1
|
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
|
2
|
Kawa AB, Hashimoto JG, Beutler MM, Guizzetti M, Wolf ME. Changes in nucleus accumbens core translatome accompanying incubation of cocaine craving. Neuropsychopharmacology 2025:10.1038/s41386-025-02112-4. [PMID: 40301580 DOI: 10.1038/s41386-025-02112-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 03/25/2025] [Accepted: 04/09/2025] [Indexed: 05/01/2025]
Abstract
In the 'incubation of cocaine craving' model of relapse, rats exhibit progressive intensification (incubation) of cue-induced craving over several weeks of forced abstinence from cocaine self-administration. The expression of incubated craving depends on plasticity of excitatory synaptic transmission in nucleus accumbens core (NAcC) medium spiny neurons (MSN). Previously, we found that the maintenance of this plasticity and the expression of incubation depends on ongoing protein translation, and the regulation of translation is altered after incubation of cocaine craving. Here we used male and female rats that express Cre recombinase in either dopamine D1 receptor- or adenosine 2a (A2a) receptor-expressing MSN to express a GFP-tagged ribosomal subunit in a cell-type specific manner, enabling us to use Translating Ribosome Affinity Purification (TRAP) to isolate actively translating mRNAs from both MSN subtypes for analysis by RNA-seq. We compared rats that self-administered saline or cocaine. Saline rats were assessed on abstinence day (AD) 1, while cocaine rats were assessed on AD1 or AD40-50. For both D1-MSN and A2a-MSN, there were few differentially translated genes between saline and cocaine AD1 groups. In contrast, pronounced differences in the translatome were observed between cocaine rats on AD1 and AD40-50, and this was far more robust in D1-MSN. Notably, all comparisons revealed sex differences in translating mRNAs. Sequencing results were validated by qRT-PCR for several genes of interest. This study, the first to combine TRAP-seq, transgenic rats, and a cocaine self-administration paradigm, identifies translating mRNAs linked to incubation of cocaine craving in D1-MSN and A2a-MSN of the NAcC.
Collapse
Affiliation(s)
- Alex B Kawa
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Joel G Hashimoto
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
- VA Portland Health Care System, Portland, OR, 97239, USA
| | - Madelyn M Beutler
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Marina Guizzetti
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
- VA Portland Health Care System, Portland, OR, 97239, USA
| | - Marina E Wolf
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|
3
|
Shi Z, Wen K, Sammudin NH, LoRocco N, Zhuang X. Erasing "bad memories": reversing aberrant synaptic plasticity as therapy for neurological and psychiatric disorders. Mol Psychiatry 2025:10.1038/s41380-025-03013-0. [PMID: 40210977 DOI: 10.1038/s41380-025-03013-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 02/24/2025] [Accepted: 04/02/2025] [Indexed: 04/12/2025]
Abstract
Dopamine modulates corticostriatal plasticity in both the direct and indirect pathways of the cortico-striato-thalamo-cortical (CSTC) loops. These gradual changes in corticostriatal synaptic strengths produce long-lasting changes in behavioral responses. Under normal conditions, these mechanisms enable the selection of the most appropriate responses while inhibiting others. However, under dysregulated dopamine conditions, including a lack of dopamine release or dopamine signaling, these mechanisms could lead to the selection of maladaptive responses and/or the inhibition of appropriate responses in an experience-dependent and task-specific manner. In this review, we propose that preventing or reversing such maladaptive synaptic strengths and erasing such aberrant "memories" could be a disease-modifying therapeutic strategy for many neurological and psychiatric disorders. We review evidence from Parkinson's disease, drug-induced parkinsonism, L-DOPA-induced dyskinesia, obsessive-compulsive disorder, substance use disorders, and depression as well as research findings on animal disease models. Altogether, these studies allude to an emerging theme in translational neuroscience and promising new directions for therapy development. Specifically, we propose that combining pharmacotherapy with behavioral therapy or with deep brain stimulation (DBS) could potentially cause desired changes in specific neural circuits. If successful, one important advantage of correcting aberrant synaptic plasticity is long-lasting therapeutic effects even after treatment has ended. We will also discuss the potential molecular targets for these therapeutic approaches, including the cAMP pathway, proteins involved in synaptic plasticity as well as pathways involved in new protein synthesis. We place special emphasis on RNA binding proteins and epitranscriptomic mechanisms, as they represent a new frontier with the distinct advantage of rapidly and simultaneously altering the synthesis of many proteins locally.
Collapse
Affiliation(s)
- Zhuoyue Shi
- The Committee on Genetics, Genomics and Systems Biology, The University of Chicago, Chicago, IL, 60637, USA
| | - Kailong Wen
- The Committee on Neurobiology, The University of Chicago, Chicago, IL, 60637, USA
| | - Nabilah H Sammudin
- The Committee on Neurobiology, The University of Chicago, Chicago, IL, 60637, USA
| | - Nicholas LoRocco
- The Interdisciplinary Scientist Training Program, The University of Chicago, Chicago, IL, 60637, USA
| | - Xiaoxi Zhuang
- The Department of Neurobiology, The University of Chicago, Chicago, IL, 60637, USA.
- The Neuroscience Institute, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
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
|
7
|
Zimbelman AR, Wong B, Murray CH, Wolf ME, Stefanik MT. Dopamine D1 and NMDA Receptor Co-Regulation of Protein Translation in Cultured Nucleus Accumbens Neurons. Neurochem Res 2024; 50:27. [PMID: 39567459 PMCID: PMC11888153 DOI: 10.1007/s11064-024-04283-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/22/2024]
Abstract
Protein translation is essential for some forms of synaptic plasticity. Here we used fluorescent noncanonical amino acid tagging (FUNCAT) to examine whether dopamine modulates protein translation in cultured nucleus accumbens (NAc) medium spiny neurons (MSN). These neurons were co-cultured with cortical neurons to restore excitatory synapses. We measured translation in MSNs under basal conditions and after disinhibiting excitatory transmission using the GABAA receptor antagonist bicuculline (2 h). Under basal conditions, translation was not altered by the D1-class receptor (D1R) agonist SKF81297 or the D2-class receptor (D2R) agonist quinpirole. Bicuculline alone robustly increased translation. This was reversed by quinpirole but not SKF81297. It was also reversed by co-incubation with the D1R antagonist SCH23390, but not the D2R antagonist eticlopride, suggesting dopaminergic tone at D1Rs. This was surprising because no dopamine neurons are present. An alternative explanation is that bicuculline activates translation by increasing glutamate tone at NMDA receptors (NMDAR) within D1R/NMDAR heteromers. Supporting this, immunocytochemistry and proximity ligation assays revealed D1R/NMDAR heteromers on NAc cells both in vitro and in vivo, confirming previous results. Furthermore, bicuculline's effect was reversed to the same extent by SCH23390 alone, the NMDAR antagonist APV alone, or SCH23390 + APV. These results suggest that: (1) excitatory transmission stimulates translation in NAc MSNs, (2) this is opposed when glutamate activates D1R/NMDAR heteromers, even in the absence of dopamine, and (3) antagonist occupation of D1Rs within the heteromers prevents their activation. Our study is the first to suggest a role for D2 receptors and D1R/NMDAR heteromers in regulating protein translation.
Collapse
Affiliation(s)
- Alexa R Zimbelman
- Department of Psychology and Neuroscience, North Central College, 30 N. Brainard St., Naperville, IL, 60540, USA
| | - Benjamin Wong
- Department of Psychology and Neuroscience, North Central College, 30 N. Brainard St., Naperville, IL, 60540, USA
| | - Conor H Murray
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
- Present Address: UCLA Center for Cannabis and Cannabinoids, Semel Institute for Neuroscience & Human Behavior, Los Angeles, CA, 90025, USA
| | - Marina E Wolf
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
- Present Address: Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97212, USA
| | - Michael T Stefanik
- Department of Psychology and Neuroscience, North Central College, 30 N. Brainard St., Naperville, IL, 60540, USA.
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA.
| |
Collapse
|
8
|
Wunsch AM, Hwang EK, Funke JR, Baker R, Moutier A, Milovanovic M, Green TA, Wolf ME. Retinoic acid-mediated homeostatic plasticity in the nucleus accumbens core contributes to incubation of cocaine craving. Psychopharmacology (Berl) 2024; 241:1983-2001. [PMID: 38935096 DOI: 10.1007/s00213-024-06612-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: 12/18/2023] [Accepted: 05/10/2024] [Indexed: 06/28/2024]
Abstract
RATIONALE Incubation of cocaine craving refers to the progressive intensification of cue-induced craving during abstinence from cocaine self-administration. We showed previously that homomeric GluA1 Ca2+-permeable AMPARs (CP-AMPAR) accumulate in excitatory synapses of nucleus accumbens core (NAcc) medium spiny neurons (MSN) after ∼1 month of abstinence and thereafter their activation is required for expression of incubation. Therefore, it is important to understand mechanisms underlying CP-AMPAR plasticity. OBJECTIVES We hypothesize that CP-AMPAR upregulation represents a retinoic acid (RA)-dependent form of homeostatic plasticity, previously described in other brain regions, in which a reduction in neuronal activity disinhibits RA synthesis, leading to GluA1 translation and CP-AMPAR synaptic insertion. We tested this using viral vectors to bidirectionally manipulate RA signaling in NAcc during abstinence following extended-access cocaine self-administration. RESULTS We used shRNA targeted to the RA degradative enzyme Cyp26b1 to increase RA signaling. This treatment accelerated incubation; rats expressed incubation on abstinence day (AD) 15, when it is not yet detected in control rats. It also accelerated CP-AMPAR synaptic insertion measured with slice physiology. CP-AMPARs were detected in Cyp26b1 shRNA-expressing MSN, but not control MSN, on AD15-18. Next, we used shRNA targeted to the major RA synthetic enzyme Aldh1a1 to reduce RA signaling. In MSN expressing Aldh1a1 shRNA, synaptic CP-AMPARs were reduced in late withdrawal (AD42-60) compared to controls. However, we did not detect an effect of this manipulation on incubated cocaine seeking (AD40). CONCLUSIONS These findings support the hypothesis that increased RA signaling during abstinence contributes to CP-AMPAR accumulation and incubation of cocaine craving.
Collapse
Affiliation(s)
- Amanda M Wunsch
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97212, USA
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
| | - Eun-Kyung Hwang
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97212, USA
| | - Jonathan R Funke
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97212, USA
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
- Graduate School of Biomedical Sciences, Tufts University, Boston, MA, 02111, USA
| | - Raines Baker
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97212, USA
- College of Education, Health, and Human Sciences, Florida State University, Tallahassee, FL, 32306, USA
| | - Alana Moutier
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97212, USA
- Yecuris Corporation, Tualatin, OR, 97062, USA
| | - Mike Milovanovic
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
| | - Thomas A Green
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Marina E Wolf
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97212, USA.
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA.
| |
Collapse
|
9
|
Kawa AB, Hashimoto JG, Beutler MM, Guizzetti M, Wolf ME. Changes in nucleus accumbens core translatome accompanying incubation of cocaine craving. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.15.613147. [PMID: 39345421 PMCID: PMC11429699 DOI: 10.1101/2024.09.15.613147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
In the 'incubation of cocaine craving' model of relapse, rats exhibit progressive intensification (incubation) of cue-induced craving over several weeks of forced abstinence from cocaine self-administration. The expression of incubated craving depends on plasticity of excitatory synaptic transmission in nucleus accumbens core (NAcC) medium spiny neurons (MSN). Previously, we found that the maintenance of this plasticity and the expression of incubation depends on ongoing protein translation, and the regulation of translation is altered after incubation of cocaine craving. Here we used male and female rats that express Cre recombinase in either dopamine D1 receptor- or adenosine 2a (A2a) receptor-expressing MSN to express a GFP-tagged ribosomal protein in a cell-type specific manner, enabling us to use Translating Ribosome Affinity Purification (TRAP) to isolate actively translating mRNAs from both MSN subtypes for analysis by RNA-seq. We compared rats that self-administered saline or cocaine. Saline rats were assessed on abstinence day (AD) 1, while cocaine rats were assessed on AD1 or AD40-50. For both D1-MSN and A2a-MSN, there were few differentially translated genes between saline and cocaine AD1 groups. In contrast, pronounced differences in the translatome were observed between cocaine rats on AD1 and AD40-50, and this was far more robust in D1-MSN. Notably, all comparisons revealed sex differences in translating mRNAs. Sequencing results were validated by qRT-PCR for several genes of interest. This study, the first to combine TRAP-seq, transgenic rats, and a cocaine self-administration paradigm, identifies translating mRNAs linked to incubation of cocaine craving in D1-MSN and A2a-MSN of the NAcC.
Collapse
Affiliation(s)
- Alex B Kawa
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239
| | - Joel G Hashimoto
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239
- VA Portland Health Care System, Portland, OR 97239
| | - Madelyn M Beutler
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239
| | - Marina Guizzetti
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239
- VA Portland Health Care System, Portland, OR 97239
| | - Marina E Wolf
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239
| |
Collapse
|
10
|
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
|
11
|
Shi Z, Wen K, Zou Z, Fu W, Guo K, Sammudin NH, Ruan X, Sullere S, Wang S, Zhang X, Thinakaran G, He C, Zhuang X. YTHDF1 mediates translational control by m6A mRNA methylation in adaptation to environmental challenges. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.07.607063. [PMID: 39149343 PMCID: PMC11326287 DOI: 10.1101/2024.08.07.607063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Animals adapt to environmental challenges with long-term changes at the behavioral, circuit, cellular, and synaptic levels which often require new protein synthesis. The discovery of reversible N6-methyladenosine (m6A) modifications of mRNA has revealed an important layer of post-transcriptional regulation which affects almost every phase of mRNA metabolism and therefore translational control. Many in vitro and in vivo studies have demonstrated the significant role of m6A in cell differentiation and survival, but its role in adult neurons is understudied. We used cell-type specific gene deletion of Mettl14, which encodes one of the subunits of the m6A methyltransferase, and Ythdf1, which encodes one of the cytoplasmic m6A reader proteins, in dopamine D1 receptor expressing or D2 receptor expressing neurons. Mettl14 or Ythdf1 deficiency blunted responses to environmental challenges at the behavioral, cellular, and molecular levels. In three different behavioral paradigms, gene deletion of either Mettl14 or Ythdf1 in D1 neurons impaired D1-dependent learning, whereas gene deletion of either Mettl14 or Ythdf1 in D2 neurons impaired D2-dependent learning. At the cellular level, modulation of D1 and D2 neuron firing in response to changes in environments was blunted in all three behavioral paradigms in mutant mice. Ythdf1 deletion resembled impairment caused by Mettl14 deletion in a cell type-specific manner, suggesting YTHDF1 is the main mediator of the functional consequences of m6A mRNA methylation in the striatum. At the molecular level, while striatal neurons in control mice responded to elevated cAMP by increasing de novo protein synthesis, striatal neurons in Ythdf1 knockout mice didn't. Finally, boosting dopamine release by cocaine drastically increased YTHDF1 binding to many mRNA targets in the striatum, especially those that encode structural proteins, suggesting the initiation of long-term neuronal and/or synaptic structural changes. While the m6A-YTHDF1 pathway has similar functional significance at cellular level, its cell type specific deficiency in D1 and D2 neurons often resulted in contrasting behavioral phenotypes, allowing us to cleanly dissociate the opposing yet cooperative roles of D1 and D2 neurons.
Collapse
Affiliation(s)
- Zhuoyue Shi
- The Department of Neurobiology, University of Chicago, Chicago, IL 60637, USA
| | - Kailong Wen
- Committee on Neurobiology, University of Chicago, Chicago, IL 60637, USA
| | - Zhongyu Zou
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Wenqin Fu
- The Department of Neurobiology, University of Chicago, Chicago, IL 60637, USA
| | - Kathryn Guo
- The Department of Neurobiology, University of Chicago, Chicago, IL 60637, USA
| | - Nabilah H Sammudin
- Committee on Neurobiology, University of Chicago, Chicago, IL 60637, USA
| | - Xiangbin Ruan
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
| | - Shivang Sullere
- Committee on Neurobiology, University of Chicago, Chicago, IL 60637, USA
| | - Shuai Wang
- Byrd Alzheimer's Center and Research Institute, University of South Florida, Tampa, FL 33613, USA
| | - Xiaochang Zhang
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
- The Neuroscience Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Gopal Thinakaran
- Byrd Alzheimer's Center and Research Institute, University of South Florida, Tampa, FL 33613, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Chuan He
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Howard Hughes Medical Institute, The University of Chicago, IL 60637, USA
| | - Xiaoxi Zhuang
- The Department of Neurobiology, University of Chicago, Chicago, IL 60637, USA
- The Neuroscience Institute, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
12
|
Fetterly TL, Catalfio AM, Ferrario CR. Effects of junk-food on food-motivated behavior and nucleus accumbens glutamate plasticity; insights into the mechanism of calcium-permeable AMPA receptor recruitment. Neuropharmacology 2024; 242:109772. [PMID: 37898332 PMCID: PMC10883075 DOI: 10.1016/j.neuropharm.2023.109772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/03/2023] [Accepted: 10/17/2023] [Indexed: 10/30/2023]
Abstract
In rats, eating obesogenic diets increases calcium-permeable AMPA receptor (CP-AMPAR) transmission in the nucleus accumbens (NAc) core, and enhances food-motivated behavior. Interestingly, these diet-induced alterations in NAc transmission are pronounced and sustained in obesity-prone (OP) male rats and absent in obesity-resistant (OR) populations. However, effects of diet manipulation on food motivation, and the mechanisms underlying this NAc plasticity in OPs is unknown. Using male selectively-bred OP and OR rats, we assessed food-motivated behavior following ad lib access to chow (CH), junk-food (JF), or 10d of JF followed by a return to chow diet (JF-Dep). Motivation for food was greater in OP than OR rats, as expected. However, JF-Dep only produced enhancements in food-seeking in OP groups, while continuous JF access reduced food-seeking in both OPs and ORs. Additionally, optogenetic, chemogenetic, and pharmacological approaches were used to examine NAc CP-AMPAR recruitment following diet manipulation and ex vivo treatment of brain slices. Reducing excitatory transmission in the NAc was sufficient to recruit CP-AMPARs to synapses in OPs, but not ORs. In OPs, JF-induced increases in CP-AMPARs occurred in mPFC-, but not BLA-to-NAc inputs. Together results show that diet differentially affects behavioral and neural plasticity in obesity susceptible populations. We also identify conditions for acute recruitment of NAc CP-AMPARs; these results suggest that synaptic scaling mechanisms contribute to NAc CP-AMPAR recruitment. Overall, this work helps elucidate how diet interacts with obesity susceptibility to influence food-motivated behavior and extends our fundamental understanding of NAc CP-AMPAR recruitment.
Collapse
Affiliation(s)
- Tracy L Fetterly
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Amanda M Catalfio
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Carrie R Ferrario
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, 48109, USA; Psychology Department (Biopsychology) University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
13
|
Castillo Díaz F, Mottarlini F, Targa G, Rizzi B, Fumagalli F, Caffino L. Recency memory is altered in cocaine-withdrawn adolescent rats: Implication of cortical mTOR signaling. Prog Neuropsychopharmacol Biol Psychiatry 2023; 127:110822. [PMID: 37442333 DOI: 10.1016/j.pnpbp.2023.110822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 06/28/2023] [Accepted: 07/06/2023] [Indexed: 07/15/2023]
Abstract
In humans, cocaine abuse during adolescence poses a significant risk for developing cognitive deficits later in life. Among the regions responsible for cognitive processes, the medial prefrontal cortex (mPFC) modulates temporal order information via mechanisms involving the mammalian-target of rapamycin (mTOR)-mediated pathway and protein synthesis regulation. Accordingly, our goal was to study the effect of repeated cocaine exposure during both adolescence and adulthood on temporal memory by studying the mTOR pathway in the mPFC. Adolescent or adult rats underwent repeated cocaine injections for 15 days and, after two weeks of withdrawal, engaged in the temporal order object recognition (TOOR) test. We found that repeated cocaine exposure during adolescence impaired TOOR performance, while control or adult-treated animals showed no impairments. Moreover, activation of the mTOR-S6-eEF2 pathway following the TOOR test was diminished only in the adolescent cocaine-treated group. Notably, inhibition of the mTOR-mediated pathway by rapamycin injection impaired TOOR performance in naïve adolescent and adult animals, revealing this pathway to be a critical component in regulating recency memory. Our data indicate that withdrawal from cocaine exposure impairs recency memory via the dysregulation of protein translation mechanisms, but only when cocaine is administered during adolescence.
Collapse
Affiliation(s)
- Fernando Castillo Díaz
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan 20133, Italy; Department of Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg 93053, Germany
| | - Francesca Mottarlini
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan 20133, Italy
| | - Giorgia Targa
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan 20133, Italy
| | - Beatrice Rizzi
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan 20133, Italy; Center for Neuroscience, University of Camerino, Camerino 62032, Italy
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan 20133, Italy.
| | - Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan 20133, Italy
| |
Collapse
|
14
|
Gupta SC, Taugher-Hebl RJ, Hardie JB, Fan R, LaLumiere RT, Wemmie JA. Effects of acid-sensing ion channel-1A (ASIC1A) on cocaine-induced synaptic adaptations. Front Physiol 2023; 14:1191275. [PMID: 37389125 PMCID: PMC10300415 DOI: 10.3389/fphys.2023.1191275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/02/2023] [Indexed: 07/01/2023] Open
Abstract
Chronic drug abuse is thought to induce synaptic changes in nucleus accumbens medium spiny neurons (MSNs) that promote subsequent craving and drug-seeking behavior. Accumulating data suggest acid-sensing ion channels (ASICs) may play a critical role. In drug naïve mice, disrupting the ASIC1A subunit produced a variety of synaptic changes reminiscent of wild-type mice following cocaine withdrawal, including increased AMPAR/NMDAR ratio, increased AMPAR rectification, and increased dendrite spine density. Importantly, these changes in Asic1a -/- mice were normalized by a single dose of cocaine. Here we sought to understand the temporal effects of cocaine exposure in Asic1a -/- mice and the cellular site of ASIC1A action. Six hours after cocaine exposure, there was no effect. However, 15 h, 24 h and 4 days after cocaine exposure there was a significant reduction in AMPAR/NMDAR ratio in Asic1a -/- mice. Within 7 days the AMPAR/NMDAR ratio had returned to baseline levels. Cocaine-evoked changes in AMPAR rectification and dendritic spine density followed a similar time course with significant reductions in rectification and dendritic spines 24 h after cocaine exposure in Asic1a -/- mice. To test the cellular site of ASIC1A action on these responses, we disrupted ASIC1A specifically in a subpopulation of MSNs. We found that effects of ASIC1A disruption were cell autonomous and restricted to neurons in which the channels are disrupted. We further tested whether ASIC1A disruption differentially affects MSNs subtypes and found AMPAR/NMDAR ratio was elevated in dopamine receptor 1-expressing MSNs, suggesting a preferential effect for these cells. Finally, we tested if protein synthesis was involved in synaptic adaptations that occurred after ASIC1A disruption, and found the protein synthesis inhibitor anisomycin normalized AMPAR-rectification and AMPAR/NMDAR ratio in drug-naïve Asic1a -/- mice to control levels, observed in wild-type mice. Together, these results provide valuable mechanistic insight into the effects of ASICs on synaptic plasticity and drug-induced effects and raise the possibility that ASIC1A might be therapeutically manipulated to oppose drug-induced synaptic changes and behavior.
Collapse
Affiliation(s)
- Subhash C. Gupta
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States
- Department of Veterans Affairs Medical Center, Iowa City, IA, United States
| | - Rebecca J. Taugher-Hebl
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States
- Department of Veterans Affairs Medical Center, Iowa City, IA, United States
| | - Jason B. Hardie
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States
- Department of Veterans Affairs Medical Center, Iowa City, IA, United States
| | - Rong Fan
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States
- Department of Veterans Affairs Medical Center, Iowa City, IA, United States
| | - Ryan T. LaLumiere
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, United States
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, United States
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, United States
| | - John A. Wemmie
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States
- Department of Veterans Affairs Medical Center, Iowa City, IA, United States
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, United States
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, United States
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, United States
- Medical Scientist Training Program, University of Iowa, Iowa City, IA, United States
- Department of Neurosurgery, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
15
|
Ketchesin KD, Becker-Krail DD, Xue X, Wilson RS, Lam TT, Williams KR, Nairn AC, Tseng GC, Logan RW. Differential Effects of Cocaine and Morphine on the Diurnal Regulation of the Mouse Nucleus Accumbens Proteome. J Proteome Res 2023. [PMID: 37311105 PMCID: PMC10392613 DOI: 10.1021/acs.jproteome.3c00126] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Substance use disorders are associated with disruptions in sleep and circadian rhythms that persist during abstinence and may contribute to relapse risk. Repeated use of substances such as psychostimulants and opioids may lead to significant alterations in molecular rhythms in the nucleus accumbens (NAc), a brain region central to reward and motivation. Previous studies have identified rhythm alterations in the transcriptome of the NAc and other brain regions following the administration of psychostimulants or opioids. However, little is known about the impact of substance use on the diurnal rhythms of the proteome in the NAc. We used liquid chromatography coupled to tandem mass spectrometry-based quantitative proteomics, along with a data-independent acquisition analysis pipeline, to investigate the effects of cocaine or morphine administration on diurnal rhythms of proteome in the mouse NAc. Overall, our data reveal cocaine and morphine differentially alter diurnal rhythms of the proteome in the NAc, with largely independent differentially expressed proteins dependent on time-of-day. Pathways enriched from cocaine altered protein rhythms were primarily associated with glucocorticoid signaling and metabolism, whereas morphine was associated with neuroinflammation. Collectively, these findings are the first to characterize the diurnal regulation of the NAc proteome and demonstrate a novel relationship between the phase-dependent regulation of protein expression and the differential effects of cocaine and morphine on the NAc proteome. The proteomics data in this study are available via ProteomeXchange with identifier PXD042043.
Collapse
Affiliation(s)
- Kyle D Ketchesin
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Darius D Becker-Krail
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Xiangning Xue
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Rashaun S Wilson
- Yale/NIDA Neuroproteomics Center, 300 George Street, New Haven, Connecticut 06511, United States
- W.M. Keck Biotechnology Mass Spectrometry (MS) & Proteomics Resource Laboratory, Yale University School of Medicine, New Haven, Connecticut 06511, United States
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - TuKiet T Lam
- Yale/NIDA Neuroproteomics Center, 300 George Street, New Haven, Connecticut 06511, United States
- W.M. Keck Biotechnology Mass Spectrometry (MS) & Proteomics Resource Laboratory, Yale University School of Medicine, New Haven, Connecticut 06511, United States
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Kenneth R Williams
- Yale/NIDA Neuroproteomics Center, 300 George Street, New Haven, Connecticut 06511, United States
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Angus C Nairn
- Yale/NIDA Neuroproteomics Center, 300 George Street, New Haven, Connecticut 06511, United States
- Department of Psychiatry, Yale University School of Medicine, Connecticut Mental Health Center, New Haven, Connecticut 06511, United States
| | - George C Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Ryan W Logan
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01655, United States
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, United States
| |
Collapse
|
16
|
Fetterly TL, Catalfio AM, Ferrario CR. Effects of junk-food on food-motivated behavior and NAc glutamate plasticity; insights into the mechanism of NAc calcium-permeable AMPA receptor recruitment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.16.540977. [PMID: 37292760 PMCID: PMC10245687 DOI: 10.1101/2023.05.16.540977] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In rats, eating obesogenic diets increase calcium-permeable AMPA receptor (CP-AMPAR) transmission in the nucleus accumbens (NAc) core, and enhances food-motivated behavior. Interestingly these diet-induced alterations in NAc transmission are pronounced in obesity-prone (OP) rats and absent in obesity-resistant (OR) populations. However, effects of diet manipulation on food motivation, and the mechanisms underlying NAc plasticity in OPs is unknown. Using male selectively-bred OP and OR rats, we assessed food-motivated behavior following ad lib access to chow (CH), junk-food (JF), or 10d of JF followed by a return to chow diet (JF-Dep). Behavioral tests included conditioned reinforcement, instrumental responding, and free consumption. Additionally, optogenetic, chemogenetic, and pharmacological approaches were used to examine NAc CP-AMPAR recruitment following diet manipulation and ex vivo treatment of brain slices. Motivation for food was greater in OP than OR rats, as expected. However, JF-Dep only produced enhancements in food-seeking in OP groups, while continuous JF access reduced food-seeking in both OPs and ORs. Reducing excitatory transmission in the NAc was sufficient to recruit CP-AMPARs to synapses in OPs, but not ORs. In OPs, JF-induced increases in CP-AMPARs occurred in mPFC-, but not BLA-to-NAc inputs. Diet differentially affects behavioral and neural plasticity in obesity susceptible populations. We also identify conditions for acute recruitment of NAc CP-AMPARs; these results suggest that synaptic scaling mechanisms contribute to NAc CP-AMPAR recruitment. Overall, this work improves our understanding of how sugary, fatty food consumption interacts with obesity susceptibility to influence food-motivated behavior. It also extends our fundamental understanding of NAc CP-AMPAR recruitment; this has important implications for motivation in the context of obesity as well as drug addiction.
Collapse
Affiliation(s)
- Tracy L. Fetterly
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Amanda M. Catalfio
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Carrie R. Ferrario
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
17
|
Coombs I, Bats C, Sexton CA, Studniarczyk D, Cull-Candy SG, Farrant M. Enhanced functional detection of synaptic calcium-permeable AMPA receptors using intracellular NASPM. eLife 2023; 12:e66765. [PMID: 37042655 PMCID: PMC10168695 DOI: 10.7554/elife.66765] [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: 01/22/2021] [Accepted: 04/11/2023] [Indexed: 04/13/2023] Open
Abstract
Calcium-permeable AMPA-type glutamate receptors (CP-AMPARs) contribute to many forms of synaptic plasticity and pathology. They can be distinguished from GluA2-containing calcium-impermeable AMPARs by the inward rectification of their currents, which reflects voltage-dependent channel block by intracellular spermine. However, the efficacy of this weakly permeant blocker is differentially altered by the presence of AMPAR auxiliary subunits - including transmembrane AMPAR regulatory proteins, cornichons, and GSG1L - which are widely expressed in neurons and glia. This complicates the interpretation of rectification as a measure of CP-AMPAR expression. Here, we show that the inclusion of the spider toxin analog 1-naphthylacetyl spermine (NASPM) in the intracellular solution results in a complete block of GluA1-mediated outward currents irrespective of the type of associated auxiliary subunit. In neurons from GluA2-knockout mice expressing only CP-AMPARs, intracellular NASPM, unlike spermine, completely blocks outward synaptic currents. Thus, our results identify a functional measure of CP-AMPARs, that is unaffected by their auxiliary subunit content.
Collapse
Affiliation(s)
- Ian Coombs
- Department of Neuroscience, Physiology and Pharmacology, University College LondonLondonUnited Kingdom
| | - Cécile Bats
- Department of Neuroscience, Physiology and Pharmacology, University College LondonLondonUnited Kingdom
| | - Craig A Sexton
- Department of Neuroscience, Physiology and Pharmacology, University College LondonLondonUnited Kingdom
| | - Dorota Studniarczyk
- Department of Neuroscience, Physiology and Pharmacology, University College LondonLondonUnited Kingdom
| | - Stuart G Cull-Candy
- Department of Neuroscience, Physiology and Pharmacology, University College LondonLondonUnited Kingdom
| | - Mark Farrant
- Department of Neuroscience, Physiology and Pharmacology, University College LondonLondonUnited Kingdom
| |
Collapse
|
18
|
Zimbelman AR, Wong B, Murray CH, Wolf ME, Stefanik MT. Dopamine D1 and NMDA receptor co-regulation of protein translation in cultured nucleus accumbens neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.02.535293. [PMID: 37034633 PMCID: PMC10081306 DOI: 10.1101/2023.04.02.535293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Protein translation is essential for some forms of synaptic plasticity. We used nucleus accumbens (NAc) medium spiny neurons (MSN), co-cultured with cortical neurons to restore excitatory synapses, to examine whether dopamine modulates protein translation in NAc MSN. FUNCAT was used to measure translation in MSNs under basal conditions and after disinhibiting excitatory transmission using the GABAA receptor antagonist bicuculline (2 hr). Under basal conditions, translation was not altered by the D1-class receptor (D1R) agonist SKF81297 or the D2-class receptor (D2R) agonist quinpirole. Bicuculline alone robustly increased translation. This was reversed by quinpirole but not SKF81297. It was also reversed by co-incubation with the D1R antagonist SCH23390, but not the D2R antagonist eticlopride, suggesting dopaminergic tone at D1Rs. This was surprising because no dopamine neurons are present. An alternative explanation is that bicuculline activates translation by increasing glutamate tone at NMDA receptors (NMDAR) within D1R/NMDAR heteromers, which have been described in other cell types. Supporting this, immunocytochemistry and proximity ligation assays revealed D1/NMDAR heteromers on NAc cells both in vitro and in vivo. Further, bicuculline's effect was reversed to the same extent by SCH23390 alone, the NMDAR antagonist APV alone, or SCH23390+APV. These results suggest that: 1) excitatory synaptic transmission stimulates translation in NAc MSNs, 2) this is opposed when glutamate activates D1R/NMDAR heteromers, even in the absence of dopamine, and 3) antagonist occupation of D1Rs within the heteromers prevents their activation. Our study is the first to suggest a role for D2 receptors and D1R/NMDAR heteromers in regulating protein translation.
Collapse
Affiliation(s)
- Alexa R. Zimbelman
- Department of Psychology and Neuroscience, North Central College, Naperville, IL 60540
| | - Benjamin Wong
- Department of Psychology and Neuroscience, North Central College, Naperville, IL 60540
| | - Conor H. Murray
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064
- Present address: Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL
| | - Marina E. Wolf
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064
- These authors contributed equally
- Present address: Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97212
| | - Michael T. Stefanik
- Department of Psychology and Neuroscience, North Central College, Naperville, IL 60540
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064
- These authors contributed equally
| |
Collapse
|
19
|
Ketchesin KD, Becker-Krail DD, Xue X, Wilson RS, Lam TT, Williams KR, Nairn AC, Tseng GC, Logan RW. Differential Effects of Cocaine and Morphine on the Diurnal Regulation of the Mouse Nucleus Accumbens Proteome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.01.530696. [PMID: 36909659 PMCID: PMC10002738 DOI: 10.1101/2023.03.01.530696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Substance use disorders (SUDs) are associated with disruptions in sleep and circadian rhythms that persist during abstinence and may contribute to relapse risk. Repeated use of substances such as psychostimulants and opioids may lead to significant alterations in molecular rhythms in the nucleus accumbens (NAc), a brain region central to reward and motivation. Previous studies have identified rhythm alterations in the transcriptome of the NAc and other brain regions following the administration of psychostimulants or opioids. However, little is known about the impact of substance use on the diurnal rhythms of the proteome in the NAc. We used liquid chromatography coupled to tandem mass spectrometry-based (LC-MS/MS) quantitative proteomics, along with a data-independent acquisition (DIA) analysis pipeline, to investigate the effects of cocaine or morphine administration on diurnal rhythms of proteome in the mouse NAc. Overall, our data reveals cocaine and morphine differentially alters diurnal rhythms of the proteome in the NAc, with largely independent differentially expressed proteins dependent on time-of-day. Pathways enriched from cocaine altered protein rhythms were primarily associated with glucocorticoid signaling and metabolism, whereas morphine was associated with neuroinflammation. Collectively, these findings are the first to characterize the diurnal regulation of the NAc proteome and demonstrate a novel relationship between phase-dependent regulation of protein expression and the differential effects of cocaine and morphine on the NAc proteome.
Collapse
|
20
|
Borland JM, Dempsey DA, Peyla AC, Hall MAL, Kohut-Jackson AL, Mermelstein PG, Meisel RL. Aggression Results in the Phosphorylation of ERK1/2 in the Nucleus Accumbens and the Dephosphorylation of mTOR in the Medial Prefrontal Cortex in Female Syrian Hamsters. Int J Mol Sci 2023; 24:1379. [PMID: 36674893 PMCID: PMC9862940 DOI: 10.3390/ijms24021379] [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] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/22/2022] [Accepted: 12/28/2022] [Indexed: 01/13/2023] Open
Abstract
Like many social behaviors, aggression can be rewarding, leading to behavioral plasticity. One outcome of reward-induced aggression is the long-term increase in the speed in which future aggression-based encounters is initiated. This form of aggression impacts dendritic structure and excitatory synaptic neurotransmission in the nucleus accumbens, a brain region well known to regulate motivated behaviors. Yet, little is known about the intracellular signaling mechanisms that drive these structural/functional changes and long-term changes in aggressive behavior. This study set out to further elucidate the intracellular signaling mechanisms regulating the plasticity in neurophysiology and behavior that underlie the rewarding consequences of aggressive interactions. Female Syrian hamsters experienced zero, two or five aggressive interactions and the phosphorylation of proteins in reward-associated regions was analyzed. We report that aggressive interactions result in a transient increase in the phosphorylation of extracellular-signal related kinase 1/2 (ERK1/2) in the nucleus accumbens. We also report that aggressive interactions result in a transient decrease in the phosphorylation of mammalian target of rapamycin (mTOR) in the medial prefrontal cortex, a major input structure to the nucleus accumbens. Thus, this study identifies ERK1/2 and mTOR as potential signaling pathways for regulating the long-term rewarding consequences of aggressive interactions. Furthermore, the recruitment profile of the ERK1/2 and the mTOR pathways are distinct in different brain regions.
Collapse
Affiliation(s)
| | - Desarae A. Dempsey
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Anna C. Peyla
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Megan A. L. Hall
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Paul G. Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Robert L. Meisel
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
21
|
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
|
22
|
Kawa AB, Hwang EK, Funke JR, Zhou H, Costa-Mattioli M, Wolf ME. Positive Allosteric Modulation of mGlu 1 Reverses Cocaine-Induced Behavioral and Synaptic Plasticity Through the Integrated Stress Response and Oligophrenin-1. Biol Psychiatry 2022; 92:871-879. [PMID: 35871097 PMCID: PMC10656746 DOI: 10.1016/j.biopsych.2022.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/21/2022] [Accepted: 05/05/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Cue-induced cocaine craving progressively intensifies (incubates) during abstinence from cocaine self-administration. Expression of incubated cocaine craving depends on elevated calcium-permeable AMPA receptors (CP-AMPARs) on medium spiny neurons in the nucleus accumbens (NAc) core. After incubation has occurred, stimulation of NAc metabotropic glutamate 1 (mGlu1) receptors or systemic administration of mGlu1 positive allosteric modulators removes CP-AMPARs from NAc synapses via dynamin-dependent internalization (mGlu1 long-term depression [LTD]) and thereby reduces incubated cocaine craving. Because mGlu1 positive allosteric modulators are potential therapeutics for cocaine craving, it is important to further define the mechanism triggering this mGlu1-LTD. METHODS Male and female rats self-administered saline or cocaine (10 days) using a long access regimen (6 h/day). Following ≥40 days of abstinence, we assessed the ability of an mGlu1 positive allosteric modulator to inhibit expression of incubated craving and remove CP-AMPARs from NAc synapses under control conditions, after blocking the integrated stress response (ISR), or after knocking down oligophrenin-1, a mediator of the ISR that can promote AMPAR endocytosis. AMPAR transmission in NAc medium spiny neurons was assessed with ex vivo slice recordings. RESULTS mGlu1 stimulation reduced cue-induced craving and removed synaptic CP-AMPARs. When the ISR was blocked prior to mGlu1 stimulation, there was no reduction in cue-induced craving, nor were CP-AMPARs removed from the synapse. Further, selective knockdown of oligophrenin-1 blocked mGlu1-LTD. CONCLUSIONS Our results indicate that mGlu1-LTD in the NAc and consequently the reduction of cue-induced seeking occur through activation of the ISR, which induces translation of oligophrenin-1. We also demonstrate CP-AMPAR accumulation and mGlu1 reversal in female rats, as previously shown in male rats.
Collapse
Affiliation(s)
- Alex B Kawa
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon
| | - Eun-Kyung Hwang
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon
| | - Jonathan R Funke
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon
| | - Hongyi Zhou
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas
| | | | - Marina E Wolf
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon.
| |
Collapse
|
23
|
Beiser T, Lisniansky E, Weitz M, Bingor A, Grad E, Rosenblum K, Thornton C, Yaka R. A functional eEF2K-eEF2 pathway in the NAc is critical for the expression of cocaine-induced psychomotor sensitisation and conditioned place preference. Transl Psychiatry 2022; 12:460. [PMID: 36319619 PMCID: PMC9626485 DOI: 10.1038/s41398-022-02232-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 01/24/2023] Open
Abstract
Recent evidence links synaptic plasticity and mRNA translation, via the eukaryotic elongation factor 2 kinase (eEF2K) and its only known substrate, eEF2. However, the involvement of the eEF2 pathway in cocaine-induced neuroadaptations and cocaine-induced behaviours is not known. Knock-in (KI) mice and shRNA were used to globally and specifically reduce eEF2K expression. Cocaine psychomotor sensitization and conditioned place preference were used to evaluate behavioural outcome. Changes in eEF2 phosphorylation were determined by western blot analyses. No effect was observed on the AMPA/NMDA receptor current ratio in the ventral tegmental area, 24 h after cocaine injection in eEF2K-KI mice compared with WT. However, development and expression of cocaine psychomotor sensitization were decreased in KI mice. Phosphorylated eEF2 was decreased one day after psychomotor sensitization and returned to baseline at seven days in the nucleus accumbens (NAc) of WT mice, but not in eEF2K-KI mice. However, one day following cocaine challenge, phosphorylated eEF2 decreased in WT but not KI mice. Importantly, specific targeting of eEF2K expression by shRNA in the NAc decreased cocaine condition place preference. These results suggest that the eEF2 pathway play a role in cocaine-induced locomotor sensitization and conditioned place preference.
Collapse
Affiliation(s)
- Tehila Beiser
- grid.9619.70000 0004 1937 0538Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Elvira Lisniansky
- grid.9619.70000 0004 1937 0538Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Moriya Weitz
- grid.9619.70000 0004 1937 0538Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Alexey Bingor
- grid.9619.70000 0004 1937 0538Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Etty Grad
- grid.9619.70000 0004 1937 0538Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Kobi Rosenblum
- grid.18098.380000 0004 1937 0562Sagol Department of Neuroscience, University of Haifa, Haifa, Israel
| | - Claire Thornton
- grid.20931.390000 0004 0425 573XDepartment of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Rami Yaka
- Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
24
|
Zinsmaier AK, Dong Y, Huang YH. Cocaine-induced projection-specific and cell type-specific adaptations in the nucleus accumbens. Mol Psychiatry 2022; 27:669-686. [PMID: 33963288 PMCID: PMC8691189 DOI: 10.1038/s41380-021-01112-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 03/29/2021] [Accepted: 04/09/2021] [Indexed: 02/03/2023]
Abstract
Cocaine craving, seeking, and relapse are mediated, in part, by cocaine-induced adaptive changes in the brain reward circuits. The nucleus accumbens (NAc) integrates and prioritizes different emotional and motivational inputs to the reward system by processing convergent glutamatergic projections from the medial prefrontal cortex, basolateral amygdala, ventral hippocampus, and other limbic and paralimbic brain regions. Medium spiny neurons (MSNs) are the principal projection neurons in the NAc, which can be divided into two major subpopulations, namely dopamine receptor D1- versus D2-expressing MSNs, with complementing roles in reward-associated behaviors. After cocaine experience, NAc MSNs exhibit complex and differential adaptations dependent on cocaine regimen, withdrawal time, cell type, location (NAc core versus shell), and related input and output projections, or any combination of these factors. Detailed characterization of these cellular adaptations has been greatly facilitated by the recent development of optogenetic/chemogenetic techniques combined with transgenic tools. In this review, we discuss such cell type- and projection-specific adaptations induced by cocaine experience. Specifically, (1) D1 and D2 NAc MSNs frequently exhibit differential adaptations in spinogenesis, glutamatergic receptor trafficking, and intrinsic membrane excitability, (2) cocaine experience differentially changes the synaptic transmission at different afferent projections onto NAc MSNs, (3) cocaine-induced NAc adaptations exhibit output specificity, e.g., being different at NAc-ventral pallidum versus NAc-ventral tegmental area synapses, and (4) the input, output, subregion, and D1/D2 cell type may together determine cocaine-induced circuit plasticity in the NAc. In light of the projection- and cell-type specificity, we also briefly discuss ensemble and circuit mechanisms contributing to cocaine craving and relapse after drug withdrawal.
Collapse
Affiliation(s)
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15219,Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219
| | - Yanhua H. Huang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219
| |
Collapse
|
25
|
Chiu AS, Kang MC, Huerta Sanchez LL, Fabella AM, Holder KN, Barger BD, Elias KN, Shin CB, Jimenez Chavez CL, Kippin TE, Szumlinski KK. Preclinical evidence to support repurposing everolimus for craving reduction during protracted drug withdrawal. Neuropsychopharmacology 2021; 46:2090-2100. [PMID: 34188183 PMCID: PMC8505628 DOI: 10.1038/s41386-021-01064-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 06/06/2021] [Accepted: 06/08/2021] [Indexed: 02/06/2023]
Abstract
Cue-elicited drug-craving is a cardinal feature of addiction that intensifies (incubates) during protracted withdrawal. In a rat model, these addiction-related behavioral pathologies are mediated, respectively, by time-dependent increases in PI3K/Akt1 signaling and reduced Group 1 metabotropic glutamate receptor (mGlu) expression, within the ventromedial prefrontal cortex (vmPFC). Herein, we examined the capacity of single oral dosing with everolimus, an FDA-approved inhibitor of the PI3K/Akt effector mTOR, to reduce incubated cocaine-craving and reverse incubation-associated changes in vmPFC kinase activity and mGlu expression. Rats were trained to lever-press for intravenous infusions of cocaine or delivery of sucrose pellets and then subjected to tests for cue-reinforced responding during early (3 days) or late (30-46 days) withdrawal. Rats were gavage-infused with everolimus (0-1.0 mg/kg), either prior to testing to examine for effects upon reinforcer-seeking behavior, or immediately following testing to probe effects upon the consolidation of extinction learning. Single oral dosing with everolimus dose-dependently blocked cocaine-seeking during late withdrawal and the effect lasted at least 24 h. No everolimus effects were observed for cue-elicited sucrose-seeking or cocaine-seeking in early withdrawal. In addition, everolimus treatment, following initial cue-testing, reduced subsequent cue hyper-responsivity exhibited observed during late withdrawal, arguing a facilitation of extinction memory consolidation. everolimus' "anti-incubation" effect was associated with a reversal of withdrawal-induced changes in indices of PI3K/Akt1/mTOR activity, as well as Homer protein and mGlu1/5 expression, within the prelimbic (PL) subregion of the prefrontal cortex. Our results indicate mTOR inhibition as a viable strategy for interrupting heightened cocaine-craving and facilitating addiction recovery during protracted withdrawal.
Collapse
Affiliation(s)
- Alvin S Chiu
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Matthew C Kang
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Laura L Huerta Sanchez
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Anne M Fabella
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Kalysta N Holder
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Brooke D Barger
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Kristina N Elias
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Christina B Shin
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, USA
| | - C Leonardo Jimenez Chavez
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Tod E Kippin
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, USA
- Department of Molecular, Developmental and Cell Biology and the Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
- Institute for Collaborative Biotechnologies, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Karen K Szumlinski
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, USA.
- Department of Molecular, Developmental and Cell Biology and the Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA.
| |
Collapse
|
26
|
Murray CH, Gaulden AD, Kawa AB, Milovanovic M, Caccamise AJ, Funke JR, Patel S, Wolf ME. CaMKII Modulates Diacylglycerol Lipase-α Activity in the Rat Nucleus Accumbens after Incubation of Cocaine Craving. eNeuro 2021; 8:ENEURO.0220-21.2021. [PMID: 34544759 PMCID: PMC8503962 DOI: 10.1523/eneuro.0220-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/10/2021] [Accepted: 08/19/2021] [Indexed: 01/04/2023] Open
Abstract
Relapse is a major challenge to the treatment of substance use disorders. A progressive increase in cue-induced drug craving, termed incubation of craving, is observed after withdrawal from multiple drugs of abuse in humans and rodents. Incubation of cocaine craving involves the strengthening of excitatory synapses onto nucleus accumbens (NAc) medium spiny neurons via postsynaptic accumulation of high-conductance Ca2+-permeable AMPA receptors. This enhances reactivity to drug-associated cues and is required for the expression of incubation. Additionally, incubation of cocaine craving is associated with loss of the synaptic depression normally triggered by stimulation of metabotropic glutamate receptor 5 (mGlu5), leading to endocannabinoid production, and expressed presynaptically via cannabinoid receptor 1 activation. Previous studies have found alterations in mGlu5 and Homer proteins associated with the loss of this synaptic depression. Here we conducted coimmunoprecipitation studies to investigate associations of diacylglycerol lipase-α (DGL), which catalyzes formation of the endocannabinoid 2-arachidonylglycerol (2-AG), with mGlu5 and Homer proteins. Although these interactions were unchanged in the NAc core at incubation-relevant withdrawal times, the association of DGL with total and phosphorylated Ca2+/calmodulin-dependent protein kinase IIα (CaMKIIα) and CaMKIIβ was increased. This would be predicted, based on other studies, to inhibit DGL activity and therefore 2-AG production. This was confirmed by measuring DGL enzymatic activity. However, the magnitude of DGL inhibition did not correlate with the magnitude of incubation of craving for individual rats. These results suggest that CaMKII contributes to the loss of mGlu5-dependent synaptic depression after incubation, but the functional significance of this loss remains unclear.
Collapse
Affiliation(s)
- Conor H Murray
- Department of Neuroscience, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064-3095
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon 97239-3098
| | - Andrew D Gaulden
- Department of Psychiatry, Vanderbilt University, Nashville, Tennessee 37240
| | - Alex B Kawa
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon 97239-3098
| | - Mike Milovanovic
- Department of Neuroscience, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064-3095
| | - Aaron J Caccamise
- Department of Neuroscience, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064-3095
| | - Jonathan R Funke
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon 97239-3098
| | - Sachin Patel
- Department of Psychiatry, Vanderbilt University, Nashville, Tennessee 37240
| | - Marina E Wolf
- Department of Neuroscience, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064-3095
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon 97239-3098
| |
Collapse
|
27
|
Li Y, He Y, Fan H, Wang Z, Huang J, Wen G, Wang X, Xie Q, Qiu P. Brain-derived neurotrophic factor upregulates synaptic GluA1 in the amygdala to promote depression in response to psychological stress. Biochem Pharmacol 2021; 192:114740. [PMID: 34419429 DOI: 10.1016/j.bcp.2021.114740] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 12/28/2022]
Abstract
Psychological stress impairs neuronal structure and function and leads to emotional disorders, but the underlying mechanisms have not yet been fully elucidated. The amygdala is closely correlated with emotional regulation. In the present study, we analyzed whether the amygdala plasticity is regulated by psychological stress and explored their regulatory mechanism. We established a mouse psychological stress model using an improved communication box, wherein mice were exposed to chronic fear and avoided physical stress interference. After the 14-day psychological stress paradigm, mice exhibited significantly increased depressive behaviors (decreased sucrose consumption in the sucrose preference test and longer immobility time in the forced swimming test). HPLC, ELISA, and molecular and morphological evidences showed that psychological stress increased the content of glutamate and the expression of glutamatergic neurons, upregulated the content of the stress hormone corticosterone, and activated the CREB/BDNF pathway in the amygdala. Furthermore, psychological stress induced an increased density of dendritic spines and LTD impairment in the amygdala. Importantly, virus-mediated silencing of BDNF in the basolateral amygdala (BLA) nuclei reversed the depression-like behaviors and the increase of synaptic GluA1 and its phosphorylation at Ser831 and Ser845 sites in psychologically stressed mice. This process was likely achieved through mTOR signaling activation. Finally, we treated primary amygdala neurons with corticosterone to mimic psychological stress; corticosterone-induced upregulation of GluA1 was prevented by BDNF and mTOR antagonists. Thus, activation of the CREB/BDNF pathway in the amygdala following psychological stress upregulates synaptic GluA1 via mTOR signaling, which dysregulates synaptic plasticity of the amygdala, eventually promoting depression.
Collapse
Affiliation(s)
- Yanning Li
- School of Forensic Medicine, Southern Medical University, Guangzhou, PR China; Department of Forensic Medicine, School of Basic Medicine, Gannan Medical University, Ganzhou, PR China
| | - Yitong He
- School of Forensic Medicine, Southern Medical University, Guangzhou, PR China
| | - Haoliang Fan
- School of Forensic Medicine, Southern Medical University, Guangzhou, PR China; School of Basic Medicine and Life Science, Hainan Medical University, Haikou, PR China
| | - Zhuo Wang
- Department of Infertility and Sexual Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China
| | - Jian Huang
- School of Forensic Medicine, Southern Medical University, Guangzhou, PR China
| | - Gehua Wen
- School of Forensic Medicine, China Medical University, Shenyang, PR China
| | - Xiaohan Wang
- School of Forensic Medicine, Southern Medical University, Guangzhou, PR China
| | - Qiqian Xie
- School of Forensic Medicine, Southern Medical University, Guangzhou, PR China
| | - Pingming Qiu
- School of Forensic Medicine, Southern Medical University, Guangzhou, PR China.
| |
Collapse
|
28
|
Wright WJ, Dong Y. Psychostimulant-Induced Adaptations in Nucleus Accumbens Glutamatergic Transmission. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a039255. [PMID: 31964644 DOI: 10.1101/cshperspect.a039255] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Carrying different aspects of emotional and motivational signals, glutamatergic synaptic projections from multiple limbic and paralimbic brain regions converge to the nucleus accumbens (NAc), in which these arousing signals are processed and prioritized for behavioral output. In animal models of drug addiction, some key drug-induced alterations at NAc glutamatergic synapses underlie important cellular and circuit mechanisms that promote subsequent drug taking, seeking, and relapse. With the focus of cocaine, we review changes at NAc glutamatergic synapses that occur after different drug procedures and abstinence durations, and the behavioral impact of these changes.
Collapse
Affiliation(s)
- William J Wright
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA
| |
Collapse
|
29
|
Ucha M, Roura-Martínez D, Ambrosio E, Higuera-Matas A. The role of the mTOR pathway in models of drug-induced reward and the behavioural constituents of addiction. J Psychopharmacol 2020; 34:1176-1199. [PMID: 32854585 DOI: 10.1177/0269881120944159] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Exposure to drugs of abuse induces neuroadaptations in critical nodes of the so-called reward systems that are thought to mediate the transition from controlled drug use to the compulsive drug-seeking that characterizes addictive disorders. These neural adaptations are likely to require protein synthesis, which is regulated, among others, by the mechanistic target of the rapamycin kinase (mTOR) signalling cascade. METHODS We have performed a narrative review of the literature available in PubMed about the involvement of the mTOR pathway in drug-reward and addiction-related phenomena. AIMS The aim of this study was to review the underlying architecture of this complex intracellular network and to discuss the alterations of its components that are evident after exposure to drugs of abuse. The aim was also to delineate the effects that manipulations of the mTOR network have on models of drug reward and on paradigms that recapitulate some of the psychological components of addiction. RESULTS There is evidence for the involvement of the mTOR pathway in the acute and rewarding effects of drugs of abuse, especially psychostimulants. However, the data regarding opiates are scarce. There is a need to use sophisticated animal models of addiction to ascertain the real role of the mTOR pathway in this pathology and not just in drug-mediated reward. The involvement of this pathway in behavioural addictions and impulsivity should also be studied in detail in the future. CONCLUSIONS Although there is a plethora of data about the modulation of mTOR by drugs of abuse, the involvement of this signalling pathway in addictive disorders requires further research.
Collapse
Affiliation(s)
- Marcos Ucha
- Department of Psychobiology, National University for Distance Learning (UNED), Madrid, Spain
| | - David Roura-Martínez
- Department of Psychobiology, National University for Distance Learning (UNED), Madrid, Spain
| | - Emilio Ambrosio
- Department of Psychobiology, National University for Distance Learning (UNED), Madrid, Spain
| | - Alejandro Higuera-Matas
- Department of Psychobiology, National University for Distance Learning (UNED), Madrid, Spain
| |
Collapse
|
30
|
AMPA receptor and metabotropic glutamate receptor 1 adaptations in the nucleus accumbens core during incubation of methamphetamine craving. Neuropsychopharmacology 2019; 44:1534-1541. [PMID: 31146278 PMCID: PMC6785134 DOI: 10.1038/s41386-019-0425-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 04/30/2019] [Accepted: 05/20/2019] [Indexed: 12/15/2022]
Abstract
Cue-induced drug craving progressively intensifies after withdrawal from self-administration of cocaine, methamphetamine, and other drugs of abuse, a phenomenon termed incubation of craving. For cocaine and methamphetamine, expression of incubated craving ultimately depends on strengthening of nucleus accumbens (NAc) synapses through an accumulation of high conductance Ca2+-permeable AMPA receptors (CP-AMPARs) that is detectable with electrophysiological approaches. This study sought to further characterize glutamate receptor adaptations in NAc core during methamphetamine incubation. Previous biochemical studies revealed that the CP-AMPARs accumulating after cocaine incubation are mainly homomeric GluA1 receptors and that their accumulation is reflected by increased cell surface GluA1. Here, for methamphetamine, we observed no significant change in surface or total GluA1 (GluA2 and GluA3 were also unchanged). Nonetheless, GluA1 translation was elevated after incubation of methamphetamine craving, as recently found for cocaine. Additionally, for cocaine, we previously observed a withdrawal-dependent decrease in mGlu1 surface expression that precedes and enables CP-AMPAR accumulation and incubation of craving, reflecting weakening of mGlu1-dependent mechanisms that normally limit synaptic CP-AMPAR levels in the NAc core. Here, we observed no change in surface or total mGlu1 protein or its coupling to Homer scaffolding proteins after methamphetamine withdrawal, nor did elevation of mGlu1 tone through repeated injections of an mGlu1-positive allosteric modulator delay incubation of craving. These findings suggest a common role for increased GluA1 translation, but not decreased mGlu1 function, in the incubation of methamphetamine and cocaine craving. We speculate that increased GluA1 translation near synapses may drive formation and synaptic insertion of homomeric GluA1 receptors in the absence of detectable changes in GluA1 protein levels.
Collapse
|
31
|
Di S, Jiang Z, Wang S, Harrison LM, Castro-Echeverry E, Stuart TC, Wolf ME, Tasker JG. Labile Calcium-Permeable AMPA Receptors Constitute New Glutamate Synapses Formed in Hypothalamic Neuroendocrine Cells during Salt Loading. eNeuro 2019; 6:ENEURO.0112-19.2019. [PMID: 31300543 PMCID: PMC6675872 DOI: 10.1523/eneuro.0112-19.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 06/13/2019] [Accepted: 06/21/2019] [Indexed: 11/21/2022] Open
Abstract
Magnocellular neuroendocrine cells (MNCs) of the hypothalamus play a critical role in the regulation of fluid and electrolyte homeostasis. They undergo a dramatic structural and functional plasticity under sustained hyperosmotic conditions, including an increase in afferent glutamatergic synaptic innervation. We tested for a postulated increase in glutamate AMPA receptor expression and signaling in magnocellular neurons of the male rat hypothalamic supraoptic nucleus (SON) induced by chronic salt loading. While without effect on GluA1-4 subunit mRNA, salt loading with 2% saline for 5-7 d resulted in a selective increase in AMPA receptor GluA1 protein expression in the SON, with no change in GluA2-4 protein expression, suggesting an increase in the ratio of GluA1 to GluA2 subunits. Salt loading induced a corresponding increase in EPSCs in both oxytocin (OT) and vasopressin (VP) neurons, with properties characteristic of calcium-permeable AMPA receptor-mediated currents. Unexpectedly, the emergent AMPA synaptic currents were silenced by blocking protein synthesis and mammalian target of rapamycin (mTOR) activity in the slices, suggesting that the new glutamate synapses induced by salt loading require continuous dendritic protein synthesis for maintenance. These findings indicate that chronic salt loading leads to the induction of highly labile glutamate synapses in OT and VP neurons that are comprised of calcium-permeable homomeric GluA1 AMPA receptors. The glutamate-induced calcium influx via calcium-permeable AMPA receptors would be expected to play a key role in the induction and/or maintenance of activity-dependent synaptic plasticity that occurs in the magnocellular neurons during chronic osmotic stimulation.
Collapse
Affiliation(s)
- Shi Di
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118
| | - ZhiYing Jiang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118
| | - Sen Wang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118
| | - Laura M Harrison
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118
| | | | - Thomas C Stuart
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118
| | - Marina E Wolf
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064
| | - Jeffrey G Tasker
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118
- Tulane Brain Institute, Tulane University, New Orleans, LA 70118
| |
Collapse
|
32
|
Ethanol Experience Enhances Glutamatergic Ventral Hippocampal Inputs to D1 Receptor-Expressing Medium Spiny Neurons in the Nucleus Accumbens Shell. J Neurosci 2019; 39:2459-2469. [PMID: 30692226 DOI: 10.1523/jneurosci.3051-18.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 01/11/2019] [Accepted: 01/15/2019] [Indexed: 02/05/2023] Open
Abstract
A growing number of studies implicate alterations in glutamatergic signaling within the reward circuitry of the brain during alcohol abuse and dependence. A key integrator of glutamatergic signaling in the reward circuit is the nucleus accumbens, more specifically, the dopamine D1 receptor-expressing medium spiny neurons (D1-MSNs) within this region, which have been implicated in the formation of dependence to many drugs of abuse including alcohol. D1-MSNs receive glutamatergic input from several brain regions; however, it is not currently known how individual inputs onto D1-MSNs are altered by alcohol experience. Here, we investigate input-specific adaptations in glutamatergic transmission in response to varying levels of alcohol experience. Virally mediated expression of Channelrhodopsin in ventral hippocampal (vHipp) glutamate neurons of male mice allowed for selective activation of vHipp to D1-MSN synapses. Therefore, we were able to compare synaptic adaptations in response to low and high alcohol experience in vitro and in vivo Alcohol experience enhanced glutamatergic activity and abolished LTD at vHipp to D1-MSN synapses. Following chronic alcohol experience, GluA2-lacking AMPARs, which are Ca permeable, were inserted into vHipp to D1-MSN synapses. These findings support the reversal of alcohol-induced insertion of Ca-permeable AMPARs and the enhancement of glutamatergic activity at vHipp to D1-MSNs as potential targets for intervention during early exposure to alcohol.SIGNIFICANCE STATEMENT Given the roles of the nucleus accumbens (NAc) in integrating cortical and allocortical information and in reward learning, it is vital to understand how inputs to this region are altered by drugs of abuse such as alcohol. The strength of excitatory inputs from the ventral hippocampus (vHipp) to the NAc has been positively associated with reward-related behaviors, but it is unclear whether or how ethanol affects these inputs. Here we show that vHipp-NAc synapses indeed are altered by ethanol exposure, with vHipp glutamatergic input to the NAc being enhanced following chronic ethanol experience. This work provides insight into ethanol-induced alterations of vHipp-NAc synapses and suggests that, similarly to drugs such as cocaine, the strengthening of these synapses promotes reward behavior.
Collapse
|
33
|
Turner BD, Kashima DT, Manz KM, Grueter CA, Grueter BA. Synaptic Plasticity in the Nucleus Accumbens: Lessons Learned from Experience. ACS Chem Neurosci 2018; 9:2114-2126. [PMID: 29280617 DOI: 10.1021/acschemneuro.7b00420] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Synaptic plasticity contributes to behavioral adaptations. As a key node in the reward pathway, the nucleus accumbens (NAc) is important for determining motivation-to-action outcomes. Across animal models of motivation including addiction, depression, anxiety, and hedonic feeding, selective recruitment of neuromodulatory signals and plasticity mechanisms have been a focus of physiologists and behaviorists alike. Experience-dependent plasticity mechanisms within the NAc vary depending on the distinct afferents and cell-types over time. A greater understanding of molecular mechanisms determining how these changes in synaptic strength track with behavioral adaptations will provide insight into the process of learning and memory along with identifying maladaptations underlying pathological behavior. Here, we summarize recent findings detailing how changes in NAc synaptic strength and mechanisms of plasticity manifest in various models of motivational disorders.
Collapse
Affiliation(s)
- Brandon D. Turner
- Vanderbilt Brain Institute, Nashville, Tennessee 37232, United States
| | - Daniel T. Kashima
- Vanderbilt Brain Institute, Nashville, Tennessee 37232, United States
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Kevin M. Manz
- Vanderbilt Brain Institute, Nashville, Tennessee 37232, United States
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Carrie A. Grueter
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Brad A. Grueter
- Vanderbilt Brain Institute, Nashville, Tennessee 37232, United States
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
34
|
Huang SH, Wu WR, Lee LM, Huang PR, Chen JC. mTOR signaling in the nucleus accumbens mediates behavioral sensitization to methamphetamine. Prog Neuropsychopharmacol Biol Psychiatry 2018; 86:331-339. [PMID: 29574227 DOI: 10.1016/j.pnpbp.2018.03.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 03/16/2018] [Accepted: 03/20/2018] [Indexed: 01/01/2023]
Abstract
Chronic psychostimulant treatment in rodents readily produces behavioral sensitization, which reflects altered brain function in response to repeated drug exposure. Numerous morphological and biochemical investigations implicate altered neural plasticity in striatal medium spiny neurons (MSNs) as an essential component in behavioral sensitization. The mammalian target of the rapamycin (mTOR) signaling pathway, a key regulator of synaptic neuroplasticity, in the ventral striatum of methamphetamine (METH) -sensitized mice was investigated to determine if a link exists with the development of METH sensitization. Behaviorally, METH-sensitized mice possessed increased levels of phosphorylated mTOR/S2448 and its down-stream regulator p70S6K and pS6 in the ventral striatum. Systemic treatment with rapamycin, a specific mTOR inhibitor, coincident with a daily METH injection suppressed the induction of METH sensitization and reduced the number of dendritic spines in the shell and core of the nucleus accumbens. The infusion of lentivirus-expressing mTOR-shRNA into the shell region of the nucleus accumbens inhibited the induction of behavioral sensitization to METH, which was comparable to the effect of rapamycin. These results suggest that mTORC1-mediated signaling in the nucleus accumbens mediates the development of behavioral sensitization to METH.
Collapse
Affiliation(s)
- Shin-Han Huang
- Department of Physiology and Pharmacology, Graduate Institute of Biomedical Sciences, School of Medicine, Chang-Gung University, Taiwan
| | - Wan-Rong Wu
- Department of Physiology and Pharmacology, Graduate Institute of Biomedical Sciences, School of Medicine, Chang-Gung University, Taiwan
| | - Li-Ming Lee
- Department of Biomedical Sciences, School of Medicine, Chang-Gung University, Taiwan
| | - Pei-Rong Huang
- Center for Molecular and Clinical Immunology, Chang-Gung University, Taiwan
| | - Jin-Chung Chen
- Department of Physiology and Pharmacology, Graduate Institute of Biomedical Sciences, School of Medicine, Chang-Gung University, Taiwan; Healthy Aging Research Center, Chang-Gung University, Taiwan; Neuroscience Research Center, Chang-Gung Memorial Hospital, Linkou, Taiwan; Chang-Gung Memorial Hospital, Keelung, Taiwan.
| |
Collapse
|
35
|
Sgritta M. Control of Cocaine Relapse: Lost in Translation. Biol Psychiatry 2018; 84:e25-e26. [PMID: 29941146 DOI: 10.1016/j.biopsych.2018.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 11/19/2022]
Affiliation(s)
- Martina Sgritta
- Department of Neuroscience and the Memory and Brain Research Center, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
36
|
Stefanik MT, Milovanovic M, Werner CT, Spainhour JCG, Wolf ME. Withdrawal From Cocaine Self-administration Alters the Regulation of Protein Translation in the Nucleus Accumbens. Biol Psychiatry 2018; 84:223-232. [PMID: 29622268 PMCID: PMC6054574 DOI: 10.1016/j.biopsych.2018.02.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/26/2018] [Accepted: 02/12/2018] [Indexed: 10/18/2022]
Abstract
BACKGROUND Cue-induced cocaine craving incubates during abstinence from cocaine self-administration. Expression of incubation ultimately depends on elevation of homomeric GluA1 alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors in the nucleus accumbens (NAc). This adaptation requires ongoing protein translation for its maintenance. Aberrant translation is implicated in central nervous system diseases, but nothing is known about glutamatergic regulation of translation in the drug-naïve NAc or after incubation. METHODS NAc tissue was obtained from drug-naïve rats and from rats after 1 or >40 days of abstinence from extended-access cocaine or saline self-administration. Newly translated proteins were labeled using 35S-Met/Cys or puromycin. We compared basal overall translation and its regulation by metabotropic glutamate receptor 1 (mGlu1), mGlu5, and N-methyl-D-aspartate receptors (NMDARs) in drug-naïve, saline control, and cocaine rats, and we compared GluA1 and GluA2 translation by immunoprecipitating puromycin-labeled proteins. RESULTS In all groups, overall translation was unaltered by mGlu1 blockade (LY367385) but increased by mGlu5 blockade (MTEP). NMDAR blockade (AVP) increased overall translation in drug-naïve and saline control rats but not in cocaine/late withdrawal rats. Cocaine/late withdrawal rats exhibited greater translation of GluA1 (but not GluA2), which was not further affected by NMDAR blockade. CONCLUSIONS Our results suggest that increased GluA1 translation contributes to the elevated homomeric GluA1 alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor levels in the NAc that mediate incubation. Additional contributions to incubation-related plasticity may result from loss of the braking influence on translation normally exerted by NMDARs. Apart from elucidating incubation-related adaptations, we found a suppressive effect of mGlu5 on NAc translation regardless of drug exposure, which is opposite to results obtained in the hippocampus and points to heterogeneity of translational regulation between brain regions.
Collapse
Affiliation(s)
- Michael T Stefanik
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin School of Medicine and Science, North Chicago, Illinois
| | - Mike Milovanovic
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin School of Medicine and Science, North Chicago, Illinois
| | - Craig T Werner
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin School of Medicine and Science, North Chicago, Illinois
| | - John C G Spainhour
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech College of Engineering and Emory School of Medicine, Atlanta, Georgia
| | - Marina E Wolf
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin School of Medicine and Science, North Chicago, Illinois.
| |
Collapse
|
37
|
Stefanik MT, Sakas C, Lee D, Wolf ME. Ionotropic and metabotropic glutamate receptors regulate protein translation in co-cultured nucleus accumbens and prefrontal cortex neurons. Neuropharmacology 2018; 140:62-75. [PMID: 30077883 DOI: 10.1016/j.neuropharm.2018.05.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 05/15/2018] [Accepted: 05/29/2018] [Indexed: 01/14/2023]
Abstract
The regulation of protein translation by glutamate receptors and its role in plasticity have been extensively studied in the hippocampus. In contrast, very little is known about glutamatergic regulation of translation in nucleus accumbens (NAc) medium spiny neurons (MSN), despite their critical role in addiction-related plasticity and recent evidence that protein translation contributes to this plasticity. We used a co-culture system, containing NAc MSNs and prefrontal cortex (PFC) neurons, and fluorescent non-canonical amino acid tagging (FUNCAT) to visualize newly synthesized proteins in neuronal processes of NAc MSNs and PFC pyramidal neurons. First, we verified that the FUNCAT signal reflects new protein translation. Next, we examined the regulation of translation by group I metabotropic glutamate receptors (mGluRs) and ionotropic glutamate receptors by incubating co-cultures with agonists or antagonists during the 2-h period of non-canonical amino acid labeling. In NAc MSNs, basal translation was modestly reduced by blocking Ca2+-permeable AMPARs whereas blocking all AMPARs or suppressing constitutive mGluR5 signaling enhanced translation. Activating group I mGluRs with dihydroxyphenylglycine increased translation in an mGluR1-dependent manner in NAc MSNs and PFC pyramidal neurons. Disinhibiting excitatory transmission with bicuculline also increased translation. In MSNs, this was reversed by antagonists of mGluR1, mGluR5, AMPARs or NMDARs. In PFC neurons, AMPAR or NMDAR antagonists blocked bicuculline-stimulated translation. Our study, the first to examine glutamatergic regulation of translation in MSNs, demonstrates regulatory mechanisms specific to MSNs that depend on the level of neuronal activation. This sets the stage for understanding how translation may be altered in addiction.
Collapse
Affiliation(s)
- Michael T Stefanik
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
| | - Courtney Sakas
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
| | - Dennis Lee
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
| | - Marina E Wolf
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA.
| |
Collapse
|
38
|
Smith ACW, Kenny PJ. MicroRNAs regulate synaptic plasticity underlying drug addiction. GENES, BRAIN, AND BEHAVIOR 2018; 17:e12424. [PMID: 28873276 PMCID: PMC5837931 DOI: 10.1111/gbb.12424] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/11/2017] [Accepted: 09/01/2017] [Indexed: 12/22/2022]
Abstract
Chronic use of drugs of abuse results in neurochemical, morphological and behavioral plasticity that underlies the emergence of compulsive drug seeking and vulnerability to relapse during periods of attempted abstinence. Identifying and reversing addiction-relevant plasticity is seen as a potential point of pharmacotherapeutic intervention in drug-addicted individuals. Despite considerable advances in our understanding of the actions of drugs of abuse in the brain, this information has thus far yielded few novel treatment options addicted individuals. MicroRNAs are small noncoding RNAs that can each regulate the translation of hundreds to thousands of messenger RNAs. The highly pleiotropic nature of miRNAs has focused attention on their contribution to addiction-relevant structural and functional plasticity in the brain and their potential utility as targets for medications development. In this review, we discuss the roles of miRNAs in synaptic plasticity underlying the development of addiction and then briefly discuss the possibility of using circulating miRNA as biomarkers for addiction.
Collapse
Affiliation(s)
- A. C. W. Smith
- The Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - P. J. Kenny
- The Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
39
|
Protein Translation in the Nucleus Accumbens Is Dysregulated during Cocaine Withdrawal and Required for Expression of Incubation of Cocaine Craving. J Neurosci 2018; 38:2683-2697. [PMID: 29431650 DOI: 10.1523/jneurosci.2412-17.2018] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 01/08/2018] [Accepted: 01/31/2018] [Indexed: 01/11/2023] Open
Abstract
Exposure to drug-associated cues can induce drug craving and relapse in abstinent addicts. Cue-induced craving that progressively intensifies ("incubates") during withdrawal from cocaine has been observed in both rats and humans. Building on recent evidence that aberrant protein translation underlies incubation-related adaptations in the NAc, we used male rats to test the hypothesis that translation is dysregulated during cocaine withdrawal and/or when rats express incubated cocaine craving. We found that intra-NAc infusion of anisomycin, a general protein translation inhibitor, or rapamycin, an inhibitor of mammalian target of rapamycin, reduced the expression of incubated cocaine craving, consistent with previous results showing that inhibition of translation in slices normalized the adaptations that maintain incubation. We then examined signaling pathways involved in protein translation using NAc synaptoneurosomes prepared after >47 d of withdrawal from cocaine or saline self-administration, or after withdrawal plus a cue-induced seeking test. The most robust changes were observed following seeking tests. Most notably, we found that eukaryotic elongation factor 2 (eEF2) and eukaryotic initiation factor 2α (eIF2α) are dephosphorylated when cocaine rats undergo a cue-induced seeking test; both effects are consistent with increased translation during the test. Blocking eIF2α dephosphorylation and thereby restoring its inhibitory influence on translation, via intra-NAc injection of Sal003 just before the test, substantially reduced cocaine seeking. These results are consistent with dysregulation of protein translation in the NAc during cocaine withdrawal, enabling cocaine cues to elicit an aberrant increase in translation that is required for the expression of incubated cocaine craving.SIGNIFICANCE STATEMENT Cue-induced cocaine craving progressively intensifies (incubates) during withdrawal in both humans and rats. This may contribute to persistent vulnerability to relapse. We previously demonstrated a role for protein translation in synaptic adaptations in the NAc closely linked to incubation. Here, we tested the hypothesis that translation is dysregulated during cocaine withdrawal, and this contributes to incubated craving. Analysis of signaling pathways regulating translation suggested that translation is enhanced when "incubated" rats undergo a cue-induced seeking test. Furthermore, intra-NAc infusions of drugs that inhibit protein translation through different mechanisms reduced expression of incubated cue-induced cocaine seeking. These results demonstrate that the expression of incubation depends on an acute increase in translation that may result from dysregulation of several pathways.
Collapse
|
40
|
Dos Santos M, Salery M, Forget B, Garcia Perez MA, Betuing S, Boudier T, Vanhoutte P, Caboche J, Heck N. Rapid Synaptogenesis in the Nucleus Accumbens Is Induced by a Single Cocaine Administration and Stabilized by Mitogen-Activated Protein Kinase Interacting Kinase-1 Activity. Biol Psychiatry 2017; 82:806-818. [PMID: 28545678 DOI: 10.1016/j.biopsych.2017.03.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 03/10/2017] [Accepted: 03/14/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Repeated cocaine exposure produces new spine formation in striatal projection neurons (SPNs) of the nucleus accumbens. However, an acute exposure to cocaine can trigger long-lasting synaptic plasticity in SPNs leading to behavioral alterations. This raises the intriguing question as to whether a single administration of cocaine could enduringly modify striatal connectivity. METHODS A three-dimensional morphometric analysis of presynaptic glutamatergic boutons and dendritic spines was performed on SPNs 1 hour and 1 week after a single cocaine administration. Time-lapse two-photon microscopy in adult slices was used to determine the precise molecular-events sequence responsible for the rapid spine formation. RESULTS A single injection triggered a rapid synaptogenesis and persistent increase in glutamatergic connectivity in SPNs from the shell part of the nucleus accumbens, specifically. Synapse formation occurred through clustered growth of active spines contacting pre-existing axonal boutons. Spine growth required extracellular signal-regulated kinase activation, while spine stabilization involved transcription-independent protein synthesis driven by mitogen-activated protein kinase interacting kinase-1, downstream from extracellular signal-regulated kinase. The maintenance of new spines driven by mitogen-activated protein kinase interacting kinase-1 was essential for long-term connectivity changes induced by cocaine in vivo. CONCLUSIONS Our study originally demonstrates that a single administration of cocaine is able to induce stable synaptic rewiring in the nucleus accumbens, which will likely influence responses to subsequent drug exposure. It also unravels a new functional role for cocaine-induced extracellular signal-regulated kinase pathway independently of nuclear targets. Finally, it reveals that mitogen-activated protein kinase interacting kinase-1 has a pivotal role in cocaine-induced connectivity.
Collapse
Affiliation(s)
- Marc Dos Santos
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Marine Salery
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Benoit Forget
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Maria Alexandra Garcia Perez
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Sandrine Betuing
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Thomas Boudier
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France; Bioinformatics Institute, Agency for Science, Technology, and Research, Singapore
| | - Peter Vanhoutte
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Jocelyne Caboche
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France.
| | - Nicolas Heck
- Neurosciences Paris Seine, Institut de Biologie Paris Seine, University Pierre and Marie Curie University of Paris 06, Sorbonne Universités, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Paris, France
| |
Collapse
|
41
|
Circuit and Synaptic Plasticity Mechanisms of Drug Relapse. J Neurosci 2017; 37:10867-10876. [PMID: 29118216 DOI: 10.1523/jneurosci.1821-17.2017] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 09/11/2017] [Accepted: 09/12/2017] [Indexed: 01/09/2023] Open
Abstract
High rates of relapse to drug use during abstinence is a defining feature of human drug addiction. This clinical scenario has been studied at the preclinical level using different animal models in which relapse to drug seeking is assessed after cessation of operant drug self-administration in rodents and monkeys. In our Society for Neuroscience (SFN) session entitled "Circuit and Synaptic Plasticity Mechanisms of Drug Relapse," we will discuss new developments of our understanding of circuits and synaptic plasticity mechanisms of drug relapse from studies combining established and novel animal models with state-of-the-art cellular, electrophysiology, anatomical, chemogenetic, and optogenetic methods. We will also discuss the translational implications of these new developments. In the mini-review that introduces our SFN session, we summarize results from our laboratories on behavioral, cellular, and circuit mechanisms of drug relapse within the context of our session.
Collapse
|
42
|
Zhu F, Wu Q, Li J, Grycel K, Liu B, Sun X, Zhou L, Jiao R, Song R, Khan YM, Wang Q, Wang L, Xu Y, Li J, Zhang B, Zhou Z. A single dose of cocaine potentiates glutamatergic synaptic transmission onto locus coeruleus neurons. Cell Calcium 2017; 67:11-20. [PMID: 29029785 DOI: 10.1016/j.ceca.2017.07.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/11/2017] [Accepted: 07/24/2017] [Indexed: 11/15/2022]
Abstract
The brainstem locus coeruleus (LC), the primary norepinephrinergic (NE) nucleus in the brain, has been implicated in the abuse of drugs such as opioids. However, whether and how the LC-NE system is involved in cocaine addiction remains elusive. Here, we demonstrated cocaine-evoked synaptic plasticity of glutamatergic transmission onto LC neurons as one of the earliest traces occurring after a single injection of cocaine. Twenty-four hours after mice were injected intraperitoneally with cocaine, the evoked α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) mediated synaptic transmission onto LC neurons were strongly potentiated without major effect on N-methyl-d-aspartate receptor (NMDAR) mediated synaptic transmission. Compared with saline-pretreated mice, AMPAR-mediated excitatory postsynaptic currents (EPSCs) of cocaine-pretreated mice showed a marked inward rectification, demonstrating the insertion of GluR2-lacking AMPARs to plasma membrane. In addition, the single injection of cocaine did not affect presynaptic glutamate release probability measured by paired pulse ratio. Furthermore, we found that the cocaine-induced potentiation of AMPAR EPSCs could be blocked by prazosin, an inhibitor of α1-adrenoreceptor (AR), indicating that cocaine increases AMPAR transmission via α1-ARs. These results reveal that LC-NE serves as an initial target of drug intake.
Collapse
Affiliation(s)
- Feipeng Zhu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China.
| | - Qihui Wu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and 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 and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Katarzyna Grycel
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Bing Liu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Xiaoxuan Sun
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Li Zhou
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Ruiyin Jiao
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Rui Song
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Younus M Khan
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, 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 and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Lun Wang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Yongxin Xu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Jin Li
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Bo Zhang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Zhuan Zhou
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China.
| |
Collapse
|
43
|
Kalinowska M, Francesconi A. Group I Metabotropic Glutamate Receptor Interacting Proteins: Fine-Tuning Receptor Functions in Health and Disease. Curr Neuropharmacol 2017; 14:494-503. [PMID: 27296642 PMCID: PMC4983749 DOI: 10.2174/1570159x13666150515234434] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 04/24/2015] [Accepted: 05/12/2015] [Indexed: 11/22/2022] Open
Abstract
Group I metabotropic glutamate receptors mediate slow excitatory neurotransmission in the central nervous system and are critical to activity-dependent synaptic plasticity, a cellular substrate of learning and memory. Dysregulated receptor signaling is implicated in neuropsychiatric conditions ranging from neurodevelopmental to neurodegenerative disorders. Importantly, group I metabotropic glutamate receptor signaling functions can be modulated by interacting proteins that mediate receptor trafficking, expression and coupling efficiency to signaling effectors. These interactions afford cell- or pathway-specific modulation to fine-tune receptor function, thus representing a potential target for pharmacological interventions in pathological conditions.
Collapse
Affiliation(s)
| | - Anna Francesconi
- Dominick P. Purpura Department of Neuroscience, Rose F. Kennedy Center, Room 706, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| |
Collapse
|
44
|
Eating 'Junk-Food' Produces Rapid and Long-Lasting Increases in NAc CP-AMPA Receptors: Implications for Enhanced Cue-Induced Motivation and Food Addiction. Neuropsychopharmacology 2016; 41:2977-2986. [PMID: 27383008 PMCID: PMC5101548 DOI: 10.1038/npp.2016.111] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 06/14/2016] [Accepted: 06/16/2016] [Indexed: 12/13/2022]
Abstract
Urges to eat are influenced by stimuli in the environment that are associated with food (food cues). Obese people are more sensitive to food cues, reporting stronger craving and consuming larger portions after food cue exposure. The nucleus accumbens (NAc) mediates cue-triggered motivational responses, and activations in the NAc triggered by food cues are stronger in people who are susceptible to obesity. This has led to the idea that alterations in NAc function similar to those underlying drug addiction may contribute to obesity, particularly in obesity-susceptible individuals. Motivational responses are mediated in part by NAc AMPA receptor (AMPAR) transmission, and recent work shows that cue-triggered motivation is enhanced in obesity-susceptible rats after 'junk-food' diet consumption. Therefore, here we determined whether NAc AMPAR expression and function is increased by 'junk-food' diet consumption in obesity-susceptible vs -resistant populations using both outbred and selectively bred models of susceptibility. In addition, cocaine-induced locomotor activity was used as a general 'read out' of mesolimbic function after 'junk-food' consumption. We found a sensitized locomotor response to cocaine in rats that gained weight on a 'junk-food' diet, consistent with greater responsivity of mesolimbic circuits in obesity-susceptible groups. In addition, eating 'junk-food' increased NAc calcium-permeable-AMPAR (CP-AMPAR) function only in obesity-susceptible rats. This increase occurred rapidly, persisted for weeks after 'junk-food' consumption ceased, and preceded the development of obesity. These data are considered in light of enhanced cue-triggered motivation and striatal function in obesity-susceptible rats and the role of NAc CP-AMPARs in enhanced motivation and addiction.
Collapse
|
45
|
Pick JE, Khatri L, Sathler MF, Ziff EB. mGluR long-term depression regulates GluA2 association with COPII vesicles and exit from the endoplasmic reticulum. EMBO J 2016; 36:232-244. [PMID: 27856517 DOI: 10.15252/embj.201694526] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 10/09/2016] [Accepted: 10/11/2016] [Indexed: 01/05/2023] Open
Abstract
mGluR long-term depression (mGluR-LTD) is a form of synaptic plasticity induced at excitatory synapses by metabotropic glutamate receptors (mGluRs). mGluR-LTD reduces synaptic strength and is relevant to learning and memory, autism, and sensitization to cocaine; however, the mechanism is not known. Here we show that activation of Group I mGluRs in medium spiny neurons induces trafficking of GluA2 from the endoplasmic reticulum (ER) to the synapse by enhancing GluA2 binding to essential COPII vesicle proteins, Sec23 and Sec13. GluA2 exit from the ER further depends on IP3 and Ryanodine receptor-controlled Ca2+ release as well as active translation. Synaptic insertion of GluA2 is coupled to removal of high-conducting Ca2+-permeable AMPA receptors from synapses, resulting in synaptic depression. This work demonstrates a novel mechanism in which mGluR signals release AMPA receptors rapidly from the ER and couple ER release to GluA2 synaptic insertion and GluA1 removal.
Collapse
Affiliation(s)
- Joseph E Pick
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Medical Center, New York, NY, USA
| | - Latika Khatri
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Medical Center, New York, NY, USA
| | - Matheus F Sathler
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Medical Center, New York, NY, USA.,Department of Pharmacology and Physiology, Fluminense Federal University, Niteroi, Brazil
| | - Edward B Ziff
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Medical Center, New York, NY, USA
| |
Collapse
|
46
|
Scheyer AF, Loweth JA, Christian DT, Uejima J, Rabei R, Le T, Dolubizno H, Stefanik MT, Murray CH, Sakas C, Wolf ME. AMPA Receptor Plasticity in Accumbens Core Contributes to Incubation of Methamphetamine Craving. Biol Psychiatry 2016; 80:661-670. [PMID: 27264310 PMCID: PMC5050076 DOI: 10.1016/j.biopsych.2016.04.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 04/01/2016] [Accepted: 04/01/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND The incubation of cue-induced drug craving in rodents provides a model of persistent vulnerability to craving and relapse in human addicts. After prolonged withdrawal, incubated cocaine craving depends on strengthening of nucleus accumbens (NAc) core synapses through incorporation of Ca2+-permeable alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (CP-AMPARs). Through metabotropic glutamate receptor 1 (mGluR1)-mediated synaptic depression, mGluR1 positive allosteric modulators remove CP-AMPARs from these synapses and thereby reduce cocaine craving. This study aimed to determine if similar plasticity accompanies incubation of methamphetamine craving. METHODS Rats self-administered saline or methamphetamine under extended-access conditions. Cue-induced seeking tests demonstrated incubation of methamphetamine craving. After withdrawal periods ranging from 1 to >40 days, rats underwent one of the following procedures: 1) whole-cell patch clamp recordings to characterize AMPAR transmission, 2) intra-NAc core injection of the CP-AMPAR antagonist 1-naphthyl acetyl spermine followed by a seeking test, or 3) systemic administration of a mGluR1 positive allosteric modulator followed by a seeking test. RESULTS Incubation of methamphetamine craving was associated with CP-AMPAR accumulation in NAc core, and both effects were maximal after ~1 week of withdrawal. Expression of incubated craving was decreased by intra-NAc core 1-naphthyl acetyl spermine injection or systemic mGluR1 positive allosteric modulator administration. CONCLUSIONS These results are the first to demonstrate a role for the NAc in the incubation of methamphetamine craving and describe adaptations in synaptic transmission associated with this model. They establish that incubation of craving and associated CP-AMPAR plasticity occur much more rapidly during withdrawal from methamphetamine compared with cocaine. However, a common mGluR1-based therapeutic strategy may be helpful for recovering cocaine and methamphetamine addicts.
Collapse
|
47
|
Abstract
Addiction is a disease of altered behavior. Addicts use drugs compulsively and will continue to do so despite negative consequences. Even after prolonged periods of abstinence, addicts are at risk of relapse, particularly when cues evoke memories that are associated with drug use. Rodent models mimic many of the core components of addiction, from the initial drug reinforcement to cue-associated relapse and continued drug intake despite negative consequences. Rodent models have also enabled unprecedented mechanistic insight into addiction, revealing plasticity of glutamatergic synaptic transmission evoked by the strong activation of mesolimbic dopamine-a defining feature of all addictive drugs-as a neural substrate for these drug-adaptive behaviors. Cell type-specific optogenetic manipulations have allowed both identification of the relevant circuits and design of protocols to reverse drug-evoked plasticity and to establish links of causality with drug-adaptive behaviors. The emergence of a circuit model for addiction will open the door for novel therapies, such as deep brain stimulation.
Collapse
Affiliation(s)
- Christian Lüscher
- Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland; .,Clinic of Neurology, Department of Clinical Neurosciences, Geneva University Hospital, CH-1211 Geneva, Switzerland
| |
Collapse
|
48
|
Scofield MD, Heinsbroek JA, Gipson CD, Kupchik YM, Spencer S, Smith ACW, Roberts-Wolfe D, Kalivas PW. The Nucleus Accumbens: Mechanisms of Addiction across Drug Classes Reflect the Importance of Glutamate Homeostasis. Pharmacol Rev 2016; 68:816-71. [PMID: 27363441 PMCID: PMC4931870 DOI: 10.1124/pr.116.012484] [Citation(s) in RCA: 415] [Impact Index Per Article: 46.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The nucleus accumbens is a major input structure of the basal ganglia and integrates information from cortical and limbic structures to mediate goal-directed behaviors. Chronic exposure to several classes of drugs of abuse disrupts plasticity in this region, allowing drug-associated cues to engender a pathologic motivation for drug seeking. A number of alterations in glutamatergic transmission occur within the nucleus accumbens after withdrawal from chronic drug exposure. These drug-induced neuroadaptations serve as the molecular basis for relapse vulnerability. In this review, we focus on the role that glutamate signal transduction in the nucleus accumbens plays in addiction-related behaviors. First, we explore the nucleus accumbens, including the cell types and neuronal populations present as well as afferent and efferent connections. Next we discuss rodent models of addiction and assess the viability of these models for testing candidate pharmacotherapies for the prevention of relapse. Then we provide a review of the literature describing how synaptic plasticity in the accumbens is altered after exposure to drugs of abuse and withdrawal and also how pharmacological manipulation of glutamate systems in the accumbens can inhibit drug seeking in the laboratory setting. Finally, we examine results from clinical trials in which pharmacotherapies designed to manipulate glutamate systems have been effective in treating relapse in human patients. Further elucidation of how drugs of abuse alter glutamatergic plasticity within the accumbens will be necessary for the development of new therapeutics for the treatment of addiction across all classes of addictive substances.
Collapse
Affiliation(s)
- M D Scofield
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - J A Heinsbroek
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - C D Gipson
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - Y M Kupchik
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - S Spencer
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - A C W Smith
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - D Roberts-Wolfe
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - P W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| |
Collapse
|
49
|
Abstract
Although it is challenging for individuals with cocaine addiction to achieve abstinence, the greatest difficulty is avoiding relapse to drug taking, which is often triggered by cues associated with prior cocaine use. This vulnerability to relapse persists for long periods (months to years) after abstinence is achieved. Here, I discuss rodent studies of cue-induced cocaine craving during abstinence, with a focus on neuronal plasticity in the reward circuitry that maintains high levels of craving. Such work has the potential to identify new therapeutic targets and to further our understanding of experience-dependent plasticity in the adult brain under normal circumstances and in the context of addiction.
Collapse
Affiliation(s)
- Marina E Wolf
- The Chicago Medical School at Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, Illinois 60064, USA
| |
Collapse
|
50
|
Oginsky MF, Maust JD, Corthell JT, Ferrario CR. Enhanced cocaine-induced locomotor sensitization and intrinsic excitability of NAc medium spiny neurons in adult but not in adolescent rats susceptible to diet-induced obesity. Psychopharmacology (Berl) 2016; 233:773-84. [PMID: 26612617 PMCID: PMC4752900 DOI: 10.1007/s00213-015-4157-x] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 11/10/2015] [Indexed: 11/29/2022]
Abstract
RATIONALE Basal and diet-induced differences in mesolimbic function, particularly within the nucleus accumbens (NAc), may contribute to human obesity; these differences may be more pronounced in susceptible populations. OBJECTIVES We examined differences in cocaine-induced behavioral plasticity in rats that are susceptible vs. resistant to diet-induced obesity and basal differences in striatal neuron function in adult and in adolescent obesity-prone and obesity-resistant rats. METHODS Susceptible and resistant outbred rats were identified based on "junk-food" diet-induced obesity. Then, the induction and expression of cocaine-induced locomotor sensitization, which is mediated by enhanced striatal function and is associated with increased motivation for rewards and reward-paired cues, were evaluated. Basal differences in mesolimbic function were examined in selectively bred obesity-prone and obesity-resistant rats (P70-80 and P30-40) using both cocaine-induced locomotion and whole-cell patch clamping approaches in NAc core medium spiny neurons (MSNs). RESULTS In rats that became obese after eating junk-food, the expression of locomotor sensitization was enhanced compared to non-obese rats, with similarly strong responses to 7.5 and 15 mg/kg cocaine. Without diet manipulation, obesity-prone rats were hyper-responsive to the acute locomotor-activating effects of cocaine, and the intrinsic excitability of NAc core MSNs was enhanced by ∼60 % at positive and negative potentials. These differences were present in adult, but not adolescent rats. Post-synaptic glutamatergic transmission was similar between groups. CONCLUSIONS Mesolimbic systems, particularly NAc MSNs, are hyper-responsive in obesity-prone individuals, and interactions between predisposition and experience influence neurobehavioral plasticity in ways that may promote weight gain and hamper weight loss in susceptible rats.
Collapse
Affiliation(s)
- Max F Oginsky
- Department of Pharmacology, University of Michigan, 1150 W. Medical Center Drive, MSRB III 1301, Ann Arbor, MI, 48109, USA
| | - Joel D Maust
- Department of Pharmacology, University of Michigan, 1150 W. Medical Center Drive, MSRB III 1301, Ann Arbor, MI, 48109, USA
| | - John T Corthell
- Department of Pharmacology, University of Michigan, 1150 W. Medical Center Drive, MSRB III 1301, Ann Arbor, MI, 48109, USA
| | - Carrie R Ferrario
- Department of Pharmacology, University of Michigan, 1150 W. Medical Center Drive, MSRB III 1301, Ann Arbor, MI, 48109, USA.
| |
Collapse
|