poorly understood but may arise as a consequence of neuroadaptations in synaptic plasticity induced by chronic nicotine. We used whole-cell patch clamp recording to investigate the effect of chronic nicotine (cNIC) on synaptic plasticity in dopamine D2 receptor-expressing medium-spiny neurons in the indirect, striatopallidal pathway in dorsolateral striatum. Mice exposed to cNIC exhibited long-term potentiation in response to high-frequency stimulation instead of the expected depression. cNIC decreased baseline AMPA/NMDA ratio, arising from increased NMDA currents enriched in the NR2B subunit with a concomitant upregulation of NMDA-only, silent synapses. These data demonstrate that cNIC can increase silent synapses in MSNs, as observed with cocaine and opiates, and alter the regulation of corticostriatal plasticity. Prior work has characterized cocaine- and morphine-induced upregulation of silent synapses in the ventral striatum; we show it can occur in the dorsal striatum, a region associated with later stages of addiction, craving, and cue-induced relapse.
Collapse
Affiliation(s)
- Jianxun Xia
- Department of Psychology, Queens College and The Graduate Center, City University of New York, Flushing, NY, USA
| | - Allison M Meyers
- Department of Psychology, Queens College and The Graduate Center, City University of New York, Flushing, NY, USA
| | - Jeff A Beeler
- Department of Psychology, Queens College and The Graduate Center, City University of New York, Flushing, NY, USA,Department of Psychology, Queens College and The Graduate Center, CUNY, 65-30 Kissena Blvd, Queens, NY 11367, USA, Tel: +718 570 0517, Fax: +773 793 2588, E-mail:
| |
Collapse
|
42
|
Bobadilla AC, Heinsbroek JA, Gipson CD, Griffin WC, Fowler CD, Kenny PJ, Kalivas PW. Corticostriatal plasticity, neuronal ensembles, and regulation of drug-seeking behavior. PROGRESS IN BRAIN RESEARCH 2017; 235:93-112. [PMID: 29054293 DOI: 10.1016/bs.pbr.2017.07.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The idea that interconnected neuronal ensembles code for specific behaviors has been around for decades; however, recent technical improvements allow studying these networks and their causal role in initiating and maintaining behavior. In particular, the role of ensembles in drug-seeking behaviors in the context of addiction is being actively investigated. Concurrent with breakthroughs in quantifying ensembles, research has identified a role for synaptic glutamate spillover during relapse. In particular, the transient relapse-associated changes in glutamatergic synapses on accumbens neurons, as well as in adjacent astroglia and extracellular matrix, are key elements of the synaptic plasticity encoded by drug use and the metaplasticity induced by drug-associated cues that precipitate drug-seeking behaviors. Here, we briefly review the recent discoveries related to ensembles in the addiction field and then endeavor to link these discoveries with drug-induced striatal plasticity and cue-induced metaplasticity toward deeper neurobiological understandings of drug seeking.
Collapse
Affiliation(s)
| | | | | | | | | | - Paul J Kenny
- Icahn School of Medicine at Mount Sinai, Icahn, New York, NY, United States
| | - Peter W Kalivas
- Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
43
|
Bidirectional Modulation of Intrinsic Excitability in Rat Prelimbic Cortex Neuronal Ensembles and Non-Ensembles after Operant Learning. J Neurosci 2017; 37:8845-8856. [PMID: 28779019 DOI: 10.1523/jneurosci.3761-16.2017] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 07/10/2017] [Accepted: 07/18/2017] [Indexed: 11/21/2022] Open
Abstract
Learned associations between environmental stimuli and rewards drive goal-directed learning and motivated behavior. These memories are thought to be encoded by alterations within specific patterns of sparsely distributed neurons called neuronal ensembles that are activated selectively by reward-predictive stimuli. Here, we use the Fos promoter to identify strongly activated neuronal ensembles in rat prelimbic cortex (PLC) and assess altered intrinsic excitability after 10 d of operant food self-administration training (1 h/d). First, we used the Daun02 inactivation procedure in male FosLacZ-transgenic rats to ablate selectively Fos-expressing PLC neurons that were active during operant food self-administration. Selective ablation of these neurons decreased food seeking. We then used male FosGFP-transgenic rats to assess selective alterations of intrinsic excitability in Fos-expressing neuronal ensembles (FosGFP+) that were activated during food self-administration and compared these with alterations in less activated non-ensemble neurons (FosGFP-). Using whole-cell recordings of layer V pyramidal neurons in an ex vivo brain slice preparation, we found that operant self-administration increased excitability of FosGFP+ neurons and decreased excitability of FosGFP- neurons. Increased excitability of FosGFP+ neurons was driven by increased steady-state input resistance. Decreased excitability of FosGFP- neurons was driven by increased contribution of small-conductance calcium-activated potassium (SK) channels. Injections of the specific SK channel antagonist apamin into PLC increased Fos expression but had no effect on food seeking. Overall, operant learning increased intrinsic excitability of PLC Fos-expressing neuronal ensembles that play a role in food seeking but decreased intrinsic excitability of Fos- non-ensembles.SIGNIFICANCE STATEMENT Prefrontal cortex activity plays a critical role in operant learning, but the underlying cellular mechanisms are unknown. Using the chemogenetic Daun02 inactivation procedure, we found that a small number of strongly activated Fos-expressing neuronal ensembles in rat PLC play an important role in learned operant food seeking. Using GFP expression to identify Fos-expressing layer V pyramidal neurons in prelimbic cortex (PLC) of FosGFP-transgenic rats, we found that operant food self-administration led to increased intrinsic excitability in the behaviorally relevant Fos-expressing neuronal ensembles, but decreased intrinsic excitability in Fos- neurons using distinct cellular mechanisms.
Collapse
|
44
|
Kim HJ, Lee JH, Yun K, Kim JH. Alterations in Striatal Circuits Underlying Addiction-Like Behaviors. Mol Cells 2017; 40:379-385. [PMID: 28724279 PMCID: PMC5523013 DOI: 10.14348/molcells.2017.0088] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/07/2017] [Accepted: 07/11/2017] [Indexed: 02/08/2023] Open
Abstract
Drug addiction is a severe psychiatric disorder characterized by the compulsive pursuit of drugs of abuse despite potential adverse consequences. Although several decades of studies have revealed that psychostimulant use can result in extensive alterations of neural circuits and physiology, no effective therapeutic strategies or medicines for drug addiction currently exist. Changes in neuronal connectivity and regulation occurring after repeated drug exposure contribute to addiction-like behaviors in animal models. Among the involved brain areas, including those of the reward system, the striatum is the major area of convergence for glutamate, GABA, and dopamine transmission, and this brain region potentially determines stereotyped behaviors. Although the physiological consequences of striatal neurons after drug exposure have been relatively well documented, it remains to be clarified how changes in striatal connectivity underlie and modulate the expression of addiction-like behaviors. Understanding how striatal circuits contribute to addiction-like behaviors may lead to the development of strategies that successfully attenuate drug-induced behavioral changes. In this review, we summarize the results of recent studies that have examined striatal circuitry and pathway-specific alterations leading to addiction-like behaviors to provide an updated framework for future investigations.
Collapse
Affiliation(s)
- Hyun Jin Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673,
Korea
| | - Joo Han Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673,
Korea
| | - Kyunghwa Yun
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673,
Korea
| | - Joung-Hun Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673,
Korea
| |
Collapse
|
45
|
Chandra R, Lobo MK. Beyond Neuronal Activity Markers: Select Immediate Early Genes in Striatal Neuron Subtypes Functionally Mediate Psychostimulant Addiction. Front Behav Neurosci 2017. [PMID: 28642692 PMCID: PMC5462953 DOI: 10.3389/fnbeh.2017.00112] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Immediate early genes (IEGs) were traditionally used as markers of neuronal activity in striatum in response to stimuli including drugs of abuse such as psychostimulants. Early studies using these neuronal activity markers led to important insights in striatal neuron subtype responsiveness to psychostimulants. Such studies have helped identify striatum as a critical brain center for motivational, reinforcement and habitual behaviors in psychostimulant addiction. While the use of IEGs as neuronal activity markers in response to psychostimulants and other stimuli persists today, the functional role and implications of these IEGs has often been neglected. Nonetheless, there is a subset of research that investigates the functional role of IEGs in molecular, cellular and behavioral alterations by psychostimulants through striatal medium spiny neuron (MSN) subtypes, the two projection neuron subtypes in striatum. This review article will address and highlight the studies that provide a functional mechanism by which IEGs mediate psychostimulant molecular, cellular and behavioral plasticity through MSN subtypes. Insight into the functional role of IEGs in striatal MSN subtypes could provide improved understanding into addiction and neuropsychiatric diseases affecting striatum, such as affective disorders and compulsive disorders characterized by dysfunctional motivation and habitual behavior.
Collapse
Affiliation(s)
- Ramesh Chandra
- Department of Anatomy and Neurobiology, University of Maryland School of MedicineBaltimore, MD, United States
| | - Mary Kay Lobo
- Department of Anatomy and Neurobiology, University of Maryland School of MedicineBaltimore, MD, United States
| |
Collapse
|
46
|
Li MH, Underhill SM, Reed C, Phillips TJ, Amara SG, Ingram SL. Amphetamine and Methamphetamine Increase NMDAR-GluN2B Synaptic Currents in Midbrain Dopamine Neurons. Neuropsychopharmacology 2017; 42:1539-1547. [PMID: 27976681 PMCID: PMC5436114 DOI: 10.1038/npp.2016.278] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 11/29/2016] [Accepted: 12/11/2016] [Indexed: 02/07/2023]
Abstract
The psychostimulants amphetamine (AMPH) and methamphetamine (MA) are widely abused illicit drugs. Here we show that both psychostimulants acutely increase NMDA receptor (NMDAR)-mediated synaptic currents and decrease AMPA receptor (AMPAR)/NMDAR ratios in midbrain dopamine neurons. The potentiation depends on the transport of AMPH into the cell by the dopamine transporter. NMDAR-GluN2B receptor inhibitors, ifenprodil, RO 25-6981, and RO 04-5595, inhibit the potentiation without affecting basal-evoked NMDA currents, indicating that NMDAR-GluN2B receptors are activated by AMPH. A selective peptide inhibitor of AMPH-dependent trafficking of the neuronal excitatory amino acid transporter 3 (EAAT3) blocks potentiation, suggesting that EAAT3 internalization increases extracellular glutamate concentrations and activates GluN2B-containing NMDARs. Experiments with the use-dependent NMDAR blocker, MK-801, indicate that potentiated NMDARs reside on the plasma membrane and are not inserted de novo. In behavioral studies, GluN2B inhibitors reduce MA-mediated locomotor activity, without affecting basal activity. These results reveal an important interaction between dopamine and glutamatergic signaling in midbrain dopamine neurons in response to acute administration of psychostimulants.
Collapse
Affiliation(s)
- Ming-Hua Li
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Suzanne M Underhill
- National Institute of Mental Health, National Institutes of Health, Laboratory of Molecular and Cellular Neurobiology, Bethesda, MD, USA
| | - Cheryl Reed
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Tamara J Phillips
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA,VA Portland Health Care System, Portland, OR, USA
| | - Susan G Amara
- National Institute of Mental Health, National Institutes of Health, Laboratory of Molecular and Cellular Neurobiology, Bethesda, MD, USA
| | - Susan L Ingram
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, USA,Department of Neurological Surgery, Oregon Health & Science University (OHSU), 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA, Tel: 503 494 1220, Fax: 503 494 2664, E-mail:
| |
Collapse
|
47
|
Scofield MD, Heinsbroek JA, Gipson CD, Kupchik YM, Spencer S, Smith ACW, Roberts-Wolfe D, Kalivas PW. The Nucleus Accumbens: Mechanisms of Addiction across Drug Classes Reflect the Importance of Glutamate Homeostasis. Pharmacol Rev 2017; 68:816-71. [PMID: 27363441 DOI: 10.1124/pr.116.012484] [Citation(s) in RCA: 379] [Impact Index Per Article: 54.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The nucleus accumbens is a major input structure of the basal ganglia and integrates information from cortical and limbic structures to mediate goal-directed behaviors. Chronic exposure to several classes of drugs of abuse disrupts plasticity in this region, allowing drug-associated cues to engender a pathologic motivation for drug seeking. A number of alterations in glutamatergic transmission occur within the nucleus accumbens after withdrawal from chronic drug exposure. These drug-induced neuroadaptations serve as the molecular basis for relapse vulnerability. In this review, we focus on the role that glutamate signal transduction in the nucleus accumbens plays in addiction-related behaviors. First, we explore the nucleus accumbens, including the cell types and neuronal populations present as well as afferent and efferent connections. Next we discuss rodent models of addiction and assess the viability of these models for testing candidate pharmacotherapies for the prevention of relapse. Then we provide a review of the literature describing how synaptic plasticity in the accumbens is altered after exposure to drugs of abuse and withdrawal and also how pharmacological manipulation of glutamate systems in the accumbens can inhibit drug seeking in the laboratory setting. Finally, we examine results from clinical trials in which pharmacotherapies designed to manipulate glutamate systems have been effective in treating relapse in human patients. Further elucidation of how drugs of abuse alter glutamatergic plasticity within the accumbens will be necessary for the development of new therapeutics for the treatment of addiction across all classes of addictive substances.
Collapse
Affiliation(s)
- M D Scofield
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - J A Heinsbroek
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - C D Gipson
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - Y M Kupchik
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - S Spencer
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - A C W Smith
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - D Roberts-Wolfe
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - P W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| |
Collapse
|
48
|
George O, Hope BT. Cortical and amygdalar neuronal ensembles in alcohol seeking, drinking and withdrawal. Neuropharmacology 2017; 122:107-114. [PMID: 28435008 DOI: 10.1016/j.neuropharm.2017.04.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 04/17/2017] [Accepted: 04/19/2017] [Indexed: 01/06/2023]
Abstract
Alcohol induces many alterations in the brain that are thought to contribute to alcohol addiction. Most of the known alterations are induced in all neurons of a brain area or all neurons of a given cell type, regardless of whether they were activated during behavior. While these alterations can have important modulatory effects on behavior, they cannot explain why animals respond specifically to alcohol-paired cues as opposed to all other non-paired cues and evoke highly specific goal-directed learned responses in models of drug craving. As an alternative, we hypothesize another class of alterations that are induced only within sparsely distributed patterns of neurons, called neuronal ensembles, that are selectively activated by alcohol-specific cues during behavior and encode the long-term memories underlying these learned behaviors in animal models of alcohol addiction. Here we review recent studies and techniques used to identify the role of neuronal ensembles in animal models of different phases of the alcohol addiction cycle. This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Olivier George
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Bruce T Hope
- National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
49
|
Changes in Appetitive Associative Strength Modulates Nucleus Accumbens, But Not Orbitofrontal Cortex Neuronal Ensemble Excitability. J Neurosci 2017; 37:3160-3170. [PMID: 28213443 DOI: 10.1523/jneurosci.3766-16.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 01/19/2017] [Accepted: 01/29/2017] [Indexed: 12/20/2022] Open
Abstract
Cues that predict the availability of food rewards influence motivational states and elicit food-seeking behaviors. If a cue no longer predicts food availability, then animals may adapt accordingly by inhibiting food-seeking responses. Sparsely activated sets of neurons, coined "neuronal ensembles," have been shown to encode the strength of reward-cue associations. Although alterations in intrinsic excitability have been shown to underlie many learning and memory processes, little is known about these properties specifically on cue-activated neuronal ensembles. We examined the activation patterns of cue-activated orbitofrontal cortex (OFC) and nucleus accumbens (NAc) shell ensembles using wild-type and Fos-GFP mice, which express green fluorescent protein (GFP) in activated neurons, after appetitive conditioning with sucrose and extinction learning. We also investigated the neuronal excitability of recently activated, GFP+ neurons in these brain areas using whole-cell electrophysiology in brain slices. Exposure to a sucrose cue elicited activation of neurons in both the NAc shell and OFC. In the NAc shell, but not the OFC, these activated GFP+ neurons were more excitable than surrounding GFP- neurons. After extinction, the number of neurons activated in both areas was reduced and activated ensembles in neither area exhibited altered excitability. These data suggest that learning-induced alterations in the intrinsic excitability of neuronal ensembles is regulated dynamically across different brain areas. Furthermore, we show that changes in associative strength modulate the excitability profile of activated ensembles in the NAc shell.SIGNIFICANCE STATEMENT Sparsely distributed sets of neurons called "neuronal ensembles" encode learned associations about food and cues predictive of its availability. Widespread changes in neuronal excitability have been observed in limbic brain areas after associative learning, but little is known about the excitability changes that occur specifically on neuronal ensembles that encode appetitive associations. Here, we reveal that sucrose cue exposure recruited a more excitable ensemble in the nucleus accumbens, but not orbitofrontal cortex, compared with their surrounding neurons. This excitability difference was not observed when the cue's salience was diminished after extinction learning. These novel data provide evidence that the intrinsic excitability of appetitive memory-encoding ensembles is regulated differentially across brain areas and adapts dynamically to changes in associative strength.
Collapse
|
50
|
Shukla A, Beroun A, Panopoulou M, Neumann PA, Grant SG, Olive MF, Dong Y, Schlüter OM. Calcium-permeable AMPA receptors and silent synapses in cocaine-conditioned place preference. EMBO J 2017; 36:458-474. [PMID: 28077487 DOI: 10.15252/embj.201695465] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 11/04/2016] [Accepted: 12/12/2016] [Indexed: 01/15/2023] Open
Abstract
Exposure to cocaine generates silent synapses in the nucleus accumbens (NAc), whose eventual unsilencing/maturation by recruitment of calcium-permeable AMPA-type glutamate receptors (CP-AMPARs) after drug withdrawal results in profound remodeling of NAc neuro-circuits. Silent synapse-based NAc remodeling was shown to be critical for several drug-induced behaviors, but its role in acquisition and retention of the association between drug rewarding effects and drug-associated contexts has remained unclear. Here, we find that the postsynaptic proteins PSD-93, PSD-95, and SAP102 differentially regulate excitatory synapse properties in the NAc. Mice deficient for either of these scaffold proteins exhibit distinct maturation patterns of silent synapses and thus provided instructive animal models to examine the role of NAc silent synapse maturation in cocaine-conditioned place preference (CPP). Wild-type and knockout mice alike all acquired cocaine-CPP and exhibited increased levels of silent synapses after drug-context conditioning. However, the mice differed in CPP retention and CP-AMPAR incorporation. Collectively, our results indicate that CP-AMPAR-mediated maturation of silent synapses in the NAc is a signature of drug-context association, but this maturation is not required for establishing or retaining cocaine-CPP.
Collapse
Affiliation(s)
- Avani Shukla
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Psychiatry and Psychotherapy, University Medical Center, Göttingen, Germany.,Cluster of Excellence "Nanoscale Microscopy and Molecular Physiology of the Brain", University Medical Center, Göttingen, Germany.,Göttingen Graduate School for Neurosciences and Molecular Biosciences, Göttingen, Germany
| | - Anna Beroun
- Cluster of Excellence "Nanoscale Microscopy and Molecular Physiology of the Brain", University Medical Center, Göttingen, Germany.,Laboratory of Neurobiology, The Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Myrto Panopoulou
- Department of Psychiatry and Psychotherapy, University Medical Center, Göttingen, Germany.,Cluster of Excellence "Nanoscale Microscopy and Molecular Physiology of the Brain", University Medical Center, Göttingen, Germany.,Göttingen Graduate School for Neurosciences and Molecular Biosciences, Göttingen, Germany
| | - Peter A Neumann
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Seth Gn Grant
- Genes to Cognition Programme, Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - M Foster Olive
- Department of Psychology, Arizona State University, Tempe, AZ, USA
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Oliver M Schlüter
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA .,Department of Psychiatry and Psychotherapy, University Medical Center, Göttingen, Germany.,Cluster of Excellence "Nanoscale Microscopy and Molecular Physiology of the Brain", University Medical Center, Göttingen, Germany
| |
Collapse
|