1
|
Tarmati V, Sepe A, Accoto A, Conversi D, Laricchiuta D, Panuccio A, Canterini S, Fiorenza MT, Cabib S, Orsini C. Genotype-dependent functional role of the anterior and posterior paraventricular thalamus in pavlovian conditioned approach. Psychopharmacology (Berl) 2025; 242:1275-1289. [PMID: 39663249 DOI: 10.1007/s00213-024-06726-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024]
Abstract
RATIONALE The specific location of deviations from normative models of brain function varies considerably across individuals with the same diagnoses. However, as pathological processes are distributed across interconnected systems, this heterogeneity of individual brain deviations may also reveal similarities and differences between disorders. The paraventricular nucleus of the thalamus (PVT) is a potential switcher to various behavioral responses where functionally distinct cell types exist across its antero-posterior axis. OBJECTIVES This study aimed to test the hypothesis that genotype-dependent differences in the anterior and posterior PVT subregions (aPVT and pPVT) are involved in the Sign-tracking (ST) behavior expressed by C57BL/6J (C57) and DBA/2J (DBA) inbred mice. METHODS Based on previous findings, male mice of the two strains were tested at ten weeks of age. The density of c-Fos immunoreactivity along the antero-posterior axis of PVT was assessed following the expression of ST behavior. Selective excitotoxic lesions of the aPVT or the pPVT by the NMDA infusion were performed prior to development of ST behavior. Finally, the distribution of neuronal populations expressing the Drd2 and Gal genes (D2R + and Gal +) was measured by in situ hybridization (ISH). RESULTS The involvement of PVT subregions in ST behavior is strain-specific, as aPVT is crucial for ST acquisition in DBA mice while pPVT is crucial for C57 mice. Despite similar antero-posterior distribution of D2R + and Gal + neurons, density of D2R + neurons differentiate aPVT in C57 and DBA mice. CONCLUSIONS These genotype-dependent results offer valuable insights into the nuanced organization of brain networks and individual variability in behavioral responses.
Collapse
Affiliation(s)
- Valeria Tarmati
- Department of Psychology, Sapienza University of Rome, Rome, Italy.
| | - Andrea Sepe
- PhD Program in Behavioral Neuroscience, Department of Psychology, Sapienza University of Rome, Rome, Italy
| | | | - David Conversi
- Department of Psychology, Sapienza University of Rome, Rome, Italy
| | - Daniela Laricchiuta
- Department of Philosophy, Social Sciences & Education, University of Perugia, Perugia, Italy
| | | | - Sonia Canterini
- Department of Psychology, Sapienza University of Rome, Rome, Italy
| | | | - Simona Cabib
- Department of Psychology, Sapienza University of Rome, Rome, Italy
- Fondazione Santa Lucia IRCCS, Rome, Rome, Italy
| | - Cristina Orsini
- Department of Psychology, Sapienza University of Rome, Rome, Italy
- Fondazione Santa Lucia IRCCS, Rome, Rome, Italy
| |
Collapse
|
2
|
Yao WD, Wu X, Kobeissi A, Phillips H, Dai H. A Prefrontal Cortex-Nucleus Accumbens Circuit Attenuates Cocaine-conditioned Place Preference Memories. RESEARCH SQUARE 2025:rs.3.rs-6355343. [PMID: 40386386 PMCID: PMC12083646 DOI: 10.21203/rs.3.rs-6355343/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
The infralimbic (IL) subregion of the prefrontal cortex (PFC), via its descending projection to the nucleus accumbens (NAc), inhibits cue-induced drug seeking and reinstatement, but the underlying mechanisms are not fully understood. Here we show that the activity of IL layer 5 pyramidal neurons projecting to the NAc shell (IL-NAcSh neurons) suppresses cocaine-associated memories. Following repeated cocaine exposures in a conditioned place preference paradigm, IL-NAcSh neurons anatomically traced by fluorescent Retrobeads undergo prolonged decrease of membrane excitability, lasting for at least 15 days after cocaine withdrawal. This persistent IL-NAcSh neuron hypoexcitability is accompanied by an increase in the rheobase, an increase in the afterhyperpolarization potential, and a decrease in the membrane input resistance. This cocaine induced neuroadapation in intrinsic excitability is not observed in prelimibic cortex neurons projecting to the NAc core (PL-NAcCo neurons), a separate descending circuit thought to promote cue-triggered drug seeking. Chemogenetic restoration of IL-NAcSh neuron activity extinguishes both the acquisition and retention of cocaine conditioned place preference memories. Our results provide direct support for the notion that the IL-NAcSh circuit serves to extinct drug associated memories and restoring the drug impaired excitability of IL-NAcSh neurons has the potential to mitigate drug-cue association memories and drug seeking.
Collapse
|
3
|
Bishir M, Huang W, Sariyer IK, Chang SL. Gender dependent modulation of opioid dependance genes and signaling pathways in HIV-1 Transgenic rats at morphine tolerance. J Neurovirol 2025:10.1007/s13365-025-01257-8. [PMID: 40332726 DOI: 10.1007/s13365-025-01257-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 04/07/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025]
Abstract
Misuse of opioids is a major comorbidity in people with HIV (PWH). Neurological abnormalities and opioid addiction seen in PWH involve the interplay among signaling pathways. However, the impact of HIV proteins on morphine dependence is understudied. We aimed to understand the modulation of the opioid dependence genes and signaling pathways in the striatum (Str) and prefrontal cortex (PFC) of PWH. HIV-1 transgenic (HIV-1Tg) rats and F344 control animals were given 2 and 4 pellets of morphine (75-mg/pellet)/placebo on Days 1 and 2, respectively, via subcutaneous implantation. On Day 5, at morphine tolerance the rats were sacrificed, Str and PFC were collected for RNA isolation and cDNA preparation. A PCR-array was used to examine the expression of the 65 opioid dependance genes. Varying numbers of genes were significantly upregulated in the Str and PFC of morphine treated rats. Fold change values were uploaded to QIAGEN Ingenuity Pathway Analysis, to study the signaling pathways associated with the treatment conditions. CREB signaling in neurons and Neuroinflammation signaling pathway were highly activated in the Str of both male and female HIV-1Tg rats given morphine. Gαq signaling and S100 family signaling were activated in female HIV-1Tg rats received morphine. Similarly, in the PFC, synthesis of IP3, CREB Signaling in neurons, Gαq signaling in males and CREB Signaling in neuron, and Gαq signaling in females were activated. Using bioinformatic analysis, we identified key signaling pathways and gender dependent changes in the opioid dependent gene expression and pathway enrichment of HIV-1Tg rats at morphine tolerance.
Collapse
Affiliation(s)
- Muhammed Bishir
- Institute of NeuroImmune Pharmacology, Seton Hall University, South Orange, NJ, 07079, USA
| | - Wenfei Huang
- Institute of NeuroImmune Pharmacology, Seton Hall University, South Orange, NJ, 07079, USA
| | - Ilker K Sariyer
- Department of Microbiology, Immunology, and Inflammation, Center for Neurovirology and GeneEditing, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA.
| | - Sulie L Chang
- Institute of NeuroImmune Pharmacology, Seton Hall University, South Orange, NJ, 07079, USA.
| |
Collapse
|
4
|
Hwang EK, Wunsch AM, Wolf ME. Retinoic acid-mediated homeostatic plasticity drives cell type-specific CP-AMPAR accumulation in nucleus accumbens core and incubation of cocaine craving. Mol Psychiatry 2025:10.1038/s41380-025-03026-9. [PMID: 40316677 DOI: 10.1038/s41380-025-03026-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 03/06/2025] [Accepted: 04/08/2025] [Indexed: 05/04/2025]
Abstract
Incubation of cocaine craving, a translationally relevant model for the persistence of drug craving during abstinence, ultimately depends on strengthening of nucleus accumbens core (NAcc) synapses through synaptic insertion of homomeric GluA1 Ca2+-permeable AMPA receptors (CP-AMPARs). Here we tested the hypothesis that CP-AMPAR upregulation results from a form of homeostatic plasticity, previously characterized in vitro and in other brain regions, that depends on retinoic acid (RA) signaling in dendrites. Under normal conditions, ongoing synaptic transmission maintains intracellular Ca2+ at levels sufficient to suppress RA synthesis. Prolonged blockade of neuronal activity results in disinhibition of RA synthesis, leading to increased GluA1 translation and synaptic insertion of homomeric GluA1 CP-AMPARs. Using slice recordings, we found that increasing RA signaling in NAcc medium spiny neurons (MSN) from drug-naïve rats rapidly upregulates CP-AMPARs. This is observed only in MSN expressing the D1 dopamine receptor. In MSN recorded from rats that have undergone incubation of craving, we observe CP-AMPAR upregulation in D1 MSN (but not D2 MSN) and the effect of exogenous RA application is occluded in these D1 MSN. Instead, interruption of RA signaling in the slice normalizes the incubation-associated elevation of synaptic CP-AMPARs. Paralleling this in vitro finding, interruption of RA signaling in the NAcc of 'incubated rats' normalizes elevated cue-induced cocaine seeking back to non-incubated levels. These results suggest that RA signaling becomes tonically active in the NAcc during cocaine withdrawal and, by maintaining elevated CP-AMPAR levels, contributes to the incubation of cocaine craving.
Collapse
Affiliation(s)
- Eun-Kyung Hwang
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97212, USA
| | - Amanda M Wunsch
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97212, USA
- National Center for Wellness and Recovery, Oklahoma State University Center for Health Sciences, Tulsa, OK, 74107, USA
| | - Marina E Wolf
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97212, USA.
| |
Collapse
|
5
|
Mount KA, Kuhn HM, Hwang EK, Beutler MM, Wolf ME. Incubation of oxycodone craving is associated with CP-AMPAR upregulation in D1 and D2 receptor-expressing medium spiny neurons in nucleus accumbens core and shell. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.06.647399. [PMID: 40236121 PMCID: PMC11996497 DOI: 10.1101/2025.04.06.647399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
A major problem in treating opioid use disorder is persistence of craving after protracted abstinence. This has been modeled in rodents using the incubation of craving model, in which cue-induced drug seeking increases over the first weeks of abstinence from drug self-administration and then remains high for an extended period. Incubation has been reported for several opioids, including oxycodone, but little is known about underlying synaptic plasticity. In contrast, it is well established that incubation of cocaine and methamphetamine craving depends on strengthening of glutamate synapses in the nucleus accumbens (NAc) through incorporation of calcium-permeable AMPARs (CP-AMPARs). CP-AMPARs have higher conductance than the calcium-impermeable AMPARs that mediate NAc excitatory transmission in drug-naïve animals, as well as other distinct properties. Here we examined AMPAR transmission in medium spiny neurons (MSN) of NAc core and shell subregions in rats during forced abstinence from extended-access oxycodone self-administration. In early abstinence (prior to incubation), CP-AMPAR levels were low. After 17-33 days of abstinence (when incubation is stably plateaued), CP-AMPAR levels were significantly elevated in both subregions. These results explain the prior demonstration that infusion of a selective CP-AMPAR antagonist into NAc core or shell subregions prevents expression of oxycodone incubation. Then, using transgenic rats, we found CP-AMPAR upregulation on both D1 and D2 receptor-expressing MSN, which contrasts with selective upregulation on D1 MSN after cocaine and methamphetamine incubation. Overall, our results demonstrate a common role for CP-AMPAR upregulation in psychostimulant and oxycodone incubation, albeit with differences in MSN subtype-specificity.
Collapse
|
6
|
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 neurons, promotes neuronal excitability, and regulates cocaine reward. SCIENCE ADVANCES 2025; 11:eads4441. [PMID: 40138397 PMCID: PMC12076537 DOI: 10.1126/sciadv.ads4441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 02/20/2025] [Indexed: 03/29/2025]
Abstract
Drugs of abuse activate defined neuronal populations in reward structures such as the nucleus accumbens (NAc), which 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, mechanisms that dictate NAc neuronal recruitment remain unknown. Here, we leveraged unbiased single-nucleus transcriptional profiling and targeted in situ detection to identify Reln (encoding the secreted glycoprotein, Reelin) as a marker of cocaine-activated neuronal populations within the rat NAc. A CRISPR interference approach enabling selective Reln knockdown in the adult NAc altered expression of calcium signaling genes, promoted a transcriptional trajectory consistent with loss of cocaine sensitivity, and decreased MSN excitability. Behaviorally, Reln knockdown prevented cocaine locomotor sensitization, abolished cocaine place preference memory, and decreased cocaine self-administration behavior. These results identify Reelin as a critical mechanistic link between neuronal activation and cocaine-induced behavioral adaptations.
Collapse
Affiliation(s)
- Kasey L. Brida
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Emily T. Jorgensen
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Robert A. Phillips
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Catherine E. Newman
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jennifer J. Tuscher
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Emily K. Morring
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Morgan E. Zipperly
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lara Ianov
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
- Civitan International Research Center,
University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kelsey D. Montgomery
- Lieber Institute for Brain Development,
Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Madhavi Tippani
- Lieber Institute for Brain Development,
Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Thomas M. Hyde
- Lieber Institute for Brain Development,
Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral
Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205,
USA
- Department of Neurology, Johns Hopkins
University School of Medicine, Baltimore, MD 21205, USA
| | - Kristen R. Maynard
- Lieber Institute for Brain Development,
Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral
Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205,
USA
- Department of Neuroscience, Johns Hopkins
University School of Medicine, Baltimore, MD 21205, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development,
Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral
Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205,
USA
- Department of Neuroscience, Johns Hopkins
University School of Medicine, Baltimore, MD 21205, USA
- The Kavli Neuroscience Discovery
Institute, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jeremy J. Day
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
7
|
Wu X, Kobeissi AM, Phillips HL, Dai H, Yao WD. A Prefrontal Cortex-Nucleus Accumbens Circuit Attenuates Cocaine-conditioned Place Preference Memories. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.21.644656. [PMID: 40196555 PMCID: PMC11974754 DOI: 10.1101/2025.03.21.644656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
The infralimbic (IL) subregion of the prefrontal cortex (PFC), via its descending projection to the nucleus accumbens (NAc), inhibits cue-induced drug seeking and reinstatement, but the underlying mechanisms are not fully understood. Here we show that the intrinsic membrane excitability of IL layer 5 pyramidal neurons projecting to the NAc shell (IL-NAcSh neurons) suppresses cocaine-associated memories. Following repeated cocaine exposures in a conditioned place preference paradigm, IL-NAcSh neurons anatomically traced by fluorescent retrobeads undergo prolonged decrease of membrane excitability, lasting for at least 15 days after cocaine withdrawal. This persistent IL-NAcSh neuron hypoexcitability was accompanied by an increase in the rheobase, an increase in the afterhyperpolarization potential, and a decrease in the membrane input resistance. This cocaine induced neuroadapation in intrinsic excitability was not observed in prelimibic cortex neurons projecting to the NAc core (PL-NAcCo neurons), a separate descending circuit thought to promote cue-triggered drug seeking. Chemogenetic restoration of IL-NAcSh neuron activity extinguishes both the acquisition and retention of cocaine conditioned place preference memories. Our results provide direct support for the notion that the IL-NAcSh circuit serves to extinct drug associated memories and restoring the drug impaired excitability of IL-NAcSh neurons has the potential to mitigate drug-cue association memories and drug seeking.
Collapse
Affiliation(s)
- Xiaobo Wu
- Departments of Psychiatry & Behavioral Sciences and of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY 13210
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Jiangsu, China, 226019
| | - Aya M. Kobeissi
- Departments of Psychiatry & Behavioral Sciences and of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY 13210
| | - Hannah L. Phillips
- Departments of Psychiatry & Behavioral Sciences and of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY 13210
| | - Huihui Dai
- Departments of Psychiatry & Behavioral Sciences and of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY 13210
| | - Wei-Dong Yao
- Departments of Psychiatry & Behavioral Sciences and of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
8
|
Liu X, Huang Y, Mu L, Friedman V, Kelly TJ, Hu Y, Yuan D, Liu QS. Epac2-mediated synaptic insertion of Ca 2+-permeable AMPARs in the nucleus accumbens contributes to incubation of cocaine craving. Neuropsychopharmacology 2025; 50:620-629. [PMID: 39702576 PMCID: PMC11845495 DOI: 10.1038/s41386-024-02030-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/04/2024] [Accepted: 11/12/2024] [Indexed: 12/21/2024]
Abstract
The accumulation of GluA2-lacking Ca2+-permeable AMPARs (CP-AMPARs) in the medium spiny neurons (MSNs) of the nucleus accumbens (NAc) is required for the expression of incubation of cocaine craving. The exchange protein directly activated by cAMP (Epac) is an intracellular effector of cAMP and a guanine nucleotide exchange factor for the small GTPase Rap1. Epac2 has been implicated in the trafficking of AMPA receptors at central synapses. We tested the hypothesis that Epac2 activation contributes to the accumulation of CP-AMPARs in NAc MSNs and incubation of cocaine craving. Here we demonstrate that the selective Epac2 agonist S-220 facilitated the synaptic insertion of GluA2-lacking CP-AMPARs at excitatory synapses onto NAc MSNs. In addition, prolonged abstinence from cocaine self-administration in rats resulted in elevated Rap1-GTP levels in the NAc, implying that Epac2 is activated during incubation. Importantly, we show that AAV-mediated shRNA knockdown of Epac2 in the NAc core attenuated the accumulation of CP-AMPARs and cue-induced drug-seeking behavior after prolonged abstinence from cocaine self-administration. In contrast, acute pharmacological inhibition of Epac2 with the selective Epac2 inhibitor ESI-05 did not alter CP-AMPARs that had already accumulated during incubation, and intra-NAc application of ESI-05 did not significantly affect cue-induced drug seeking following prolonged abstinence. Taken together, these results suggest that Epac2 activation during the period of incubation, but not during cue-induced drug seeking, leads to the accumulation of CP-AMPARs in NAc MSNs, which in turn contributes to incubation of cocaine craving.
Collapse
Affiliation(s)
- Xiaojie Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Yao Huang
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Lianwei Mu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Vladislav Friedman
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Thomas J Kelly
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ying Hu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Dong Yuan
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Qing-Song Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
9
|
Chow JJ, Pitts KM, Negishi K, Madangopal R, Dong Y, Wolf ME, Shaham Y. Neurobiology of the incubation of drug craving: An update. Pharmacol Rev 2025; 77:100022. [PMID: 40148031 PMCID: PMC11964951 DOI: 10.1016/j.pharmr.2024.100022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 11/21/2024] [Indexed: 03/29/2025] Open
Abstract
Relapse to drug use is often preceded by drug craving. Clinical observations in the 1980s led clinical investigators to postulate that cue-induced cocaine craving may increase during abstinence. Over 2 decades ago, investigators identified an analogous phenomenon in rats of time-dependent increases in drug-seeking behavior during homecage abstinence and termed it incubation of cocaine craving. In 2011, we reviewed the first decade of studies on brain mechanisms of incubation of drug craving. In this review, we provide an update on incubation-related brain mechanisms from studies published since 2011. We first review studies using the standard method of incubation after homecage-forced abstinence from cocaine, methamphetamine, opioid drugs, and nicotine. Next, we review studies using newer methods to study incubation after voluntary abstinence in the drug environment. In these studies, abstinence is achieved by either providing rats alternative nondrug rewards in a choice setting or introducing rats to adverse consequences to drug seeking or taking. We then discuss translational human studies on incubation of cue-induced drug craving. We conclude by discussing several emerging topics, including sex differences in incubation of drug craving, role of sleep patterns, and similarities and differences in mechanisms of incubation of craving across drug classes and abstinence conditions. Our 2 main conclusions are as follows: (1) across drug classes, there are both similarities and differences in mechanisms of incubation of drug craving after forced abstinence, and (2) the method used to achieve abstinence (forced or voluntary) can influence the mechanisms controlling incubation of drug craving or its expression. SIGNIFICANCE STATEMENT: This article reviews results from preclinical and clinical studies published since 2011 on neurobiological mechanisms of incubation of drug craving after homecage-forced abstinence or voluntary abstinence in the drug environment. This article also reviews translational human studies on incubation of cue-induced subjective drug craving and brain response during abstinence. The results of the studies reviewed indicate that multiple brain mechanisms control incubation of drug craving after homecage-forced abstinence or voluntary abstinence.
Collapse
Affiliation(s)
- Jonathan J Chow
- Behavioral Neuroscience Research Branch, IRP/NIDA/NIH, Baltimore, Maryland.
| | - Kayla M Pitts
- Behavioral Neuroscience Research Branch, IRP/NIDA/NIH, Baltimore, Maryland
| | - Kenichiro Negishi
- Behavioral Neuroscience Research Branch, IRP/NIDA/NIH, Baltimore, Maryland
| | | | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Marina E Wolf
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon
| | - Yavin Shaham
- Behavioral Neuroscience Research Branch, IRP/NIDA/NIH, Baltimore, Maryland.
| |
Collapse
|
10
|
Guo C, Ma YY. Epac2 activation drives calcium permeable-AMPA receptor accumulation, fueling the incubation of cocaine craving. Neuropsychopharmacology 2025; 50:618-619. [PMID: 39775174 PMCID: PMC11845452 DOI: 10.1038/s41386-024-02043-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025]
Affiliation(s)
- Changyong Guo
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yao-Ying Ma
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
11
|
Namba MD, Goldberg SL, Side CM, Yadlapalli A, Curran-Alfaro CM, Kress D, Fogarty MF, Mohr ALA, Barker JM. Dissociating the incubation of appetitive and consummatory behavior in a model of oral cocaine self-administration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637922. [PMID: 39990380 PMCID: PMC11844540 DOI: 10.1101/2025.02.12.637922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Cocaine use disorder remains a persistent public health dilemma that currently lacks effective treatment strategies. One key impediment to successful treatment outcomes is increased drug craving that occurs over the course of abstinence and subsequent relapse to drug use. This phenomenon, known as the incubation of drug craving, has been modeled extensively in rodent models of intravenous drug self-administration. As commonly implemented, the design of intravenous self-administration preclinical studies precludes disentangling appetitive and consummatory behaviors as drug seeking (appetitive) and taking (consummatory) is simultaneous. Here, we employed a model of oral cocaine self-administration to interrogate the incubation of drug vs nondrug craving, where the route of administration is identical between reinforcers and appetitive and consummatory behaviors are dissociable. Oral self-administration of cocaine produced detectable levels of cocaine and its metabolite, benzoylecgonine, within the blood and brain, and blood and brain levels of both substrates correlated with cocaine consumption. When tested for seeking-(lever pressing) and taking-related (magazine head entries) behavior after 1 or 21 days of forced abstinence from cocaine or saccharin, we observed incubation of lever pressing among cocaine-administering mice and incubation of magazine entries among saccharin-administering mice. These behavioral changes were accompanied by reduced expression of the glial glutamate transporter GLT-1 within the nucleus accumbens (NAc) of cocaine self-administering mice, regardless of abstinence. Altogether, these results underscore the utility of this model of cocaine self-administration, highlight the conserved nature of incubated cocaine seeking across routes of administration, and demonstrate the dissociable neurobehavioral sequelae of the incubation of reward seeking across reinforcer types.
Collapse
|
12
|
Cheng Z, Zhao F, Piao J, Yang W, Cui R, Li B. Rasd2 regulates depression-like behaviors via DRD2 neurons in the prelimbic cortex afferent to nucleus accumbens core circuit. Mol Psychiatry 2025; 30:435-449. [PMID: 39097664 PMCID: PMC11746134 DOI: 10.1038/s41380-024-02684-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 07/18/2024] [Accepted: 07/24/2024] [Indexed: 08/05/2024]
Abstract
Depressive symptoms, such as anhedonia, decreased social interaction, and lack of motivation, implicate brain reward systems in the pathophysiology of depression. Exposure to chronic stress impairs the function of brain reward circuits and is well-known to be involved in the etiology of depression. A transcriptomic analysis found that stress alters the expression of Rasd2 in mice prefrontal cortex (PFC). Similarly, in our previous study, acute fasting decreased Rasd2 expression in mice PFC, and RASD2 modulated dopamine D2 receptor (DRD2)-mediated antidepressant-like effects in ovariectomized mice. This research suggests the role of RASD2 in stress-induced depression and its underlying neural mechanisms that require further investigation. Here, we show that 5-day unpredictable mild stress (5-d UMS) exposure reduces RASD2 expression in both the nucleus accumbens (NAc) and medial prefrontal cortex (mPFC) of mice, while overexpression (but not knock-down) of Rasd2 in the NAc core (NAcc) alleviates 5-d UMS-induced depression-like behaviors and activates the DRD2-cAMP-PKA-DARPP-32 signaling pathway. Further studies investigated neuronal projections between the mPFC (Cg1, PrL, and IL) and NAcc, labeled by the retrograde tracer Fluorogold. Depression-like behaviors induced by 5-d UMS were only related to inhibition of the PrL-NAcc circuit. DREADD (Designer receptors exclusively activated by designer drug) analysis found that the activation of PrL-NAcc glutaminergic projection alleviated depression-like behaviors and increased DRD2- and RASD2-positive neurons in the NAcc. Using Drd2-cre transgenic mice, we constructed mice with Rasd2 overexpression in DRD2PrL-NAcc neurons, finding that Rasd2 overexpression ameliorated 5-d UMS-induced depression-like behaviors. These findings demonstrate a critical role for RASD2 modulation of DRD2PrL-NAcc neurons in 5-d UMS-induced depression-like behaviors. In addition, the study identifies a new potential strategy for precision medical treatment of depression.
Collapse
Affiliation(s)
- Ziqian Cheng
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, 130041, PR China
- Engineering Lab on Screening of Antidepressant Drugs, Jilin Province Development and Reform Commission, Changchun, 130041, PR China
| | - Fangyi Zhao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, 130041, PR China
- Engineering Lab on Screening of Antidepressant Drugs, Jilin Province Development and Reform Commission, Changchun, 130041, PR China
| | - Jingjing Piao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, 130041, PR China
- Engineering Lab on Screening of Antidepressant Drugs, Jilin Province Development and Reform Commission, Changchun, 130041, PR China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, 130041, PR China
- Engineering Lab on Screening of Antidepressant Drugs, Jilin Province Development and Reform Commission, Changchun, 130041, PR China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, 130041, PR China.
- Engineering Lab on Screening of Antidepressant Drugs, Jilin Province Development and Reform Commission, Changchun, 130041, PR China.
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, 130041, PR China.
- Engineering Lab on Screening of Antidepressant Drugs, Jilin Province Development and Reform Commission, Changchun, 130041, PR China.
| |
Collapse
|
13
|
Chen Y, Fu H, Korada A, Lange MA, Rayanki C, Lu T, Lai D, Fang S, Guo C, Ma YY. Decoding Secondary Motor Cortex Neuronal Activity during Cocaine Self-Administration: Insights from Longitudinal in vivo Calcium Imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.20.594996. [PMID: 38826399 PMCID: PMC11142101 DOI: 10.1101/2024.05.20.594996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Background We recently reported that the risk of cocaine relapse is linked to hyperexcitability in the secondary motor cortex (M2) after prolonged withdrawal following intravenous self-administration (IVSA). However, the neuronal mechanisms underlying drug-taking behaviors and the response of M2 neurons to contingent drug delivery remain poorly understood. Methods Mice received cocaine as reinforcement (RNF) following active lever presses (ALP) but not inactive lever presses (ILP). Using in vivo calcium imaging during cocaine IVSA, we tracked M2 neuronal activity with single-cell resolution. We then analyzed Ca 2+ transients in M2 at the early vs . late stages during the 1-hr daily sessions on IVSA Day 1 and Day 5. Results M2 neurons adapted to both operant behaviors and drug exposure history. Specifically, saline mice showed a reduction in both saline taking behaviors and Ca 2+ transient frequency with the 1-hr session. In contrast, cocaine mice maintained high ALP and RNF counts, with increased Ca 2+ transient frequency and amplitude on Day 1, persisting through Day 5. Compared to saline controls, cocaine mice exhibited a lower % of positively responsive neurons (Pos) and higher % of negatively responsive neurons (Neg) before ALP and after RNF, a difference not seen before ILP. Furthermore, as drug-taking behaviors progressed during the daily session, cocaine mice showed greater neuronal engagement with a larger population, particularly linked to ALP and RNF, with reduced overlap in neurons associated with ILP. Conclusion The M2 undergoes dynamic neuronal adaptations during early drug-taking behaviors, indicating its role as a potential substrate mediating the persistence of drug-seeking behaviors in cocaine relapse.
Collapse
|
14
|
Wolf ME. Targeting Neuroplasticity in Substance Use Disorders: Implications for Therapeutics. Annu Rev Pharmacol Toxicol 2025; 65:259-280. [PMID: 39374445 PMCID: PMC11864087 DOI: 10.1146/annurev-pharmtox-061724-080548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
The last two decades have witnessed substantial advances in identifying synaptic plasticity responsible for behavioral changes in animal models of substance use disorder. We have learned the most about cocaine-induced plasticity in the nucleus accumbens and its relationship to cocaine seeking, so that is the focus in this review. Synaptic plasticity pointing to potential therapeutic targets has been identified mainly using two drug self-administration models: extinction-reinstatement and abstinence models. A relationship between cocaine seeking and potentiated AMPAR transmission in nucleus accumbens is indicated by both models. In particular, an atypical subpopulation-Ca2+-permeable or CP-AMPARs-mediates cue-induced seeking that persists even after long periods of abstinence, modeling the persistent vulnerability to relapse that represents a major challenge in treating substance use disorder. We review strategies to reverse CP-AMPAR plasticity; strategies targeting other components of excitatory synapses, including dysregulated glutamate uptake and release; and behavioral interventions that can be augmented by harnessing synaptic plasticity.
Collapse
Affiliation(s)
- Marina E Wolf
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA;
| |
Collapse
|
15
|
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 PMCID: PMC11616716 DOI: 10.1016/j.neuroscience.2024.11.005] [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: 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
|
16
|
Webb SM, Miller BW, Wroten MG, Sacramento A, Travis KO, Kippin TE, Ben-Shahar O, Szumlinski KK. Replication and extension of the subregion selectivity of glutamate-related changes within the nucleus accumbens associated with the incubation of cocaine-craving. Pharmacol Biochem Behav 2024; 245:173889. [PMID: 39389205 DOI: 10.1016/j.pbb.2024.173889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 09/10/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024]
Abstract
Cue-elicited drug-seeking behavior intensifies with the passage of time during withdrawal from drug taking and this "incubation of cocaine-craving" involves alterations in nucleus accumbens (NA) glutamate transmission. Here, we employed a combination of in vivo microdialysis and immunoblotting approaches to further examine changes in biochemical indices of glutamate transmission within NA subregions that accompany the incubation of cocaine-craving exhibited by male rats with a 10-day history of 6-h access to intravenous cocaine (0.25 mg/infusion). Immunoblotting on whole cell lysates from the core subregion (NAc core) revealed interactions between cocaine self-administration history, withdrawal and drug cue re-exposure for Homer2a/b, mGlu1, and GluN2b expression, as well as indices of Akt and ERK activity. With the exception of PKCε phosphorylation, most protein changes within the shell subregion (NAc shell) depended on drug cue re-exposure and cocaine history rather than varying in a consistent time-dependent manner. Reduced basal extracellular glutamate content was apparent only in the NAc core of cocaine-experienced rats during protracted (30 days) withdrawal and this was accompanied by a markedly blunted capacity of the mGlu1/5 agonist DHPG to elevate glutamate levels within this subregion. Finally, over-expressing neither Homer1c nor Homer2b within the NAc core during protracted cocaine withdrawal altered the magnitude of cue-elicited responding, its extinction or cocaine-primed reinstatement of drug-seeking behavior. The present findings are consistent with the extant literature implicating changes in Group 1 mGlu receptor function within the NAc core subregion as central to incubated cocaine-craving and provide further evidence against a major role for Homer proteins in gating incubated cocaine-craving. Further, our results provide novel correlational evidence implicating elevated Akt and blunted ERK activity within the NAc core as potential contributors to the expression of incubated cocaine-craving, worthy of future investigation.
Collapse
Affiliation(s)
- Sierra M Webb
- Department of Psychological and Brain Sciences, MC-9660, University of California Santa Barbara, Santa Barbara, CA 93106-9660, United States of America
| | - Bailey W Miller
- Department of Psychological and Brain Sciences, MC-9660, University of California Santa Barbara, Santa Barbara, CA 93106-9660, United States of America
| | - Melissa G Wroten
- Department of Psychological and Brain Sciences, MC-9660, University of California Santa Barbara, Santa Barbara, CA 93106-9660, United States of America
| | - Arianne Sacramento
- Department of Psychological and Brain Sciences, MC-9660, University of California Santa Barbara, Santa Barbara, CA 93106-9660, United States of America
| | - Katherine O Travis
- Department of Psychological and Brain Sciences, MC-9660, University of California Santa Barbara, Santa Barbara, CA 93106-9660, United States of America
| | - Tod E Kippin
- Department of Psychological and Brain Sciences, MC-9660, University of California Santa Barbara, Santa Barbara, CA 93106-9660, United States of America; Department of Molecular, Cellular and Developmental Biology and the Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA 93106, United States of America
| | - Osnat Ben-Shahar
- Department of Psychological and Brain Sciences, MC-9660, University of California Santa Barbara, Santa Barbara, CA 93106-9660, United States of America
| | - Karen K Szumlinski
- Department of Psychological and Brain Sciences, MC-9660, University of California Santa Barbara, Santa Barbara, CA 93106-9660, United States of America; Department of Molecular, Cellular and Developmental Biology and the Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA 93106, United States of America.
| |
Collapse
|
17
|
Kuhn HM, Serrano LC, Stys GA, Smith BL, Speckmaier J, Dawson BD, Murray BR, He J, Robison AJ, Eagle AL. Lateral entorhinal cortex neurons that project to nucleus accumbens mediate contextual associative memory. Learn Mem 2024; 31:a054026. [PMID: 39592189 PMCID: PMC11606517 DOI: 10.1101/lm.054026.124] [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: 06/04/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024]
Abstract
The lateral entorhinal cortex (LEC) contains glutamatergic projections that innervate the nucleus accumbens (NAc) and may be involved in the encoding of contextual associations with both positive and negative valences, such as those encountered in drug cues or fear conditioning. To determine whether LEC-NAc neurons are activated by the encoding and recall of contexts associated with cocaine or footshock, we measured c-fos expression in these neurons and found that LEC-NAc neurons are activated in both contexts. Specifically, activation patterns of the LEC-NAc were observed in a novel context and reexposure to the same context, highlighting the specific role for LEC-NAc neurons in encoding rather than the valence of a specific event-related memory. Using a combination of circuit-specific chemogenetic tools and behavioral assays, we selectively inactivated LEC-NAc neurons in mice during the encoding and retrieval of memories of contexts associated with cocaine or footshock. Chemogenetic inactivation of LEC-NAc neurons impaired the formation of both positive and negative context-associated memories without affecting the retrieval of an established memory. This finding suggests a critical role for this circuit in the initial encoding of contextual associations. In summary, LEC-NAc neurons facilitate the encoding of contextual information, guiding motivational behaviors without directly mediating the hedonic or aversive properties of these associations.
Collapse
Affiliation(s)
- Hayley M Kuhn
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, USA
| | | | - Grace A Stys
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, USA
| | - Brianna L Smith
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, USA
| | | | | | - Brooklynn R Murray
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, USA
| | - Jin He
- Department of Biochemistry, Michigan State University, East Lansing, Michigan 48824, USA
| | - Alfred J Robison
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, USA
| | - Andrew L Eagle
- Department of Neuroscience, The University of Texas at Dallas, Richardson, Texas 75080, USA
| |
Collapse
|
18
|
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
|
19
|
Hwang EK, Wunsch AM, Wolf ME. Retinoic acid-mediated homeostatic plasticity drives cell type-specific CP-AMPAR accumulation in nucleus accumbens core and incubation of cocaine craving. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.611703. [PMID: 39314388 PMCID: PMC11419102 DOI: 10.1101/2024.09.12.611703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Incubation of cocaine craving, a translationally relevant model for the persistence of drug craving during abstinence, ultimately depends on strengthening of nucleus accumbens core (NAcc) synapses through synaptic insertion of homomeric GluA1 Ca2+-permeable AMPA receptors (CP-AMPARs). Here we tested the hypothesis that CP-AMPAR upregulation results from a form of homeostatic plasticity, previously characterized in vitro and in other brain regions, that depends on retinoic acid (RA) signaling in dendrites. Under normal conditions, ongoing synaptic transmission maintains intracellular Ca2+ at levels sufficient to suppress RA synthesis. Prolonged blockade of neuronal activity results in disinhibition of RA synthesis, leading to increased GluA1 translation and synaptic insertion of homomeric GluA1 CP-AMPARs. Using slice recordings, we found that increasing RA signaling in NAcc medium spiny neurons (MSN) from drug-naïve rats rapidly upregulates CP-AMPARs, and that this pathway is operative only in MSN expressing the D1 dopamine receptor. In MSN recorded from rats that have undergone incubation of craving, this effect of RA is occluded; instead, interruption of RA signaling in the slice normalizes the incubation-associated elevation of synaptic CP-AMPARs. Paralleling this in vitro finding, interruption of RA signaling in the NAcc of 'incubated rats' normalizes the incubation-associated elevation of cue-induced cocaine seeking. These results suggest that RA signaling becomes tonically active in the NAcc during cocaine withdrawal and, by maintaining elevated CP-AMPAR levels, contributes to the incubation of cocaine craving.
Collapse
Affiliation(s)
- Eun-Kyung Hwang
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, U.S.A. 97212
| | - Amanda M Wunsch
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, U.S.A. 97212
| | - Marina E Wolf
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, U.S.A. 97212
| |
Collapse
|
20
|
Kim YJ, Kim K, Lee Y, Min HW, Ko YH, Lee BR, Hur KH, Kim SK, Lee SY, Jang CG. The mutated cytoplasmic fragile X messenger ribonucleoprotein 1 (FMR1)-interacting protein 2 (CYFIP2 S968F) regulates cocaine-induced reward behaviour and plasticity in the nucleus accumbens. Br J Pharmacol 2024; 181:3327-3345. [PMID: 38751203 DOI: 10.1111/bph.16427] [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: 12/21/2023] [Revised: 04/02/2024] [Accepted: 04/25/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND AND PURPOSE Cytoplasmic fragile X messenger ribonucleoprotein 1 (FMR1)-interacting protein 2 (CYFIP2), as a component of the Wiskott-Aldrich syndrome protein family verprolin-homologous protein (WAVE) regulatory complex, is involved in actin polymerization, contributing to neuronal development and structural plasticity. Mutating serine-968 to phenylalanine (S968F) in CYFIP2 causes an altered cocaine response in mice. The neuronal mechanisms underlying this response remain unknown. EXPERIMENTAL APPROACH We performed cocaine reward-related behavioural tests and examined changes in synaptic protein phenotypes and neuronal morphology in the nucleus accumbens (NAc), using CYFIP2 S968F knock-in mice to investigate the role of CYFIP2 in regulating cocaine reward. KEY RESULTS CYFIP2 S968F mutation attenuated cocaine-induced behavioural sensitization and conditioned place preference. Cocaine-induced c-Fos was not observed in the NAc of CYFIP2 S968F knock-in mice. However, c-Fos induction was still evident in the medial prefrontal cortex (mPFC). CYFIP2 S968F mutation altered cocaine-associated CYFIP2 signalling, glutamatergic protein expression and synaptic density in the NAc following cocaine exposure. To further determine the role of CYFIP2 in NAc neuronal activity and the mPFC projecting to the NAc activity-mediating reward response, we used optogenetic tools to stimulate the NAc or mPFC-NAc pathway and observed that optogenetic activation of the NAc or mPFC-NAc pathway induced reward-related behaviours. This effect was not observed in the S968F mutation in CYFIP2. CONCLUSION AND IMPLICATIONS These results suggest that CYFIP2 plays a role in controlling cocaine-mediated neuronal function and structural plasticity in the NAc, and that CYFIP2 could serve as a target for regulating cocaine reward.
Collapse
Affiliation(s)
- Young-Jung Kim
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Kyungin Kim
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Youyoung Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hee-Won Min
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yong-Hyun Ko
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Bo-Ram Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Kwang-Hyun Hur
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Seon-Kyung Kim
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Seok-Yong Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
21
|
Wingert JC, Ramos JD, Reynolds SX, Gonzalez AE, Rose RM, Hegarty DM, Aicher SA, Bailey LG, Brown TE, Abbas AI, Sorg BA. Perineuronal Nets in the Rat Medial Prefrontal Cortex Alter Hippocampal-Prefrontal Oscillations and Reshape Cocaine Self-Administration Memories. J Neurosci 2024; 44:e0468242024. [PMID: 38991791 PMCID: PMC11340292 DOI: 10.1523/jneurosci.0468-24.2024] [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: 03/11/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024] Open
Abstract
The medial prefrontal cortex (mPFC) is a major contributor to relapse to cocaine in humans and to reinstatement in rodent models of cocaine use disorder. The output from the mPFC is potently modulated by parvalbumin (PV)-containing fast-spiking interneurons, the majority of which are surrounded by perineuronal nets. We previously showed that treatment with chondroitinase ABC (ABC) reduced the consolidation and reconsolidation of a cocaine conditioned place preference memory. However, self-administration memories are more difficult to disrupt. Here we report in male rats that ABC treatment in the mPFC attenuated the consolidation and blocked the reconsolidation of a cocaine self-administration memory. However, reconsolidation was blocked when rats were given a novel, but not familiar, type of retrieval session. Furthermore, ABC treatment prior to, but not after, memory retrieval blocked reconsolidation. This same treatment did not alter a sucrose memory, indicating specificity for cocaine-induced memory. In naive rats, ABC treatment in the mPFC altered levels of PV intensity and cell firing properties. In vivo recordings from the mPFC and dorsal hippocampus (dHIP) during the novel retrieval session revealed that ABC prevented reward-associated increases in high-frequency oscillations and synchrony of these oscillations between the dHIP and mPFC. Together, this is the first study to show that ABC treatment disrupts reconsolidation of the original memory when combined with a novel retrieval session that elicits coupling between the dHIP and mPFC. This coupling after ABC treatment may serve as a fundamental signature for how to disrupt reconsolidation of cocaine memories and reduce relapse.
Collapse
Affiliation(s)
- Jereme C Wingert
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, Oregon 97232
| | - Jonathan D Ramos
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, Oregon 97232
| | | | - Angela E Gonzalez
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, Oregon 97232
- Program in Neuroscience, Washington State University, Vancouver, Washington 98686
| | - R Mae Rose
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, Oregon 97232
| | - Deborah M Hegarty
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon 97239
| | - Sue A Aicher
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon 97239
| | - Lydia G Bailey
- Program in Neuroscience, Washington State University, Pullman, Washington 99164
| | - Travis E Brown
- Program in Neuroscience, Washington State University, Pullman, Washington 99164
| | - Atheir I Abbas
- Departments of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon 97239
- Psychiatry, Oregon Health & Science University, Portland, Oregon 97239
- Research Division, VA Portland Health Care System, Portland, Oregon 97239
| | - Barbara A Sorg
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, Oregon 97232
- Program in Neuroscience, Washington State University, Vancouver, Washington 98686
| |
Collapse
|
22
|
Xu Y, Lin Y, Yu M, Zhou K. The nucleus accumbens in reward and aversion processing: insights and implications. Front Behav Neurosci 2024; 18:1420028. [PMID: 39184934 PMCID: PMC11341389 DOI: 10.3389/fnbeh.2024.1420028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/26/2024] [Indexed: 08/27/2024] Open
Abstract
The nucleus accumbens (NAc), a central component of the brain's reward circuitry, has been implicated in a wide range of behaviors and emotional states. Emerging evidence, primarily drawing from recent rodent studies, suggests that the function of the NAc in reward and aversion processing is multifaceted. Prolonged stress or drug use induces maladaptive neuronal function in the NAc circuitry, which results in pathological conditions. This review aims to provide comprehensive and up-to-date insights on the role of the NAc in motivated behavior regulation and highlights areas that demand further in-depth analysis. It synthesizes the latest findings on how distinct NAc neuronal populations and pathways contribute to the processing of opposite valences. The review examines how a range of neuromodulators, especially monoamines, influence the NAc's control over various motivational states. Furthermore, it delves into the complex underlying mechanisms of psychiatric disorders such as addiction and depression and evaluates prospective interventions to restore NAc functionality.
Collapse
Affiliation(s)
| | | | | | - Kuikui Zhou
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| |
Collapse
|
23
|
Chalhoub RM, Testen A, Hopkins J, Carthy C, Kalivas PW. Formation of an Enduring Ensemble of Accumbens Neurons Leads to Prepotent Seeking for Cocaine Over Natural Reward Cues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.05.606522. [PMID: 39149274 PMCID: PMC11326163 DOI: 10.1101/2024.08.05.606522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Neuronal activity in the nucleus accumbens core (NAcore) is necessary for reward-seeking behaviors. We hypothesized that the differential encoding of natural and drug rewards in the NAcore contributes to substance use disorder. We leveraged single-cell calcium imaging of dopamine D1- and D2-receptor-expressing medium spiny neurons (MSNs) in the NAcore of mice to examine differences between sucrose and cocaine rewarded (self-administration) and unrewarded (abstinent and cue-induced) seeking. Activity was time-locked to nose-poking for reward, clustered, and compared between sucrose and cocaine. Only in cocaine-trained mice were excited D1-MSNs securely stable, capable of decoding nose-poking in all rewarded and unrewarded sessions and correlated with the intensity of nose-poking for unrewarded seeking. Furthermore, D1-MSNs formed a stable ensemble predictive of seeking behavior after extended cocaine, but not sucrose abstinence. The excited D1-MSN ensemble uniquely drives cue-induced cocaine seeking and may contribute to why drug seeking is prepotent over natural reward seeking in cocaine use disorder.
Collapse
Affiliation(s)
- Reda M Chalhoub
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
- Medical Scientist Training Program, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Anze Testen
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jordan Hopkins
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Camille Carthy
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
- Ralph Johnson Veterans Administration, Charleston, South Carolina, USA
| |
Collapse
|
24
|
De Almeida SS, Drinkuth CR, Sartor GC. Comparing withdrawal- and anxiety-like behaviors following oral and subcutaneous oxycodone administration in C57BL/6 mice. Behav Pharmacol 2024; 35:269-279. [PMID: 38847447 PMCID: PMC11226370 DOI: 10.1097/fbp.0000000000000780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
Excessive prescribing and misuse of prescription opioids, such as oxycodone, significantly contributed to the current opioid crisis. Although oxycodone is typically consumed orally by humans, parenteral routes of administration have primarily been used in preclinical models of oxycodone dependence. To address this issue, more recent studies have used oral self-administration procedures to study oxycodone seeking and withdrawal in rodents. Behavioral differences, however, following oral oxycodone intake versus parenteral oxycodone administration remain unclear. Thus, the goal of the current studies was to compare anxiety- and withdrawal-like behaviors using established opioid dependence models of either home cage oral intake of oxycodone (0.5 mg/ml) or repeated subcutaneous (s.c.) injections of oxycodone (10 mg/kg) in male and female mice. Here, mice received 10 days of oral or s.c. oxycodone administration, and following 72 h of forced abstinence, anxiety- and withdrawal-like behaviors were measured using elevated zero maze, open field, and naloxone-induced precipitated withdrawal procedures. Global withdrawal scores were increased to a similar degree following oral and s.c. oxycodone use, while both routes of oxycodone administration had minimal effects on anxiety-like behaviors. When examining individual withdrawal-like behaviors, mice receiving s.c. oxycodone exhibited more paw tremors and jumps during naloxone-induced precipitated withdrawal compared with oral oxycodone mice. These results indicate that both models of oxycodone administration are sufficient to elevate global withdrawal scores, but, when compared with oral consumption, s.c. oxycodone injections yielded more pronounced effects on some withdrawal-like behaviors.
Collapse
Affiliation(s)
| | | | - Gregory C. Sartor
- Department of Pharmaceutical Sciences, Institute for the Brain and Cognitive Sciences (IBACS), University of Connecticut, Storrs, CT 06269
| |
Collapse
|
25
|
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
|
26
|
Namba MD, Xie Q, Park K, Jackson JG, Barker JM. EcoHIV Infection Modulates the Effects of Cocaine Exposure Pattern and Abstinence on Cocaine Seeking and Neuroimmune Protein Expression in Male Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.15.589615. [PMID: 38659915 PMCID: PMC11042347 DOI: 10.1101/2024.04.15.589615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Cocaine use disorders (CUDs) and human immunodeficiency virus (HIV) remain persistent public health dilemmas throughout the world. One major hurdle for treating CUD is the increase in cocaine craving and seeking behavior that occurs over a protracted period of abstinence, an effect known as the incubation of craving. Little is known about how HIV may modulate this process. Thus, we sought to examine the impact of chronic HIV infection on the incubation of cocaine craving and associated changes in the central and peripheral immune systems. Here, mice were inoculated with EcoHIV, which is a chimeric HIV-1 construct that produces chronic HIV infection in mice. EcoHIV- and sham-infected mice were conditioned with cocaine daily or intermittently in a conditioned place preference (CPP) paradigm, followed by 1 or 21 days of forced abstinence prior to assessing preference for the cocaine-paired chamber. Under both conditioning regimens, sham mice exhibited incubation of cocaine CPP after 21 days of abstinence. EcoHIV-infected mice conditioned daily with cocaine showed enhanced cocaine seeking at both abstinence timepoints, whereas infected mice conditioned intermittently showed a reversal of the incubation effect, with higher cocaine seeking after 1 day of abstinence compared to 21 days. Analysis of corticolimbic CX3CL1-CX3CR1 and glutamate receptor expression revealed alterations in medial prefrontal cortex (mPFC) CX3CL1 and nucleus accumbens (NAc) GluN2A receptors that correlated with cocaine seeking following daily cocaine exposure. Moreover, examination of peripheral immune markers showed that the effect of abstinence and EcoHIV infection on these measures depended on the cocaine exposure regimen. Altogether, these results highlight the importance of cocaine abstinence and exposure pattern as critical variables that modulate HIV-associated neuroimmune outcomes and relapse vulnerability.
Collapse
Affiliation(s)
- Mark D. Namba
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Qiaowei Xie
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
- Graduate Program in Pharmacology and Physiology, College of Medicine, Drexel University
| | - Kyewon Park
- Center for AIDS Research (CFAR), University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua G. Jackson
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Jacqueline M. Barker
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| |
Collapse
|
27
|
Wang Y, Li D, Widjaja J, Guo R, Cai L, Yan R, Ozsoy S, Allocca G, Fang J, Dong Y, Tseng GC, Huang C, Huang YH. An EEG Signature of MCH Neuron Activities Predicts Cocaine Seeking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.586887. [PMID: 38586019 PMCID: PMC10996698 DOI: 10.1101/2024.03.27.586887] [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
Background Identifying biomarkers that predict substance use disorder (SUD) propensity may better strategize anti-addiction treatment. The melanin-concentrating hormone (MCH) neurons in the lateral hypothalamus (LH) critically mediates interactions between sleep and substance use; however, their activities are largely obscured in surface electroencephalogram (EEG) measures, hindering the development of biomarkers. Methods Surface EEG signals and real-time Ca2+ activities of LH MCH neurons (Ca2+MCH) were simultaneously recorded in male and female adult rats. Mathematical modeling and machine learning were then applied to predict Ca2+MCH using EEG derivatives. The robustness of the predictions was tested across sex and treatment conditions. Finally, features extracted from the EEG-predicted Ca2+MCH either before or after cocaine experience were used to predict future drug-seeking behaviors. Results An EEG waveform derivative - a modified theta-to-delta ratio (EEG Ratio) - accurately tracks real-time Ca2+MCH in rats. The prediction was robust during rapid eye movement sleep (REMS), persisted through REMS manipulations, wakefulness, circadian phases, and was consistent across sex. Moreover, cocaine self-administration and long-term withdrawal altered EEG Ratio suggesting shortening and circadian redistribution of synchronous MCH neuron activities. In addition, features of EEG Ratio indicative of prolonged synchronous MCH neuron activities predicted lower subsequent cocaine seeking. EEG Ratio also exhibited advantages over conventional REMS measures for the predictions. Conclusions The identified EEG Ratio may serve as a non-invasive measure for assessing MCH neuron activities in vivo and evaluating REMS; it may also serve as a potential biomarker predicting drug use propensity.
Collapse
Affiliation(s)
- Yao Wang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | - Danyang Li
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | | | - Rong Guo
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | - Li Cai
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | - Rongzhen Yan
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | - Sahin Ozsoy
- Somnivore Pty. Ltd., Bacchus Marsh, VIC, Australia 3340
| | - Giancarlo Allocca
- Somnivore Pty. Ltd., Bacchus Marsh, VIC, Australia 3340
- Department of Pharmacology and Therapeutics, The University of Melbourne, VIC, Australia 3010
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, VIC, Australia
| | - Jidong Fang
- Department of Psychiatry and Behavioral Health, Penn State College of Medicine, Hershey, PA 17033
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | - George C. Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | - Chengcheng Huang
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | - Yanhua H. Huang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| |
Collapse
|
28
|
Zhao J, Zhang G, Xu D. The effect of reward on motor learning: different stage, different effect. Front Hum Neurosci 2024; 18:1381935. [PMID: 38532789 PMCID: PMC10963647 DOI: 10.3389/fnhum.2024.1381935] [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: 02/06/2024] [Accepted: 02/29/2024] [Indexed: 03/28/2024] Open
Abstract
Motor learning is a prominent and extensively studied subject in rehabilitation following various types of neurological disorders. Motor repair and rehabilitation often extend over months and years post-injury with a slow pace of recovery, particularly affecting the fine movements of the distal extremities. This extended period can diminish the motivation and persistence of patients, a facet that has historically been overlooked in motor learning until recent years. Reward, including monetary compensation, social praise, video gaming, music, and virtual reality, is currently garnering heightened attention for its potential to enhance motor motivation and improve function. Numerous studies have examined the effects and attempted to explore potential mechanisms in various motor paradigms, yet they have yielded inconsistent or even contradictory results and conclusions. A comprehensive review is necessary to summarize studies on the effects of rewards on motor learning and to deduce a central pattern from these existing studies. Therefore, in this review, we initially outline a framework of motor learning considering two major types, two major components, and three stages. Subsequently, we summarize the effects of rewards on different stages of motor learning within the mentioned framework and analyze the underlying mechanisms at the level of behavior or neural circuit. Reward accelerates learning speed and enhances the extent of learning during the acquisition and consolidation stages, possibly by regulating the balance between the direct and indirect pathways (activating more D1-MSN than D2-MSN) of the ventral striatum and by increasing motor dynamics and kinematics. However, the effect varies depending on several experimental conditions. During the retention stage, there is a consensus that reward enhances both short-term and long-term memory retention in both types of motor learning, attributed to the LTP learning mechanism mediated by the VTA-M1 dopaminergic projection. Reward is a promising enhancer to bolster waning confidence and motivation, thereby increasing the efficiency of motor learning and rehabilitation. Further exploration of the circuit and functional connections between reward and the motor loop may provide a novel target for neural modulation to promote motor behavior.
Collapse
Affiliation(s)
- Jingwang Zhao
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Rehabilitation Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guanghu Zhang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Rehabilitation Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dongsheng Xu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Rehabilitation Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai, China
- Department of Rehabilitation Medicine, Shuguang Hospital, Shanghai, China
| |
Collapse
|
29
|
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
|
30
|
Liu X, Lu T, Chen X, Huang S, Zheng W, Zhang W, Meng S, Yan W, Shi L, Bao Y, Xue Y, Shi J, Yuan K, Han Y, Lu L. Memory consolidation drives the enhancement of remote cocaine memory via prefrontal circuit. Mol Psychiatry 2024; 29:730-741. [PMID: 38221548 DOI: 10.1038/s41380-023-02364-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 11/22/2023] [Accepted: 12/01/2023] [Indexed: 01/16/2024]
Abstract
Remote memory usually decreases over time, whereas remote drug-cue associated memory exhibits enhancement, increasing the risk of relapse during abstinence. Memory system consolidation is a prerequisite for remote memory formation, but neurobiological underpinnings of the role of consolidation in the enhancement of remote drug memory are unclear. Here, we found that remote cocaine-cue associated memory was enhanced in rats that underwent self-administration training, together with a progressive increase in the response of prelimbic cortex (PrL) CaMKII neurons to cues. System consolidation was required for the enhancement of remote cocaine memory through PrL CaMKII neurons during the early period post-training. Furthermore, dendritic spine maturation in the PrL relied on the basolateral amygdala (BLA) input during the early period of consolidation, contributing to remote memory enhancement. These findings indicate that memory consolidation drives the enhancement of remote cocaine memory through a time-dependent increase in activity and maturation of PrL CaMKII neurons receiving a sustained BLA input.
Collapse
Affiliation(s)
- Xiaoxing Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China
| | - Tangsheng Lu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Xuan Chen
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China
- Xinxiang Medical University, Xinxiang, 453003, China
| | - Shihao Huang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Wei Zheng
- Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, 100871, Beijing, China
| | - Wen Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China
| | - Shiqiu Meng
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China
| | - Wei Yan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China
| | - Le Shi
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China
| | - Yanping Bao
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China
| | - Yanxue Xue
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China.
| | - Jie Shi
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China
| | - Kai Yuan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China
| | - Ying Han
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China.
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China.
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China.
- Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, 100871, Beijing, China.
- Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences (No. 2018RU006), Dongcheng, Beijing, China.
| |
Collapse
|
31
|
Wingert JC, Ramos JD, Reynolds SX, Gonzalez AE, Rose RM, Hegarty DM, Aicher SA, Bailey LG, Brown TE, Abbas AI, Sorg BA. Perineuronal nets in the rat medial prefrontal cortex alter hippocampal-prefrontal oscillations and reshape cocaine self-administration memories. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.05.577568. [PMID: 38370716 PMCID: PMC10871211 DOI: 10.1101/2024.02.05.577568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
The medial prefrontal cortex (mPFC) is a major contributor to relapse to cocaine in humans and to reinstatement behavior in rodent models of cocaine use disorder. Output from the mPFC is modulated by parvalbumin (PV)-containing fast-spiking interneurons, the majority of which are surrounded by perineuronal nets (PNNs). Here we tested whether chondroitinase ABC (ABC)- mediated removal of PNNs prevented the acquisition or reconsolidation of a cocaine self-administration memory. ABC injections into the dorsal mPFC prior to training attenuated the acquisition of cocaine self-administration. Also, ABC given 3 days prior to but not 1 hr after memory reactivation blocked cue-induced reinstatement. However, reduced reinstatement was present only in rats given a novel reactivation contingency, suggesting that PNNs are required for the updating of a familiar memory. In naive rats, ABC injections into mPFC did not alter excitatory or inhibitory puncta on PV cells but reduced PV intensity. Whole-cell recordings revealed a greater inter-spike interval 1 hr after ABC, but not 3 days later. In vivo recordings from the mPFC and dorsal hippocampus (dHIP) during novel memory reactivation revealed that ABC in the mPFC prevented reward-associated increases in beta and gamma activity as well as phase-amplitude coupling between the dHIP and mPFC. Together, our findings show that PNN removal attenuates the acquisition of cocaine self-administration memories and disrupts reconsolidation of the original memory when combined with a novel reactivation session. Further, reduced dHIP/mPFC coupling after PNN removal may serve as a key biomarker for how to disrupt reconsolidation of cocaine memories and reduce relapse.
Collapse
|
32
|
Yang H, Zhang X, Zhang M, Lu Y, Xie B, Sun S, Yu H, Cong B, Luo Y, Ma C, Wen D. Roles of lncLingo2 and its derived miR-876-5p in the acquisition of opioid reinforcement. Addict Biol 2024; 29:e13375. [PMID: 38380802 PMCID: PMC10898844 DOI: 10.1111/adb.13375] [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: 11/14/2023] [Revised: 01/13/2024] [Accepted: 01/15/2024] [Indexed: 02/22/2024]
Abstract
Recent studies found that non-coding RNAs (ncRNAs) played crucial roles in drug addiction through epigenetic regulation of gene expression and underlying drug-induced neuroadaptations. In this study, we characterized lncRNA transcriptome profiles in the nucleus accumbens (NAc) of mice exhibiting morphine-conditioned place preference (CPP) and explored the prospective roles of novel differentially expressed lncRNA, lncLingo2 and its derived miR-876-5p in the acquisition of opioids-associated behaviours. We found that the lncLingo2 was downregulated within the NAc core (NAcC) but not in the NAc shell (NAcS). This downregulation was found to be associated with the development of morphine CPP and heroin intravenous self-administration (IVSA). As Mfold software revealed that the secondary structures of lncLingo2 contained the sequence of pre-miR-876, transfection of LV-lncLingo2 into HEK293 cells significantly upregulated miR-876 expression and the changes of mature miR-876 are positively correlated with lncLingo2 expression in NAcC of morphine CPP trained mice. Delivering miR-876-5p mimics into NAcC also inhibited the acquisition of morphine CPP. Furthermore, bioinformatics analysis and dual-luciferase assay confirmed that miR-876-5p binds to its target gene, Kcnn3, selectively and regulates morphine CPP training-induced alteration of Kcnn3 expression. Lastly, the electrophysiological analysis indicated that the currents of small conductance calcium-activated potassium (SK) channel was increased, which led to low neuronal excitability in NAcC after CPP training, and these changes were reversed by lncLingo2 overexpression. Collectively, lncLingo2 may function as a precursor of miR-876-5p in NAcC, hence modulating the development of opioid-associated behaviours in mice, which may serve as an underlying biomarker and therapeutic target of opioid addiction.
Collapse
Affiliation(s)
- Hongyu Yang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
| | - Xiuning Zhang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
| | - Minglong Zhang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
- Department of GeneticsQiqihar Medical UniversityQiqiharHeilongjiang ProvinceChina
| | - Yun Lu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
| | - Bing Xie
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
| | - Shaoguang Sun
- Department of Biochemistry and Molecular Biology, Key Laboratory of Medical Biotechnology of Hebei ProvinceHebei Medical UniversityShijiazhuangChina
- Key Laboratory of Neural and Vascular BiologyMinistry of EducationShijiazhuangHebei ProvinceChina
| | - Hailei Yu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
| | - Bin Cong
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
| | - Yixiao Luo
- Hunan Province People's HospitalThe First‐Affiliated Hospital of Hunan Normal UniversityChangshaChina
| | - Chunling Ma
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
- Key Laboratory of Neural and Vascular BiologyMinistry of EducationShijiazhuangHebei ProvinceChina
| | - Di Wen
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
- Key Laboratory of Neural and Vascular BiologyMinistry of EducationShijiazhuangHebei ProvinceChina
| |
Collapse
|
33
|
Suzuki T, Hattori S, Mizukami H, Nakajima R, Hibi Y, Kato S, Matsuzaki M, Ikebe R, Miyakawa T, Yamakawa K. Inversed Effects of Nav1.2 Deficiency at Medial Prefrontal Cortex and Ventral Tegmental Area for Prepulse Inhibition in Acoustic Startle Response. Mol Neurobiol 2024; 61:622-634. [PMID: 37650965 DOI: 10.1007/s12035-023-03610-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 08/20/2023] [Indexed: 09/01/2023]
Abstract
Numerous pathogenic variants of SCN2A gene, encoding voltage-gated sodium channel α2 subunit Nav1.2 protein, have been identified in a wide spectrum of neuropsychiatric disorders including schizophrenia. However, pathological mechanisms for the schizophrenia-relevant behavioral abnormalities caused by the variants remain poorly understood. Here in this study, we characterized mouse lines with selective Scn2a deletion at schizophrenia-related brain regions, medial prefrontal cortex (mPFC) or ventral tegmental area (VTA), obtained by injecting adeno-associated viruses (AAV) expressing Cre recombinase into homozygous Scn2a-floxed (Scn2afl/fl) mice, in which expression of the Scn2a was locally deleted in the presence of Cre recombinase. The mice lacking Scn2a in the mPFC exhibited a tendency for a reduction in prepulse inhibition (PPI) in acoustic startle response. Conversely, the mice lacking Scn2a in the VTA showed a significant increase in PPI. We also found that the mice lacking Scn2a in the mPFC displayed increased sociability, decreased locomotor activity, and increased anxiety-like behavior, while the mice lacking Scn2a in the VTA did not show any other abnormalities in these parameters except for vertical activity which is one of locomotor activities. These results suggest that Scn2a-deficiencies in mPFC and VTA are inversely relevant for the schizophrenic phenotypes in patients with SCN2A variants.
Collapse
Affiliation(s)
- Toshimitsu Suzuki
- Department of Neurodevelopmental Disorder Genetics, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan.
| | - Satoko Hattori
- Division of Systems Medical Science, Center for Medical Science, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
- Research Creation Support Center, Aichi Medical University, Nagakute, Aichi, 480-1195, Japan
| | - Hiroaki Mizukami
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, 329-0498, Japan
| | - Ryuichi Nakajima
- Division of Systems Medical Science, Center for Medical Science, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Yurina Hibi
- Department of Neurodevelopmental Disorder Genetics, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan
| | - Saho Kato
- Department of Neurodevelopmental Disorder Genetics, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan
| | - Mahoro Matsuzaki
- Department of Neurodevelopmental Disorder Genetics, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan
| | - Ryu Ikebe
- Department of Neurodevelopmental Disorder Genetics, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Center for Medical Science, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Kazuhiro Yamakawa
- Department of Neurodevelopmental Disorder Genetics, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan
| |
Collapse
|
34
|
Johnson CS, Chapp AD, Lind EB, Thomas MJ, Mermelstein PG. Sex differences in mouse infralimbic cortex projections to the nucleus accumbens shell. Biol Sex Differ 2023; 14:87. [PMID: 38082417 PMCID: PMC10712109 DOI: 10.1186/s13293-023-00570-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND The nucleus accumbens (NAc) is an important region in motivation and reward. Glutamatergic inputs from the infralimbic cortex (ILC) to the shell region of the NAc (NAcSh) have been implicated in driving the motivation to seek reward through repeated action-based behavior. While this has primarily been studied in males, observed sex differences in motivational circuitry and behavior suggest that females may be more sensitive to rewarding stimuli. These differences have been implicated for the observed vulnerability in women to substance use disorders. METHODS We used an optogenetic self-stimulation task in addition to ex vivo electrophysiological recordings of NAcSh neurons in mouse brain slices to investigate potential sex differences in ILC-NAcSh circuitry in reward-seeking behavior. Glutamatergic neurons in the ILC were infected with an AAV delivering DNA encoding for channelrhodopsin. Entering the designated active corner of an open field arena resulted in photostimulation of the ILC terminals in the NAcSh. Self-stimulation occurred during two consecutive days of testing over three consecutive weeks: first for 10 Hz, then 20 Hz, then 30 Hz. Whole-cell recordings of medium spiny neurons in the NAcSh assessed both optogenetically evoked local field potentials and intrinsic excitability. RESULTS Although both sexes learned to seek the active zone, within the first day, females entered the zone more than males, resulting in a greater amount of photostimulation. Increasing the frequency of optogenetic stimulation amplified female reward-seeking behavior. Males were less sensitive to ILC stimulation, with higher frequencies and repeated days required to increase male reward-seeking behavior. Unexpectedly, ex vivo optogenetic local field potentials in the NAcSh were greater in slices from male animals. In contrast, female medium-spiny neurons (MSNs) displayed significantly greater intrinsic neuronal excitability. CONCLUSIONS Taken together, these data indicate that there are sex differences in the motivated behavior driven by glutamate within the ILC-NAcSh circuit. Though glutamatergic signaling was greater in males, heightened intrinsic excitability in females appears to drive this sex difference.
Collapse
Affiliation(s)
- Caroline S Johnson
- Department of Neuroscience, School of Medicine, University of Minnesota, 4-140 Jackson Hall, 321 Church St SE, Minneapolis, MN, 55455, USA
| | - Andrew D Chapp
- Department of Neuroscience, School of Medicine, University of Minnesota, 4-140 Jackson Hall, 321 Church St SE, Minneapolis, MN, 55455, USA
- Medical Discovery Team on Addiction, University of Minnesota, 3-432 McGuire Translational Research Facility, 2001 6th St SE, Minneapolis, MN, 55455, USA
| | - Erin B Lind
- Department of Neuroscience, School of Medicine, University of Minnesota, 4-140 Jackson Hall, 321 Church St SE, Minneapolis, MN, 55455, USA
- Medical Discovery Team on Addiction, University of Minnesota, 3-432 McGuire Translational Research Facility, 2001 6th St SE, Minneapolis, MN, 55455, USA
| | - Mark J Thomas
- Department of Neuroscience, School of Medicine, University of Minnesota, 4-140 Jackson Hall, 321 Church St SE, Minneapolis, MN, 55455, USA
- Medical Discovery Team on Addiction, University of Minnesota, 3-432 McGuire Translational Research Facility, 2001 6th St SE, Minneapolis, MN, 55455, USA
| | - Paul G Mermelstein
- Department of Neuroscience, School of Medicine, University of Minnesota, 4-140 Jackson Hall, 321 Church St SE, Minneapolis, MN, 55455, USA.
- Medical Discovery Team on Addiction, University of Minnesota, 3-432 McGuire Translational Research Facility, 2001 6th St SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
35
|
Huang D, Ma YY. Increased Excitability of Layer 2 Cortical Pyramidal Neurons in the Supplementary Motor Cortex Underlies High Cocaine-Seeking Behaviors. Biol Psychiatry 2023; 94:875-887. [PMID: 37330163 PMCID: PMC10721734 DOI: 10.1016/j.biopsych.2023.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 05/29/2023] [Accepted: 06/07/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND Most efforts in addiction research have focused on the involvement of the medial prefrontal cortex, including the infralimbic, prelimbic, and anterior cingulate cortical areas, in cocaine-seeking behaviors. However, no effective prevention or treatment for drug relapse is available. METHODS We focused instead on the motor cortex, including both the primary and supplementary motor areas (M1 and M2, respectively). Addiction risk was evaluated by testing cocaine seeking after intravenous self-administration (IVSA) of cocaine in Sprague Dawley rats. The causal relationship between the excitability of cortical pyramidal neurons (CPNs) in M1/M2 and addiction risk was explored by ex vivo whole-cell patch clamp recordings and in vivo pharmacological or chemogenetic manipulation. RESULTS Our recordings showed that on withdrawal day 45 (WD45) after IVSA, cocaine, but not saline, increased the excitability of CPNs in the cortical superficial layers (primarily layer 2, denoted L2) but not in layer 5 (L5) in M2. Bilateral microinjection of the GABAA (gamma-aminobutyric acid A) receptor agonist muscimol to the M2 area attenuated cocaine seeking on WD45. More specifically, chemogenetic inhibition of CPN excitability in L2 of M2 (denoted M2-L2) by the DREADD (designer receptor exclusively activated by designer drugs) agonist compound 21 prevented drug seeking on WD45 after cocaine IVSA. This chemogenetic inhibition of M2-L2 CPNs had no effects on sucrose seeking. In addition, neither pharmacological nor chemogenetic inhibition manipulations altered general locomotor activity. CONCLUSIONS Our results indicate that cocaine IVSA induces hyperexcitability in the motor cortex on WD45. Importantly, the increased excitability in M2, particularly in L2, could be a novel target for preventing drug relapse during withdrawal.
Collapse
Affiliation(s)
- Donald Huang
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana
| | - Yao-Ying Ma
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
36
|
Wingert JC, Anguiano JN, Ramos JD, Blacktop JM, Gonzalez AE, Churchill L, Sorg BA. Enhanced expression of parvalbumin and perineuronal nets in the medial prefrontal cortex after extended-access cocaine self-administration in rats. Addict Biol 2023; 28:e13334. [PMID: 37855072 DOI: 10.1111/adb.13334] [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: 05/09/2023] [Revised: 07/27/2023] [Accepted: 08/28/2023] [Indexed: 10/20/2023]
Abstract
The medial prefrontal cortex (mPFC) drives cocaine-seeking behaviour in rodent models of cocaine use disorder. Parvalbumin (PV)-containing GABAergic interneurons powerfully control the output of the mPFC, yet few studies have focused on how these neurons modulate cocaine-seeking behaviour. Most PV neurons are surrounded by perineuronal nets (PNNs), which regulate the firing of PV neurons. We examined staining intensity and number of PV and PNNs after long-access (6 h/day) cocaine self-administration in rats followed by either 8-10 days extinction ± cue-induced reinstatement or short-term (1-2 days) or long-term (30-31 days) abstinence ± cue-induced reinstatement. The intensity of PNNs was increased in the prelimbic and infralimbic PFC after long-term abstinence in the absence of cue reinstatement and after cue reinstatement following both daily extinction sessions and after a 30-day abstinence period. PV intensity was increased after 30 days of abstinence in the prelimbic but not infralimbic PFC. Enzymatic removal of PNNs with chondroitinase ABC (ABC) in the prelimbic PFC did not prevent incubation of cue-induced reinstatement but decreased cocaine-seeking behaviour at both 2 and 31 days of abstinence, and this decrease at 31 days was accompanied by reduced c-Fos levels in the prelimbic PFC. Increases in PNN intensity have generally been associated with the loss of plasticity, suggesting that the persistent and chronic nature of cocaine use disorder may in part be attributed to long-lasting increases in PNN intensity that reduce the ability of stimuli to alter synaptic input to underlying PV neurons.
Collapse
Affiliation(s)
- Jereme C Wingert
- Neuroscience, Washington State University, Vancouver, Washington, USA
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, Oregon, USA
| | - Jonathan N Anguiano
- Neuroscience, Washington State University, Vancouver, Washington, USA
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, Oregon, USA
| | - Jonathan D Ramos
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, Oregon, USA
| | - Jordan M Blacktop
- Neuroscience, Washington State University, Vancouver, Washington, USA
| | - Angela E Gonzalez
- Neuroscience, Washington State University, Vancouver, Washington, USA
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, Oregon, USA
| | - Lynn Churchill
- Neuroscience, Washington State University, Pullman, Washington, USA
| | - Barbara A Sorg
- Neuroscience, Washington State University, Vancouver, Washington, USA
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, Oregon, USA
| |
Collapse
|
37
|
Namba MD, Xie Q, Barker JM. Advancing the preclinical study of comorbid neuroHIV and substance use disorders: Current perspectives and future directions. Brain Behav Immun 2023; 113:453-475. [PMID: 37567486 PMCID: PMC10528352 DOI: 10.1016/j.bbi.2023.07.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/23/2023] [Accepted: 07/30/2023] [Indexed: 08/13/2023] Open
Abstract
Human immunodeficiency virus (HIV) remains a persistent public health concern throughout the world. Substance use disorders (SUDs) are a common comorbidity that can worsen treatment outcomes for people living with HIV. The relationship between HIV infection and SUD outcomes is likely bidirectional, making clear interrogation of neurobehavioral outcomes challenging in clinical populations. Importantly, the mechanisms through which HIV and addictive drugs disrupt homeostatic immune and CNS function appear to be highly overlapping and synergistic within HIV-susceptible reward and motivation circuitry in the central nervous system. Decades of animal research have revealed invaluable insights into mechanisms underlying the pathophysiology SUDs and HIV, although translational studies examining comorbid SUDs and HIV are very limited due to the technical challenges of modeling HIV infection preclinically. In this review, we discuss preclinical animal models of HIV and highlight key pathophysiological characteristics of each model, with a particular emphasis on rodent models of HIV. We then review the implementation of these models in preclinical SUD research and identify key gaps in knowledge in the field. Finally, we discuss how cutting-edge behavioral neuroscience tools, which have revealed key insights into the neurobehavioral mechanisms of SUDs, can be applied to preclinical animal models of HIV to reveal potential, novel treatment avenues for comorbid HIV and SUDs. Here, we argue that future preclinical SUD research would benefit from incorporating comorbidities such as HIV into animal models and would facilitate the discovery of more refined, subpopulation-specific mechanisms and effective SUD prevention and treatment targets.
Collapse
Affiliation(s)
- Mark D Namba
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Qiaowei Xie
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Jacqueline M Barker
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA.
| |
Collapse
|
38
|
Bijoch Ł, Klos J, Pękała M, Fiołna K, Kaczmarek L, Beroun A. Diverse processing of pharmacological and natural rewards by the central amygdala. Cell Rep 2023; 42:113036. [PMID: 37616162 DOI: 10.1016/j.celrep.2023.113036] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/03/2023] [Accepted: 08/11/2023] [Indexed: 08/25/2023] Open
Abstract
The central amygdala (CeA) with its medial (CeM) and lateral (CeL) nuclei is the brain hub for processing stimuli with emotional context. CeL nucleus gives a strong inhibitory input to the CeM, and this local circuitry assigns values (positive or negative) to incoming stimuli, guiding appropriate behavior (approach or avoid). However, the particular involvement of CeA in processing such emotionally relevant information and adaptations of the CeA circuitry are not yet well understood. In this study, we examined synaptic plasticity in the CeA after exposure to two types of rewards, pharmacological (cocaine) and natural (sugar). We found that both rewards engage CeM, where they generate silent synapses resulting in the strengthening of the network. However, only cocaine triggers plasticity in the CeL, which leads to the weakening of its excitatory inputs. Finally, chemogenetic inhibition of CeM attenuates animal preference for sugar, while activation delays cocaine-induced increase in locomotor activity.
Collapse
Affiliation(s)
- Łukasz Bijoch
- Nencki-EMBL Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Laboratory of Neuronal Plasticity, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, L. Pasteura 3, 02-093 Warsaw, Poland
| | - Joanna Klos
- Nencki-EMBL Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Laboratory of Neuronal Plasticity, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, L. Pasteura 3, 02-093 Warsaw, Poland
| | - Martyna Pękała
- Nencki-EMBL Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Laboratory of Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, L. Pasteura 3, 02-093 Warsaw, Poland
| | - Kristina Fiołna
- Nencki-EMBL Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Laboratory of Neuronal Plasticity, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, L. Pasteura 3, 02-093 Warsaw, Poland; Nencki-EMBL Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Laboratory of Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, L. Pasteura 3, 02-093 Warsaw, Poland
| | - Leszek Kaczmarek
- Nencki-EMBL Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Laboratory of Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, L. Pasteura 3, 02-093 Warsaw, Poland
| | - Anna Beroun
- Nencki-EMBL Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Laboratory of Neuronal Plasticity, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, L. Pasteura 3, 02-093 Warsaw, Poland.
| |
Collapse
|
39
|
Ngbokoli ML, Douton JE, Carelli RM. Prelimbic cortex and nucleus accumbens core resting state signaling dynamics as a biomarker for cocaine seeking behaviors. ADDICTION NEUROSCIENCE 2023; 7:100097. [PMID: 37396409 PMCID: PMC10310298 DOI: 10.1016/j.addicn.2023.100097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Substance use disorders (SUDs) are characterized by maladaptive signaling in the prefrontal cortex and associated regions, however precisely how these drug-induced abnormalities may be linked to drug seeking/taking behaviors is not well understood. Here, in vivo local field potential (LFP) electrophysiology was used in rats to examine the relationship between overall spontaneous (resting state) activity within the prelimbic cortex (PrL) and nucleus accumbens (NAc) core, and their functional connectivity, to cocaine taking and seeking behaviors. Adult, male Sprague-Dawley rats were trained to self-administer either intravenous cocaine (0.33 mg/inf) or water reinforcement during 6-hour daily sessions over 2 weeks; extinction sessions were completed immediately after self-administration training and following 30 days experimenter-imposed abstinence. Rest LFP recordings were completed during 3 recording periods (15 min each in a chamber different from the self-administration context) conducted (1) prior to self-administration training (rest LFP 1) (2) immediately after 2 weeks of self-administration training (rest LFP 2) and (3) following 1 month abstinence (rest LFP 3). Our findings show that resting state LFP power in the PrL recorded prior to training (Rest LFP 1) was positively correlated with total cocaine intake and escalation of cocaine seeking at the beta frequency range. Immediately after self-administration training (Rest LFP 2) power in the NAc core at gamma frequency was negatively correlated with incubation of cocaine craving. For rats trained to self-administer water, no significant correlations were observed. Together, these findings show that resting state LFP at specific timepoints in the addiction cycle can serve as unique predictors (biomarkers) of cocaine use disorders.
Collapse
Affiliation(s)
- Metika L. Ngbokoli
- Department of Psychology and Neuroscience, The University of North Carolina, CB #3270 Davie Hall, Chapel Hill, NC 27599, USA
| | - Joaquin E. Douton
- Department of Psychology and Neuroscience, The University of North Carolina, CB #3270 Davie Hall, Chapel Hill, NC 27599, USA
| | - Regina M. Carelli
- Department of Psychology and Neuroscience, The University of North Carolina, CB #3270 Davie Hall, Chapel Hill, NC 27599, USA
| |
Collapse
|
40
|
He Y, Wang J, Li KL, Wang YQ, Freyberg Z, Dong Y. Membrane excitability of nucleus accumbens neurons gates the incubation of cocaine craving. Neuropsychopharmacology 2023; 48:1318-1327. [PMID: 37041207 PMCID: PMC10354025 DOI: 10.1038/s41386-023-01580-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/15/2023] [Accepted: 03/28/2023] [Indexed: 04/13/2023]
Abstract
After drug withdrawal, a key factor triggering relapse is progressively intensified cue-associated drug craving, termed incubation of drug craving. After withdrawal from cocaine self-administration, incubation of cocaine craving develops more reliably in rats compared to mice. This species difference provides an opportunity to determine rat-specific cellular adaptations, which may constitute the critical mechanisms that contribute to incubated cocaine craving in humans. Expression of incubated cocaine seeking is mediated, in part, by cocaine-induced cellular adaptations in medium spiny neurons (MSNs) within the nucleus accumbens (NAc). In rats, decreased membrane excitability in NAc MSNs is a prominent cellular adaptation, which is induced after cocaine self-administration and lasts throughout prolonged drug withdrawal. Here, we show that, similar to rats, mice exhibit decreased membrane excitability of dopamine D1 receptor (D1)-, but not D2 (D2)-, expressing MSNs within the NAc shell (NAcSh) after 1 d withdrawal from cocaine self-administration. However, in contrast to rats, this membrane adaptation does not persist in mice, diminishing after 45-d withdrawal. We also find that restoring the membrane excitability of NAcSh MSNs after cocaine withdrawal decreases cocaine seeking in rats. This suggests that drug-induced membrane adaptations are essential for behavioral expression of incubated cocaine craving. In mice, however, experimentally inducing hypoactivity of D1 NAcSh MSNs after cocaine withdrawal does not alter cocaine seeking, suggesting that MSN hypo-excitability alone is insufficient to increase cocaine seeking. Together, our results demonstrate an overall permissive role of cocaine-induced hypoactivity of NAcSh MSNs in gating increased cocaine seeking after prolonged cocaine withdrawal.
Collapse
Affiliation(s)
- Yi He
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Junshi Wang
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - King-Lun Li
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Yao Q Wang
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15260, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA.
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
41
|
Kalivas PW, Gourley SL, Paulus MP. Intrusive thinking: Circuit and synaptic mechanisms of a transdiagnostic psychiatric symptom. Neurosci Biobehav Rev 2023; 150:105196. [PMID: 37094741 PMCID: PMC10249786 DOI: 10.1016/j.neubiorev.2023.105196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 04/17/2023] [Accepted: 04/21/2023] [Indexed: 04/26/2023]
Abstract
Spontaneous thought is an adaptive cognitive process that can produce novel and insightful thought sequences useful in guiding future behavior. In many psychiatric disorders, spontaneous thinking becomes intrusive and uncontrolled, and can trigger symptoms such as craving, repetitive negative thinking and trauma-related memories. We link studies using clinical imaging and rodent modeling towards understanding the neurocircuitry and neuroplasticity of intrusive thinking. We propose a framework in which drugs or stress change the homeostatic set point of brain reward circuitry, which then impacts subsequent plasticity induced by drug/stress conditioned cues (metaplastic allostasis). We further argue for the importance of examining not only the canonical pre- and postsynapse, but also the adjacent astroglial protrusions and extracellular matrix that together form the tetrapartite synapse and that plasticity throughout the tetrapartite synapse is necessary for cue-induced drug or stress behaviors. This analysis reveals that drug use or trauma cause long-lasting allostatic brain plasticity that sets the stage for subsequent drug/trauma-associated cues to induce transient plasticity that can lead to intrusive thinking.
Collapse
Affiliation(s)
- Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
| | - Shannon L Gourley
- Emory National Primate Research Center, Emory University, Department of Pediatrics and Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA; Emory National Primate Research Center, Emory University, Emory University School of Medicine, Atlanta, GA, USA
| | | |
Collapse
|
42
|
Mango D, Ledonne A. Updates on the Physiopathology of Group I Metabotropic Glutamate Receptors (mGluRI)-Dependent Long-Term Depression. Cells 2023; 12:1588. [PMID: 37371058 DOI: 10.3390/cells12121588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Group I metabotropic glutamate receptors (mGluRI), including mGluR1 and mGluR5 subtypes, modulate essential brain functions by affecting neuronal excitability, intracellular calcium dynamics, protein synthesis, dendritic spine formation, and synaptic transmission and plasticity. Nowadays, it is well appreciated that the mGluRI-dependent long-term depression (LTD) of glutamatergic synaptic transmission (mGluRI-LTD) is a key mechanism by which mGluRI shapes connectivity in various cerebral circuitries, directing complex brain functions and behaviors, and that it is deranged in several neurological and psychiatric illnesses, including neurodevelopmental disorders, neurodegenerative diseases, and psychopathologies. Here, we will provide an updated overview of the physiopathology of mGluRI-LTD, by describing mechanisms of induction and regulation by endogenous mGluRI interactors, as well as functional physiological implications and pathological deviations.
Collapse
Affiliation(s)
- Dalila Mango
- School of Pharmacy, University of Rome "Tor Vergata", 00133 Rome, Italy
- Laboratory of Pharmacology of Synaptic Plasticity, European Brain Research Institute, 00161 Rome, Italy
| | - Ada Ledonne
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
- Department of Experimental Neuroscience, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
| |
Collapse
|
43
|
Kumaresan V, Lim Y, Juneja P, Tipton AE, de Guglielmo G, Carrette LLG, Kallupi M, Maturin L, Liu Y, George O, Zhang H. Abstinence from Escalation of Cocaine Intake Changes the microRNA Landscape in the Cortico-Accumbal Pathway. Biomedicines 2023; 11:1368. [PMID: 37239038 PMCID: PMC10216163 DOI: 10.3390/biomedicines11051368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 05/28/2023] Open
Abstract
Cocaine administration alters the microRNA (miRNA) landscape in the cortico-accumbal pathway. These changes in miRNA can play a major role in the posttranscriptional regulation of gene expression during withdrawal. This study aimed to investigate the changes in microRNA expression in the cortico-accumbal pathway during acute withdrawal and protracted abstinence following escalated cocaine intake. Small RNA sequencing (sRNA-seq) was used to profile miRNA transcriptomic changes in the cortico-accumbal pathway [infralimbic- and prelimbic-prefrontal cortex (IL and PL) and nucleus accumbens (NAc)] of rats with extended access to cocaine self-administration followed by an 18-h withdrawal or a 4-week abstinence. An 18-h withdrawal led to differential expression (fold-change > 1.5 and p < 0.05) of 21 miRNAs in the IL, 18 miRNAs in the PL, and two miRNAs in the NAc. The mRNAs potentially targeted by these miRNAs were enriched in the following pathways: gap junctions, neurotrophin signaling, MAPK signaling, and cocaine addiction. Moreover, a 4-week abstinence led to differential expression (fold-change > 1.5 and p < 0.05) of 23 miRNAs in the IL, seven in the PL, and five miRNAs in the NAc. The mRNAs potentially targeted by these miRNAs were enriched in pathways including gap junctions, cocaine addiction, MAPK signaling, glutamatergic synapse, morphine addiction, and amphetamine addiction. Additionally, the expression levels of several miRNAs differentially expressed in either the IL or the NAc were significantly correlated with addiction behaviors. Our findings highlight the impact of acute and protracted abstinence from escalated cocaine intake on miRNA expression in the cortico-accumbal pathway, a key circuit in addiction, and suggest developing novel biomarkers and therapeutic approaches to prevent relapse by targeting abstinence-associated miRNAs and their regulated mRNAs.
Collapse
Affiliation(s)
- Vidhya Kumaresan
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA;
| | - Yolpanhchana Lim
- Department of Psychiatry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA; (Y.L.); (P.J.); (Y.L.)
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| | - Poorva Juneja
- Department of Psychiatry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA; (Y.L.); (P.J.); (Y.L.)
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| | - Allison E. Tipton
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA;
| | - Giordano de Guglielmo
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; (G.d.G.); (L.L.G.C.); (M.K.); (L.M.); (O.G.)
| | - Lieselot L. G. Carrette
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; (G.d.G.); (L.L.G.C.); (M.K.); (L.M.); (O.G.)
| | - Marsida Kallupi
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; (G.d.G.); (L.L.G.C.); (M.K.); (L.M.); (O.G.)
| | - Lisa Maturin
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; (G.d.G.); (L.L.G.C.); (M.K.); (L.M.); (O.G.)
| | - Ying Liu
- Department of Psychiatry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA; (Y.L.); (P.J.); (Y.L.)
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| | - Olivier George
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; (G.d.G.); (L.L.G.C.); (M.K.); (L.M.); (O.G.)
| | - Huiping Zhang
- Department of Psychiatry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA; (Y.L.); (P.J.); (Y.L.)
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
44
|
Wang L, Gao M, Wang Q, Sun L, Younus M, Ma S, Liu C, Shi L, Lu Y, Zhou B, Sun S, Chen G, Li J, Zhang Q, Zhu F, Wang C, Zhou Z. Cocaine induces locomotor sensitization through a dopamine-dependent VTA-mPFC-FrA cortico-cortical pathway in male mice. Nat Commun 2023; 14:1568. [PMID: 36944634 PMCID: PMC10030897 DOI: 10.1038/s41467-023-37045-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 02/28/2023] [Indexed: 03/23/2023] Open
Abstract
As a central part of the mammalian brain, the prefrontal cortex (PFC) has been implicated in regulating cocaine-induced behaviors including compulsive seeking and reinstatement. Although dysfunction of the PFC has been reported in animal and human users with chronic cocaine abuse, less is known about how the PFC is involved in cocaine-induced behaviors. By using two-photon Ca2+ imaging to simultaneously record tens of intact individual networking neurons in the frontal association cortex (FrA) in awake male mice, here we report that a systematic acute cocaine exposure decreased the FrA neural activity in mice, while the chemogenetic intervention blocked the cocaine-induced locomotor sensitization. The hypoactivity of FrA neurons was critically dependent on both dopamine transporters and dopamine transmission in the ventromedial PFC (vmPFC). Both dopamine D1R and D2R neurons in the vmPFC projected to and innervated FrA neurons, the manipulation of which changed the cocaine-induced hypoactivity of the FrA and locomotor sensitization. Together, this work demonstrates acute cocaine-induced hypoactivity of FrA neurons in awake mice, which defines a cortico-cortical projection bridging dopamine transmission and cocaine sensitization.
Collapse
Affiliation(s)
- Lun Wang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Min Gao
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
- Joint Graduate Program of Peking-Tsinghua-NIBS, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Qinglong Wang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Liyuan Sun
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Muhammad Younus
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Sixing Ma
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Can Liu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Li Shi
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Yang Lu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Bo Zhou
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Suhua Sun
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Guoqing Chen
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Jie Li
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Quanfeng Zhang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Feipeng Zhu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Changhe Wang
- Neuroscience Research Center, Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
- Department of Neurology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Zhuan Zhou
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.
| |
Collapse
|
45
|
Nett KE, Zimbelman AR, McGregor MS, Alizo Vera V, Harris MR, LaLumiere RT. Infralimbic Projections to the Nucleus Accumbens Shell and Amygdala Regulate the Encoding of Cocaine Extinction Learning. J Neurosci 2023; 43:1348-1359. [PMID: 36657972 PMCID: PMC9987566 DOI: 10.1523/jneurosci.2023-22.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/21/2022] [Accepted: 12/30/2022] [Indexed: 01/20/2023] Open
Abstract
Prior evidence indicates that the infralimbic cortex (IL) mediates the ongoing inhibition of cocaine seeking following self-administration and extinction training in rats, specifically through projections to the nucleus accumbens shell (NAshell). Our own data indicate that IL activity immediately following an unreinforced lever press is critical for encoding the extinction contingencies in such procedures. Whether extinction encoding requires activity in the IL exclusively or also activity in its outputs, such as those to the NAshell and amygdala, is unknown. To address this issue, we used a closed-loop optogenetic approach in female and male Sprague Dawley rats to silence IL-NAshell or IL-amygdala activity following an unreinforced lever press during extinction training. Optical illumination (20 s) was given either immediately after a lever press or following a 20 s delay. IL-NAshell inhibition immediately following an unreinforced lever press increased lever pressing during extinction training and impaired retention of extinction learning, as assessed during subsequent extinction sessions without optical inhibition. Likewise, IL-amygdala inhibition given in the same manner impaired extinction retention during sessions without inhibition. Control experiments indicate that critical encoding of extinction learning does not require activity in these pathways beyond the initial 20 s post-lever press period, as delayed IL-NAshell and IL-amygdala inhibition had no effect on extinction learning. These results suggest that a larger network extending from the IL to the NAshell and amygdala is involved in encoding extinction contingencies following cocaine self-administration.SIGNIFICANCE STATEMENT Infralimbic cortex (IL) activity following an unreinforced lever press during extinction learning encodes the extinction of cocaine-seeking behavior. However, the larger circuitry controlling such encoding has not been investigated. Using closed-loop optogenetic pathway targeting, we found that inhibition of IL projections to the nucleus accumbens shell and to the amygdala impaired the extinction of cocaine seeking. Importantly, these effects were only observed when activity was disrupted during the first 20 s post-lever press and not when given following a 20 s delay. These findings suggest that successful cocaine extinction encoding requires activity across a larger circuit beyond simply inputs to the IL.
Collapse
Affiliation(s)
- Kelle E Nett
- Interdisciplinary Neuroscience Program, University of Iowa, Iowa City, Iowa 52242
| | - Alexa R Zimbelman
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, Iowa 52242
| | - Matthew S McGregor
- Interdisciplinary Neuroscience Program, University of Iowa, Iowa City, Iowa 52242
| | - Vanessa Alizo Vera
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, Iowa 52242
| | - Molly R Harris
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, Iowa 52242
| | - Ryan T LaLumiere
- Interdisciplinary Neuroscience Program, University of Iowa, Iowa City, Iowa 52242
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, Iowa 52242
- Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa 52242
| |
Collapse
|
46
|
Li JY, Yu YJ, Su CL, Shen YQ, Chang CH, Gean PW. Modulation of methamphetamine memory reconsolidation by neural projection from basolateral amygdala to nucleus accumbens. Neuropsychopharmacology 2023; 48:478-488. [PMID: 36109595 PMCID: PMC9852248 DOI: 10.1038/s41386-022-01417-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/21/2022] [Accepted: 07/28/2022] [Indexed: 02/02/2023]
Abstract
Drug-associated conditioned cues promote subjects to recall drug reward memory, resulting in drug-seeking and reinstatement. A consolidated memory becomes unstable after recall, such that the amnestic agent can disrupt the memory during the reconsolidation stage, which implicates a potential therapeutic strategy for weakening maladaptive memories. The basolateral amygdala (BLA) involves the association of conditioned cues with reward and aversive valences and projects the information to the nucleus accumbens (NAc) that mediates reward-seeking. However, whether the BLA-NAc projection plays a role in drug-associated memory reactivation and reconsolidation is unknown. We used methamphetamine (MeAM) conditioned place preference (CPP) to investigate the role of BLA-NAc neural projection in the memory reconsolidation. Two weeks before CPP training, we infused adeno-associated virus (AAV) carrying the designer receptor exclusively activated by designer drugs (DREADD) or control constructs. We infused clozapine-N-oxide (CNO) after the recall test to manipulate the neural activity of BLA-NAc projections in mice. We found that after recall, DREADD-mediated inhibition of BLA neurons projecting to the NAc core blunted consolidated MeAM-associated memory. Inhibition of BLA glutamatergic nerve terminals in the NAc core 1 h after recall disrupted consolidated MeAM-associated memory. However, inhibiting this pathway after the time window of reconsolidation failed to affect memory. Furthermore, under the condition without memory retrieval, DREADD-mediated activation of BLA-NAc core projection was required for amnesic agents to disrupt consolidated MeAM-associated memory. Our findings provide evidence that the BLA-NAc pathway activity is involved in the post-retrieval processing of MeAM-associated memory in CPP.
Collapse
Affiliation(s)
- Jia-Ying Li
- Department of Pharmacology, National Cheng-Kung University, Tainan, 701, Taiwan, ROC
| | - Yang-Jung Yu
- Department of Pharmacology, National Cheng-Kung University, Tainan, 701, Taiwan, ROC
| | - Chun-Lin Su
- Department of Pharmacology, National Cheng-Kung University, Tainan, 701, Taiwan, ROC
| | - Yu-Qi Shen
- Department of Pharmacology, National Cheng-Kung University, Tainan, 701, Taiwan, ROC
| | - Chih-Hua Chang
- Department of Pharmacology, National Cheng-Kung University, Tainan, 701, Taiwan, ROC.
- Department of Biotechnology and Bioindustry Sciences, National Cheng-Kung University, Tainan, 701, Taiwan, ROC.
| | - Po-Wu Gean
- Department of Pharmacology, National Cheng-Kung University, Tainan, 701, Taiwan, ROC.
- Department of Biotechnology and Bioindustry Sciences, National Cheng-Kung University, Tainan, 701, Taiwan, ROC.
| |
Collapse
|
47
|
Moschak TM, Sloand TJ, Carelli RM. Prelimbic Cortex Activity during a Distress Tolerance Task Predicts Cocaine-Seeking Behavior in Male, But Not Female Rats. J Neurosci 2023; 43:647-655. [PMID: 36639895 PMCID: PMC9888504 DOI: 10.1523/jneurosci.1718-22.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/04/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022] Open
Abstract
Distress tolerance (DT) is defined as the ability to persist in challenging goal-directed behavior in the face of stress, and individuals with low DT exhibit heightened drug-seeking behavior. However, no preclinical studies have examined the neurobiology underlying this phenomenon. To assess this, in vivo electrophysiology was used in Long Evans male and female rats during a DT task to record neural activity in the prelimbic cortex (PrL), a brain region implicated in drug-seeking. Rats were first assessed for DT, defined as the amount of time elapsed before rats quit seeking reward in an increasingly difficult operant task. Subsequently, rats underwent 2 weeks of self-administration for either water/saline or cocaine for 6 h/day. Animals then began a 1 month period of experimenter-imposed abstinence to induce heightened drug-seeking behavior. On day 28 of abstinence, DT and neural activity were reassessed; and on day 30, cocaine-seeking behavior was examined under extinction. Males had significantly higher DT than females and exhibited significantly more phasic PrL activity during the DT task. Furthermore, in male rats with a history of cocaine, PrL activity shifted to track DT; and this change in activity significantly correlated with the change in DT. Additionally, male (but not female) rats with low DT after 28 d of abstinence had significantly heightened drug-seeking behavior. Finally, PrL activity during the DT task predicted cocaine-seeking behavior. Collectively, these data demonstrate an important role for the PrL in DT in males, and link this neural activity and behavior to drug-seeking, particularly in males.SIGNIFICANCE STATEMENT Distress tolerance (DT) is defined as the ability to persist in challenging goal-directed behavior in the face of stress, and individuals with low DT exhibit heightened drug-seeking. Here, we investigated the role of the prelimbic cortex (PrL) in DT and its relationship to cocaine-seeking in male and female rats. We found that males had significantly higher DT than females and exhibited significantly more PrL activity during the DT task. Furthermore, male (but not female) rats with low DT after 28 d of abstinence had significantly heightened drug-seeking behavior. Finally, PrL activity during the DT task predicted cocaine-seeking. These data demonstrate an important role for the PrL in DT and link this neural activity and behavior to drug-seeking in males.
Collapse
Affiliation(s)
- Travis M Moschak
- Department of Biological Sciences, University of Texas at El Paso, El Paso, Texas 79902
| | - T Joseph Sloand
- Department of Psychology & Neuroscience, University of North Carolina Chapel Hill, Chapel Hill, North Carolina 27599
| | - Regina M Carelli
- Department of Psychology & Neuroscience, University of North Carolina Chapel Hill, Chapel Hill, North Carolina 27599
| |
Collapse
|
48
|
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: 11] [Impact Index Per Article: 5.5] [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
|
49
|
Preventing incubation of drug craving to treat drug relapse: from bench to bedside. Mol Psychiatry 2023; 28:1415-1429. [PMID: 36646901 DOI: 10.1038/s41380-023-01942-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 12/24/2022] [Accepted: 01/06/2023] [Indexed: 01/17/2023]
Abstract
In 1986, Gawin and Kleber reported a progressive increase in cue-induced drug craving in individuals with cocaine use disorders during prolonged abstinence. After years of controversy, as of 2001, this phenomenon was confirmed in rodent studies using self-administration model, and defined as the incubation of drug craving. The intensification of cue-induced drug craving after withdrawal exposes abstinent individuals to a high risk of relapse, which urged us to develop effective interventions to prevent incubated craving. Substantial achievements have been made in deciphering the neural mechanisms, with potential implications for reducing drug craving and preventing the relapse. The present review discusses promising drug targets that have been well investigated in animal studies, including some neurotransmitters, neuropeptides, neurotrophic factors, and epigenetic markers. We also discuss translational exploitation and challenges in the field of the incubation of drug craving, providing insights into future investigations and highlighting the potential of pharmacological interventions, environment-based interventions, and neuromodulation techniques.
Collapse
|
50
|
Huerta Sanchez LL, Sankaran M, Li TL, Doan H, Chiu A, Shulman E, Shab G, Kippin TE, Szumlinski KK. Profiling prefrontal cortex protein expression in rats exhibiting an incubation of cocaine craving following short-access self-administration procedures. Front Psychiatry 2023; 13:1031585. [PMID: 36684008 PMCID: PMC9846226 DOI: 10.3389/fpsyt.2022.1031585] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/25/2022] [Indexed: 01/05/2023] Open
Abstract
Introduction Incubation of drug-craving refers to a time-dependent increase in drug cue-elicited craving that occurs during protracted withdrawal. Historically, rat models of incubated cocaine craving employed extended-access (typically 6 h/day) intravenous drug self-administration (IV-SA) procedures, although incubated cocaine craving is reported to occur following shorter-access IV-SA paradigms. The notoriously low-throughput of extended-access IV-SA prompted us to determine whether two different short-access IV-SA procedures akin to those in the literature result in qualitatively similar changes in glutamate receptor expression and the activation of downstream signaling molecules within prefrontal cortex (PFC) subregions as those reported previously by our group under 6h-access conditions. Methods For this, adult, male Sprague-Dawley rats were trained to intravenously self-administer cocaine for 2 h/day for 10 consecutive days (2-h model) or for 6 h on day 1 and 2 h/day for the remaining 9 days of training (Mixed model). A sham control group was also included that did not self-administer cocaine. Results On withdrawal day 3 or 30, rats were subjected to a 2-h test of cue-reinforced responding in the absence of cocaine and a time-dependent increase in drug-seeking was observed under both IV-SA procedures. Immunoblotting of brain tissue collected immediately following the cue test session indicated elevated phospho-Akt1, phospho-CaMKII and Homer2a/b expression within the prelimbic subregion of the PFC of cocaine-incubated rats. However, we failed to detect incubation-related changes in Group 1 metabotropic glutamate receptor or ionotropic glutamate receptor subunit expression in either subregion. Discussion These results highlight further a role for Akt1-related signaling within the prelimbic cortex in driving incubated cocaine craving, and provide novel evidence supporting a potential role also for CaMKII-dependent signaling through glutamate receptors in this behavioral phenomenon.
Collapse
Affiliation(s)
- Laura L. Huerta Sanchez
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Mathangi Sankaran
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Taylor L. Li
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Hoa Doan
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Alvin Chiu
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Eleanora Shulman
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Gabriella Shab
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Tod E. Kippin
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, United States
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Karen K. Szumlinski
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, United States
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States
| |
Collapse
|