1
|
Hu YB, Deng X, Liu L, Cao CC, Su YW, Gao ZJ, Cheng X, Kong D, Li Q, Shi YW, Wang XG, Ye X, Zhao H. Distinct roles of excitatory and inhibitory neurons in the medial prefrontal cortex in the expression and reconsolidation of methamphetamine-associated memory in male mice. Neuropsychopharmacology 2024; 49:1827-1838. [PMID: 38730034 PMCID: PMC11473735 DOI: 10.1038/s41386-024-01879-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024]
Abstract
Methamphetamine, a commonly abused drug, is known for its high relapse rate. The persistence of addictive memories associated with methamphetamine poses a significant challenge in preventing relapse. Memory retrieval and subsequent reconsolidation provide an opportunity to disrupt addictive memories. However, the key node in the brain network involved in methamphetamine-associated memory retrieval has not been clearly defined. In this study, using the conditioned place preference in male mice, whole brain c-FOS mapping and functional connectivity analysis, together with chemogenetic manipulations of neural circuits, we identified the medial prefrontal cortex (mPFC) as a critical hub that integrates inputs from the retrosplenial cortex and the ventral tegmental area to support both the expression and reconsolidation of methamphetamine-associated memory during its retrieval. Surprisingly, with further cell-type specific analysis and manipulation, we also observed that methamphetamine-associated memory retrieval activated inhibitory neurons in the mPFC to facilitate memory reconsolidation, while suppressing excitatory neurons to aid memory expression. These findings provide novel insights into the neural circuits and cellular mechanisms involved in the retrieval process of addictive memories. They suggest that targeting the balance between excitation and inhibition in the mPFC during memory retrieval could be a promising treatment strategy to prevent relapse in methamphetamine addiction.
Collapse
Affiliation(s)
- Yu-Bo Hu
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xi Deng
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Lu Liu
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Can-Can Cao
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Ya-Wen Su
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Zhen-Jie Gao
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xin Cheng
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Deshan Kong
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Qi Li
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yan-Wei Shi
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xiao-Guang Wang
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xiaojing Ye
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| | - Hu Zhao
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
2
|
Jun DJ, Shannon R, Tschida K, Smith DM. The Infralimbic, but not the Prelimbic Cortex is needed for a Complex Olfactory Memory Task. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618554. [PMID: 39463969 PMCID: PMC11507807 DOI: 10.1101/2024.10.15.618554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The medial prefrontal cortex (mPFC) plays a key role in memory and behavioral flexibility, and a growing body of evidence suggests that the prelimbic (PL) and infralimbic (IL) subregions contribute differently to these processes. Studies of fear conditioning and goal-directed learning suggest that the PL promotes behavioral responses and memory retrieval, while the IL inhibits them. Other studies have shown that the mPFC is engaged under conditions of high interference. This raises the possibility that the PL and IL play differing roles in resolving interference. To examine this, we first used chemogenetics (DREADDs) to suppress mPFC neuronal activity and tested subjects on a conditional discrimination task known to be sensitive to muscimol inactivation. After confirming the effectiveness of the DREADD procedures, we conducted a second experiment to examine the PL and IL roles in a high interference memory task. We trained rats on two consecutive sets of conflicting odor discrimination problems, A and B, followed by test sessions involving a mid-session switch between the problem sets. Controls repeatedly performed worse on Set A, suggesting that learning Set B inhibited the rats' ability to retrieve Set A memories (i.e. retroactive interference). PL inactivation rats performed similarly to controls. However, IL inactivation rats did not show this effect, suggesting that the IL plays a critical role in suppressing the retrieval of previously acquired memories that may interfere with retrieval of more recent memories. These results suggest that the IL plays a critical role in memory control processes needed for resolving interference.
Collapse
Affiliation(s)
- Dahae J. Jun
- Department of Psychology, Cornell University, 211 Uris Hall Ithaca, NY 14853 United States
| | - Rebecca Shannon
- Department of Psychology, Cornell University, 211 Uris Hall Ithaca, NY 14853 United States
| | - Katherine Tschida
- Department of Psychology, Cornell University, 211 Uris Hall Ithaca, NY 14853 United States
| | - David M. Smith
- Department of Psychology, Cornell University, 211 Uris Hall Ithaca, NY 14853 United States
| |
Collapse
|
3
|
Liu S, Nawarawong N, Liu X, Liu QS, Olsen CM. Dissociable dorsal medial prefrontal cortex ensembles are necessary for cocaine seeking and fear conditioning in mice. Transl Psychiatry 2024; 14:387. [PMID: 39313502 PMCID: PMC11420216 DOI: 10.1038/s41398-024-03068-7] [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: 02/01/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/25/2024] Open
Abstract
The dorsal medial prefrontal cortex (dmPFC) plays a dual role in modulating drug seeking and fear-related behaviors. Learned associations between cues and drug seeking are encoded by a specific ensemble of neurons. This study explored the stability of a dmPFC cocaine seeking ensemble over 2 weeks and its influence on persistent cocaine seeking and fear memory retrieval. In the first series of experiments, we trained TetTag c-fos-driven-EGFP mice in cocaine self-administration and tagged strongly activated neurons with EGFP during the initial day 7 cocaine seeking session. Subsequently, a follow-up seeking test was conducted 2 weeks later to examine ensemble reactivation between two seeking sessions via c-Fos immunostaining. In the second series of experiments, we co-injected viruses expressing TRE-cre and a cre-dependent inhibitory PSAM-GlyR into the dmPFC of male and female c-fos-tTA mice to enable "tagging" of cocaine seeking ensemble or cued fear ensemble neurons with inhibitory chemogenetic receptors. These c-fos-tTA mice have the c-fos promoter that drives expression of the tetracycline transactivator (tTA). The tTA can bind to the tetracycline response element (TRE) site on the viral construct, resulting in the expression of cre-recombinase, which enables the expression of cre-dependent inhibitory chemogenetic receptors and fluorescent reporters. Then we investigated ensemble contribution to subsequent cocaine seeking and fear recall during inhibition of the tagged ensemble by administering uPSEM792s (0.3 mg/kg), a selective ligand for PSAM-GlyR. In both sexes, there was a positive association between the persistence of cocaine seeking and the proportion of reactivated EGFP+ neurons within the dmPFC. More importantly, inhibition of the cocaine seeking ensemble suppressed cocaine seeking, but not recall of fear memory, while inhibition of the fear ensemble reduced conditioned freezing but not cocaine seeking. The results demonstrate that cocaine and fear recall ensembles in the dmPFC are stable, but largely exclusive from one another.
Collapse
Affiliation(s)
- Shuai Liu
- Department of Pharmacology & Toxicology, Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Departments of Pharmacology & Toxicology and Neurosurgery, Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Natalie Nawarawong
- Department of Pharmacology & Toxicology, Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Xiaojie Liu
- Department of Pharmacology & Toxicology, Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Qing-Song Liu
- Department of Pharmacology & Toxicology, Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Christopher M Olsen
- Department of Pharmacology & Toxicology, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
4
|
Vlkolinsky R, Khom S, Vozella V, Bajo M, Roberto M. Withdrawal from chronic alcohol impairs the serotonin-mediated modulation of GABAergic transmission in the infralimbic cortex in male rats. Neurobiol Dis 2024; 199:106590. [PMID: 38996987 PMCID: PMC11412312 DOI: 10.1016/j.nbd.2024.106590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/14/2024] Open
Abstract
The infralimbic cortex (IL) is part of the medial prefrontal cortex (mPFC), exerting top-down control over structures that are critically involved in the development of alcohol use disorder (AUD). Activity of the IL is tightly controlled by γ-aminobutyric acid (GABA) transmission, which is susceptible to chronic alcohol exposure and withdrawal. This inhibitory control is regulated by various neuromodulators, including 5-hydroxytryptamine (5-HT; serotonin). We used chronic intermittent ethanol vapor inhalation exposure, a model of AUD, in male Sprague-Dawley rats to induce alcohol dependence (Dep) followed by protracted withdrawal (WD; 2 weeks) and performed ex vivo electrophysiology using whole-cell patch clamp to study GABAergic transmission in layer V of IL pyramidal neurons. We found that WD increased frequencies of spontaneous inhibitory postsynaptic currents (sIPSCs), whereas miniature IPSCs (mIPSCs; recorded in the presence of tetrodotoxin) were unaffected by either Dep or WD. The application of 5-HT (50 μM) increased sIPSC frequencies and amplitudes in naive and Dep rats but reduced sIPSC frequencies in WD rats. Additionally, 5-HT2A receptor antagonist M100907 and 5-HT2C receptor antagonist SB242084 reduced basal GABA release in all groups to a similar extent. The blockage of either 5-HT2A or 5-HT2C receptors in WD rats restored the impaired response to 5-HT, which then resembled responses in naive rats. Our findings expand our understanding of synaptic inhibition in the IL in AUD, indicating that antagonism of 5-HT2A and 5-HT2C receptors may restore GABAergic control over IL pyramidal neurons. SIGNIFICANCE STATEMENT: Impairment in the serotonergic modulation of GABAergic inhibition in the medial prefrontal cortex contributes to alcohol use disorder (AUD). We used a well-established rat model of AUD and ex vivo whole-cell patch-clamp electrophysiology to characterize the serotonin modulation of GABAergic transmission in layer V infralimbic (IL) pyramidal neurons in ethanol-naive, ethanol-dependent (Dep), and ethanol-withdrawn (WD) male rats. We found increased basal inhibition following WD from chronic alcohol and altered serotonin modulation. Exogenous serotonin enhanced GABAergic transmission in naive and Dep rats but reduced it in WD rats. 5-HT2A and 5-HT2C receptor blockage in WD rats restored the typical serotonin-mediated enhancement of GABAergic inhibition. Our findings expand our understanding of synaptic inhibition in the infralimbic neurons in AUD.
Collapse
Affiliation(s)
- Roman Vlkolinsky
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Sophia Khom
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Pharmaceutical Sciences, University of Vienna, Vienna, 1090, Austria.
| | - Valentina Vozella
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Michal Bajo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
5
|
Vierkant V, Xie X, Huang Z, He L, Bancroft E, Wang X, Nguyen T, Srinivasan R, Zhou Y, Wang J. Optogenetic inhibition of light-captured alcohol-taking striatal engrams facilitates extinction and suppresses reinstatement. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024. [PMID: 39095328 DOI: 10.1111/acer.15412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/09/2024] [Accepted: 07/16/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Alcohol use disorder (AUD) is a complex condition, and it remains unclear which specific neuronal substrates mediate alcohol-seeking and -taking behaviors. Engram cells and their related ensembles, which encode learning and memory, may play a role in this process. We aimed to assess the precise neural substrates underlying alcohol-seeking and -taking behaviors and determine how they may affect one another. METHODS Using FLiCRE (Fast Light and Calcium-Regulated Expression; a newly developed technique which permits the trapping of acutely activated neuronal ensembles) and operant self-administration (OSA), we tagged striatal neurons activated during alcohol-taking behaviors. We used FLiCRE to express an inhibitory halorhodopsin in alcohol-taking neurons, permitting loss-of-function manipulations. RESULTS We found that the inhibition of OSA-tagged alcohol-taking neurons decreased both alcohol-seeking and -taking behaviors in future OSA trials. In addition, optogenetic inhibition of these OSA-tagged alcohol-taking neurons during extinction training facilitated the extinction of alcohol-seeking behaviors. Furthermore, inhibition of these OSA-tagged alcohol-taking neurons suppressed the reinstatement of alcohol-seeking behaviors, but, interestingly, it did not significantly suppress alcohol-taking behaviors during reinstatement. CONCLUSIONS Our findings suggest that alcohol-taking neurons are crucial for future alcohol-seeking behaviors during extinction and reinstatement. These results may help in the development of new therapeutic approaches to enhance extinction and suppress relapse in individuals with AUD.
Collapse
Affiliation(s)
- Valerie Vierkant
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Xueyi Xie
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Zhenbo Huang
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Lian He
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas, USA
| | - Eric Bancroft
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Xuehua Wang
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Tran Nguyen
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Rahul Srinivasan
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Yubin Zhou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas, USA
| | - Jun Wang
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| |
Collapse
|
6
|
Giannone F, Ebrahimi C, Endrass T, Hansson AC, Schlagenhauf F, Sommer WH. Bad habits-good goals? Meta-analysis and translation of the habit construct to alcoholism. Transl Psychiatry 2024; 14:298. [PMID: 39030169 PMCID: PMC11271507 DOI: 10.1038/s41398-024-02965-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 05/19/2024] [Accepted: 05/24/2024] [Indexed: 07/21/2024] Open
Abstract
Excessive alcohol consumption remains a global public health crisis, with millions suffering from alcohol use disorder (AUD, or simply "alcoholism"), leading to significantly reduced life expectancy. This review examines the interplay between habitual and goal-directed behaviors and the associated neurobiological changes induced by chronic alcohol exposure. Contrary to a strict habit-goal dichotomy, our meta-analysis of the published animal experiments combined with a review of human studies reveals a nuanced transition between these behavioral control systems, emphasizing the need for refined terminology to capture the probabilistic nature of decision biases in individuals with a history of chronic alcohol exposure. Furthermore, we distinguish habitual responding from compulsivity, viewing them as separate entities with diverse roles throughout the stages of the addiction cycle. By addressing species-specific differences and translational challenges in habit research, we provide insights to enhance future investigations and inform strategies for combatting AUD.
Collapse
Affiliation(s)
- F Giannone
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159, Mannheim, Germany
| | - C Ebrahimi
- Faculty of Psychology, Institute of Clinical Psychology and Psychotherapy, Technische Universität Dresden, 01062, Dresden, Germany
| | - T Endrass
- Faculty of Psychology, Institute of Clinical Psychology and Psychotherapy, Technische Universität Dresden, 01062, Dresden, Germany
| | - A C Hansson
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159, Mannheim, Germany
| | - F Schlagenhauf
- Department of Psychotherapy, Campus Charité Mitte, Charité Universitätsmedizin Berlin & St. Hedwig Hospital, 10117, Berlin, Germany
| | - W H Sommer
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159, Mannheim, Germany.
- Bethania Hospital for Psychiatry, Psychosomatics and Psychotherapy, Greifswald, Germany.
- German Center for Mental Health (DZPG), Partner Site Mannheim-Heidelberg-Ulm, 68159, Mannheim, Germany.
| |
Collapse
|
7
|
Patel RR, Gandhi P, Spencer K, Salem NA, Erikson CM, Borgonetti V, Vlkolinsky R, Rodriguez L, Nadav T, Bajo M, Roberts AJ, Dayne Mayfield R, Roberto M. Functional and morphological adaptation of medial prefrontal corticotropin releasing factor receptor 1-expressing neurons in male mice following chronic ethanol exposure. Neurobiol Stress 2024; 31:100657. [PMID: 38983690 PMCID: PMC11231756 DOI: 10.1016/j.ynstr.2024.100657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/13/2024] [Accepted: 06/15/2024] [Indexed: 07/11/2024] Open
Abstract
Chronic ethanol dependence and withdrawal activate corticotropin releasing factor (CRF)-containing GABAergic neurons in the medial prefrontal cortex (mPFC), which tightly regulate glutamatergic pyramidal neurons. Using male CRF1:GFP reporter mice, we recently reported that CRF1-expressing (mPFCCRF1+) neurons predominantly comprise mPFC prelimbic layer 2/3 pyramidal neurons, undergo profound adaptations following chronic ethanol exposure, and regulate anxiety and conditioned rewarding effects of ethanol. To explore the effects of acute and chronic ethanol exposure on glutamate transmission, the impact of chronic alcohol on spine density and morphology, as well as persistent changes in dendritic-related gene expression, we employed whole-cell patch-clamp electrophysiology, diOlistic labeling for dendritic spine analysis, and dendritic gene expression analysis to further characterize mPFCCRF1+ and mPFCCRF1- prelimbic layer 2/3 pyramidal neurons. We found increased glutamate release in mPFCCRF1+ neurons with ethanol dependence, which recovered following withdrawal. In contrast, we did not observe significant changes in glutamate transmission in neighboring mPFCCRF1- neurons. Acute application of 44 mM ethanol significantly reduced glutamate release onto mPFCCRF1+ neurons, which was observed across all treatment groups. However, this sensitivity to acute ethanol was only evident in mPFCCRF1- neurons during withdrawal. In line with alterations in glutamate transmission, we observed a decrease in total spine density in mPFCCRF1+ neurons during dependence, which recovered following withdrawal, while again no changes were observed in mPFCCRF- neurons. Given the observed decreases in mPFCCRF1+ stubby spines during withdrawal, we then identified persistent changes at the dendritic gene expression level in mPFCCRF1+ neurons following withdrawal that may underlie these structural adaptations. Together, these findings highlight the varying responses of mPFCCRF1+ and mPFCCRF1- cell-types to acute and chronic ethanol exposure, as well as withdrawal, revealing specific functional, morphological, and molecular adaptations that may underlie vulnerability to ethanol and the lasting effects of ethanol dependence.
Collapse
Affiliation(s)
- Reesha R. Patel
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Pauravi Gandhi
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Kathryn Spencer
- Core Microscopy Facility, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Nihal A. Salem
- Department of Neuroscience, The University of Texas at Austin, Austin, TX, 78712, USA
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Chloe. M. Erikson
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Vittoria Borgonetti
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Roman Vlkolinsky
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Larry Rodriguez
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Tali Nadav
- Animal Models Core Facility, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Michal Bajo
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Amanda J. Roberts
- Animal Models Core Facility, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - R. Dayne Mayfield
- Department of Neuroscience, The University of Texas at Austin, Austin, TX, 78712, USA
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| |
Collapse
|
8
|
Vierkant V, Xie X, Huang Z, He L, Bancroft E, Wang X, Srinivisan R, Zhou Y, Wang J. Optogenetic inhibition of light-captured alcohol-taking striatal engrams facilitates extinction and suppresses reinstatement. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.02.597021. [PMID: 38853893 PMCID: PMC11160798 DOI: 10.1101/2024.06.02.597021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Background Alcohol use disorder (AUD) is a complex condition, and it remains unclear which specific neuronal substrates mediate alcohol-seeking and -taking behaviors. Engram cells and their related ensembles, which encode learning and memory, may play a role in this process. We aimed to assess the precise neural substrates underlying alcohol-seeking and -taking behaviors and determine how they may affect one another. Methods Using FLiCRE (Fast Light and Calcium-Regulated Expression; a newly developed technique which permits the trapping of acutely activated neuronal ensembles) and operant-self administration (OSA), we tagged striatal neurons activated during alcohol-taking behaviors. We used FLiCRE to express an inhibitory halorhodopsin in alcohol-taking neurons, permitting loss-of-function manipulations. Results We found that the inhibition of OSA-tagged alcohol-taking neurons decreased both alcohol-seeking and -taking behaviors in future OSA trials. In addition, optogenetic inhibition of these OSA-tagged alcohol-taking neurons during extinction training facilitated the extinction of alcohol-seeking behaviors. Furthermore, inhibition of these OSA-tagged alcohol-taking neurons suppressed the reinstatement of alcohol-seeking behaviors, but, interestingly, it did not significantly suppress alcohol-taking behaviors during reinstatement. Conclusions Our findings suggest that alcohol-taking neurons are crucial for future alcohol-seeking behaviors during extinction and reinstatement. These results may help in the development of new therapeutic approaches to enhance extinction and suppress relapse in individuals with AUD.
Collapse
|
9
|
Arnold ME, Schank JR. Sex differences in neuronal activation in the cortex and midbrain during quinine-adulterated alcohol intake. Alcohol Alcohol 2024; 59:agae031. [PMID: 38742547 PMCID: PMC11091839 DOI: 10.1093/alcalc/agae031] [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: 01/10/2024] [Revised: 04/10/2024] [Accepted: 04/23/2024] [Indexed: 05/16/2024] Open
Abstract
AIMS Continued alcohol consumption despite negative consequences is a core symptom of alcohol use disorder. This is modeled in mice by pairing negative stimuli with alcohol, such as adulterating alcohol solution with quinine. Mice consuming alcohol under these conditions are considered to be engaging in aversion-resistant intake. Previously, we have observed sex differences in this behavior, with females more readily expressing aversion-resistant consumption. We also identified three brain regions that exhibited sex differences in neuronal activation during quinine-alcohol drinking: ventromedial prefrontal cortex (vmPFC), posterior insular cortex (PIC), and ventral tegmental area (VTA). Specifically, male mice showed increased activation in vmPFC and PIC, while females exhibited increased activation in VTA. In this study, we aimed to identify what specific type of neurons are activated in these regions during quinine-alcohol drinking. METHOD We assessed quinine-adulterated alcohol intake using the two-bottle choice procedure. We also utilized RNAscope in situ hybridization in the three brain regions that previously exhibited a sex difference to examine colocalization of Fos, glutamate, GABA, and dopamine. RESULT Females showed increased aversion-resistant alcohol consumption compared to males. We also found that males had higher colocalization of glutamate and Fos in vmPFC and PIC, while females had greater dopamine and Fos colocalization in the VTA. CONCLUSIONS Collectively, these experiments suggest that glutamatergic output from the vmPFC and PIC may have a role in suppressing, and dopaminergic activity in the VTA may promote, aversion-resistant alcohol consumption. Future experiments will examine neuronal circuits that contribute to sex differences in aversion resistant consumption.
Collapse
Affiliation(s)
- Miranda E Arnold
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 D. W. Brooks Drive, Athens, GA 30602, United States
| | - Jesse R Schank
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 D. W. Brooks Drive, Athens, GA 30602, United States
| |
Collapse
|
10
|
Luft JG, Popik B, Gonçalves DA, Cruz FC, de Oliveira Alvares L. Distinct engrams control fear and extinction memory. Hippocampus 2024; 34:230-240. [PMID: 38396226 DOI: 10.1002/hipo.23601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/06/2023] [Accepted: 02/04/2024] [Indexed: 02/25/2024]
Abstract
Memories are stored in engram cells, which are necessary and sufficient for memory recall. Recalling a memory might undergo reconsolidation or extinction. It has been suggested that the original memory engram is reactivated during reconsolidation so that memory can be updated. Conversely, during extinction training, a new memory is formed that suppresses the original engram. Nonetheless, it is unknown whether extinction creates a new engram or modifies the original fear engram. In this study, we utilized the Daun02 procedure, which uses c-Fos-lacZ rats to induce apoptosis of strongly activated neurons and examine whether a new memory trace emerges as a result of a short or long reactivation, or if these processes rely on modifications within the original engram located in the basolateral amygdala (BLA) and infralimbic (IL) cortex. By eliminating neurons activated during consolidation and reactivation, we observed significant impacts on fear memory, highlighting the importance of the BLA engram in these processes. Although we were unable to show any impact when removing the neurons activated after the test of a previously extinguished memory in the BLA, disrupting the IL extinction engram reactivated the aversive memory that was suppressed by the extinction memory. Thus, we demonstrated that the IL cortex plays a crucial role in the network involved in extinction, and disrupting this specific node alone is sufficient to impair extinction behavior. Additionally, our findings indicate that extinction memories rely on the formation of a new memory, supporting the theory that extinction memories rely on the formation of a new memory, whereas the reconsolidation process reactivates the same original memory trace.
Collapse
Affiliation(s)
- Jordana Griebler Luft
- Laboratório de Neurobiologia da Memória, Departamento de Biofísica, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Bruno Popik
- Laboratório de Neurobiologia da Memória, Departamento de Biofísica, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Débora Aguirre Gonçalves
- Laboratório de Neurobiologia da Memória, Departamento de Biofísica, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fabio Cardoso Cruz
- Departamento de Farmacologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Lucas de Oliveira Alvares
- Laboratório de Neurobiologia da Memória, Departamento de Biofísica, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
11
|
Liu S, Nawarawong N, Liu X, Liu QS, Olsen CM. Dissociable dorsal medial prefrontal cortex ensembles are necessary for cocaine seeking and fear conditioning in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.17.585444. [PMID: 38562850 PMCID: PMC10983871 DOI: 10.1101/2024.03.17.585444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The dmPFC plays a dual role in modulating drug seeking and fear-related behaviors. Learned associations between cues and drug seeking are encoded by a specific ensemble of neurons. This study explored the stability of a dmPFC cocaine seeking ensemble over two weeks and its influence on persistent cocaine seeking and fear memory retrieval. In the first series of experiments, we trained TetTag mice in cocaine self-administration and tagged strongly activated neurons with EGFP during the initial day 7 cocaine seeking session. Subsequently, a follow-up seeking test was conducted two weeks later to examine ensemble reactivation between two seeking sessions via c-Fos immunostaining. In the second series of experiments, we co-injected viruses expressing TRE-cre and a cre-dependent inhibitory PSAM-GlyR into the dmPFC of male and female c-fos -tTA mice to enable "tagging" of cocaine seeking ensemble or cued fear ensemble neurons with an inhibitory chemogenetic receptors. Then we investigated their contribution to subsequent cocaine seeking and fear recall during inhibition of the tagged ensemble by administering uPSEM792s (0.3 mg/kg), a selective ligand for PSAM-GlyR. In both sexes, there was a positive association between the persistence of cocaine seeking and the proportion of reactivated EGFP+ neurons within the dmPFC. More importantly, inhibition of the cocaine seeking ensemble suppressed cocaine seeking, but not recall of fear memory, while inhibition of the fear ensemble reduced conditioned freezing but not cocaine seeking. The results demonstrate that cocaine and fear recall ensembles in the dmPFC are stable, but largely exclusive from one another.
Collapse
|
12
|
Tian W, Zhao D, Ding J, Zhan S, Zhang Y, Etkin A, Wu W, Yuan TF. An electroencephalographic signature predicts craving for methamphetamine. Cell Rep Med 2024; 5:101347. [PMID: 38151021 PMCID: PMC10829728 DOI: 10.1016/j.xcrm.2023.101347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/17/2023] [Accepted: 11/28/2023] [Indexed: 12/29/2023]
Abstract
Craving is central to methamphetamine use disorder (MUD) and both characterizes the disease and predicts relapse. However, there is currently a lack of robust and reliable biomarkers for monitoring craving and diagnosing MUD. Here, we seek to identify a neurobiological signature of craving based on individual-level functional connectivity pattern differences between healthy control and MUD subjects. We train high-density electroencephalography (EEG)-based models using data recorded during the resting state and then calculate imaginary coherence features between the band-limited time series across different brain regions of interest. Our prediction model demonstrates that eyes-open beta functional connectivity networks have significant predictive value for craving at the individual level and can also identify individuals with MUD. These findings advance the neurobiological understanding of craving through an EEG-tailored computational model of the brain connectome. Dissecting neurophysiological features provides a clinical avenue for personalized treatment of MUD.
Collapse
Affiliation(s)
- Weiwen Tian
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai 200030, China
| | - Di Zhao
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai 200030, China
| | - Jinjun Ding
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai 200030, China
| | - Shulu Zhan
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai 200030, China
| | - Yi Zhang
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai 200030, China
| | - Amit Etkin
- Department of Psychiatry and Behavioral Science, Stanford University, Stanford, CA 94305, USA; Wu Tsai Neuroscience Institute, Stanford University, Stanford, CA 94305, USA; Alto Neuroscience, Inc., Los Altos, CA 94022, USA
| | - Wei Wu
- Department of Psychiatry and Behavioral Science, Stanford University, Stanford, CA 94305, USA; Wu Tsai Neuroscience Institute, Stanford University, Stanford, CA 94305, USA; Alto Neuroscience, Inc., Los Altos, CA 94022, USA.
| | - Ti-Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai 200030, China; Institute of Mental Health and Drug Discovery, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226019, China.
| |
Collapse
|
13
|
Pang B, Wu X, Chen H, Yan Y, Du Z, Yu Z, Yang X, Wang W, Lu K. Exploring the memory: existing activity-dependent tools to tag and manipulate engram cells. Front Cell Neurosci 2024; 17:1279032. [PMID: 38259503 PMCID: PMC10800721 DOI: 10.3389/fncel.2023.1279032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/17/2023] [Indexed: 01/24/2024] Open
Abstract
The theory of engrams, proposed several years ago, is highly crucial to understanding the progress of memory. Although it significantly contributes to identifying new treatments for cognitive disorders, it is limited by a lack of technology. Several scientists have attempted to validate this theory but failed. With the increasing availability of activity-dependent tools, several researchers have found traces of engram cells. Activity-dependent tools are based on the mechanisms underlying neuronal activity and use a combination of emerging molecular biological and genetic technology. Scientists have used these tools to tag and manipulate engram neurons and identified numerous internal connections between engram neurons and memory. In this review, we provide the background, principles, and selected examples of applications of existing activity-dependent tools. Using a combination of traditional definitions and concepts of engram cells, we discuss the applications and limitations of these tools and propose certain developmental directions to further explore the functions of engram cells.
Collapse
Affiliation(s)
- Bo Pang
- The Second Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Xiaoyan Wu
- The First Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Hailun Chen
- The Second Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Yiwen Yan
- School of Basic Medicine Science, Southern Medical University, Guangzhou, China
| | - Zibo Du
- The First Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Zihan Yu
- School of Basic Medicine Science, Southern Medical University, Guangzhou, China
| | - Xiai Yang
- Department of Neurology, Ankang Central Hospital, Ankang, China
| | - Wanshan Wang
- Laboratory Animal Management Center, Southern Medical University, Guangzhou, China
- Guangzhou Southern Medical Laboratory Animal Sci. and Tech. Co., Ltd., Guangzhou, China
| | - Kangrong Lu
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Southern Medical University, Guangzhou, China
| |
Collapse
|
14
|
Davidson CJ, Mascarin AT, Yahya MA, Rubio FJ, Gheidi A. Approaches and considerations of studying neuronal ensembles: a brief review. Front Cell Neurosci 2023; 17:1310724. [PMID: 38155864 PMCID: PMC10752959 DOI: 10.3389/fncel.2023.1310724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/27/2023] [Indexed: 12/30/2023] Open
Abstract
First theorized by Hebb, neuronal ensembles have provided a framework for understanding how the mammalian brain operates, especially regarding learning and memory. Neuronal ensembles are discrete, sparsely distributed groups of neurons that become activated in response to a specific stimulus and are thought to provide an internal representation of the world. Beyond the study of region-wide or projection-wide activation, the study of ensembles offers increased specificity and resolution to identify and target specific memories or associations. Neuroscientists interested in the neurobiology of learning, memory, and motivated behavior have used electrophysiological-, calcium-, and protein-based proxies of neuronal activity in preclinical models to better understand the neurobiology of learned and motivated behaviors. Although these three approaches may be used to pursue the same general goal of studying neuronal ensembles, technical differences lead to inconsistencies in the output and interpretation of data. This mini-review highlights some of the methodologies used in electrophysiological-, calcium-, and protein-based studies of neuronal ensembles and discusses their strengths and weaknesses.
Collapse
Affiliation(s)
- Cameron J. Davidson
- William Beaumont School of Medicine, Oakland University, Rochester, MI, United States
| | - Alixandria T. Mascarin
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Majd A. Yahya
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - F. Javier Rubio
- Neuronal Ensembles in Addiction Section, Behavioral Neuroscience Research Branch, Intramural Research Program/National Institute on Drug Abuse/National Institutes of Health, Bethesda, MD, United States
| | - Ali Gheidi
- Department of Biomedical Sciences, Mercer University, Macon, GA, United States
| |
Collapse
|
15
|
Doyle MA, Taylor A, Winder DG. Neural Circuitries and Alcohol Use Disorder: Cutting Corners in the Cycle. Curr Top Behav Neurosci 2023. [PMID: 38082108 DOI: 10.1007/7854_2023_454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2024]
Abstract
An implicit tenet of the alcohol use disorder (AUD) research field is that knowledge of how alcohol interacts with the brain is critical to the development of an understanding of vulnerability to AUD and treatment approaches. Gaining this understanding requires the mapping of brain function critical to specific components of this heterogeneous disorder. Early approaches in humans and animal models focused on the determination of specific brain regions sensitive to alcohol action and their participation in AUD-relevant behaviors. Broadly speaking, this research has focused on three domains, Binge/Intoxication, Negative Affect/Withdrawal, and Preoccupation/Anticipation, with a number of regions identified as participating in each. With the generational advances in technologies that the field of neuroscience has undergone over the last two decades, this focus has shifted to a circuit-based analysis. A wealth of new data has sharpened the field's focus on the specific roles of the interconnectivity of multiple brain regions in AUD and AUD-relevant behaviors, as well as demonstrating that the three major domains described above have much fuzzier edges than originally thought.In this chapter, we very briefly review brain regions previously implicated in aspects of AUD-relevant behavior from animal model research. Next, we move to a more in-depth overview of circuit-based approaches, and the utilization of these approaches in current AUD research.
Collapse
Affiliation(s)
- Marie A Doyle
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Anne Taylor
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Danny G Winder
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
16
|
Augustin SM, Gracias AL, Luo G, Anumola RC, Lovinger DM. Striatonigral direct pathway 2-arachidonoylglycerol contributes to ethanol effects on synaptic transmission and behavior. Neuropsychopharmacology 2023; 48:1941-1951. [PMID: 37528221 PMCID: PMC10584873 DOI: 10.1038/s41386-023-01671-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 07/12/2023] [Indexed: 08/03/2023]
Abstract
Endocannabinoids (eCB) and cannabinoid receptor 1 (CB1) play important roles in mediating short- and long-term synaptic plasticity in many brain regions involved in learning and memory, as well as the reinforcing effects of misused substances. Ethanol-induced plasticity and neuroadaptations predominantly occur in striatal direct pathway projecting medium spiny neurons (dMSNs). It is hypothesized that alterations in eCB neuromodulation may be involved. Recent work has implicated a role of eCB 2-arachidonoylglycerol (2-AG) in the rewarding effects of ethanol. However, there is insufficient research to answer which cellular subtype is responsible for mediating the 2-AG eCB signal that might be involved in the rewarding properties of ethanol and the mechanisms by which that occurs. To examine the role of dMSN mediated 2-AG signaling in ethanol related synaptic transmission and behaviors, we used conditional knockout mice in which the 2-AG-synthesizing enzyme diacylglycerol lipase α (DGLα) was deleted in dMSNs, DGLαD1-Cre+. Using acute brain slice photometry and a genetically encoded fluorescent eCB sensor, GRABeCB2.0, to assess real-time eCB mediated activity of sensorimotor inputs from primary motor cortices (M1/M2) to the dorsolateral striatum, we showed that DGLαD1-Cre+ mice had blunted evoked eCB-mediated presynaptic eCB signaling compared to littermate controls. Furthermore, ethanol induced eCB inhibition was significantly reduced in DGLαD1-Cre+ deficient mice. Additionally, there was a reduction in the duration of loss of righting reflex (LORR) to a high dose of ethanol in the DGLαD1-Cre+ mice compared to controls. These mice also showed a male-specific decrease in ethanol preference accompanied by an increase in ethanol-induced water consumption in a voluntary drinking paradigm. There were no significant differences observed in sucrose and quinine consumption between the genotypes. These findings reveal a novel role for dMSN mediated 2-AG signaling in modulating ethanol effects on presynaptic function and behavior.
Collapse
Affiliation(s)
- Shana M Augustin
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA.
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| | - Alexa L Gracias
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Guoxiang Luo
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Rishitha C Anumola
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - David M Lovinger
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
17
|
Matchynski JI, Cilley TS, Sadik N, Makki KM, Wu M, Manwar R, Woznicki AR, Kallakuri S, Arfken CL, Hope BT, Avanaki K, Conti AC, Perrine SA. Quantification of prefrontal cortical neuronal ensembles following conditioned fear learning in a Fos-LacZ transgenic rat with photoacoustic imaging in Vivo. PHOTOACOUSTICS 2023; 33:100551. [PMID: 38021296 PMCID: PMC10658601 DOI: 10.1016/j.pacs.2023.100551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 05/19/2023] [Accepted: 08/26/2023] [Indexed: 12/01/2023]
Abstract
Understanding the neurobiology of complex behaviors requires measurement of activity in the discrete population of active neurons, neuronal ensembles, which control the behavior. Conventional neuroimaging techniques ineffectively measure neuronal ensemble activity in the brain in vivo because they assess the average regional neuronal activity instead of the specific activity of the neuronal ensemble that mediates the behavior. Our functional molecular photoacoustic tomography (FM-PAT) system allows direct imaging of Fos-dependent neuronal ensemble activation in Fos-LacZ transgenic rats in vivo. We tested four experimental conditions and found increased FM-PAT signal in prefrontal cortical areas in rats undergoing conditioned fear or novel context exposure. A parallel immunofluorescence ex vivo study of Fos expression found similar findings. These findings demonstrate the ability of FM-PAT to measure Fos-expressing neuronal ensembles directly in vivo and support a mechanistic role for the prefrontal cortex in higher-order processing of response to specific stimuli or environmental cues.
Collapse
Affiliation(s)
- James I. Matchynski
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
- Translational Neuroscience Program, Wayne State University School of Medicine, Detroit, MI, USA
- John D. Dingell Veterans Affairs Medical Center, Detroit, MI, USA
- Wayne State MD/PhD Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Timothy S. Cilley
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nareen Sadik
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kassem M. Makki
- John D. Dingell Veterans Affairs Medical Center, Detroit, MI, USA
| | - Min Wu
- John D. Dingell Veterans Affairs Medical Center, Detroit, MI, USA
| | - Rayyan Manwar
- University of Illinois at Chicago, Department of Bioengineering, Chicago, IL, USA
| | | | - Srinivasu Kallakuri
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Cynthia L. Arfken
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
- Translational Neuroscience Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Bruce T. Hope
- The National Institute on Drug Abuse (NIDA), Intramural Research Program, Baltimore, MD, USA
| | - Kamran Avanaki
- University of Illinois at Chicago, Department of Bioengineering, Chicago, IL, USA
| | - Alana C. Conti
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
- Translational Neuroscience Program, Wayne State University School of Medicine, Detroit, MI, USA
- John D. Dingell Veterans Affairs Medical Center, Detroit, MI, USA
| | - Shane A. Perrine
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
- Translational Neuroscience Program, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
18
|
Zhao D, Zeng N, Zhang HB, Zhang Y, Shan J, Luo H, Zangen A, Yuan TF. Deep magnetic stimulation targeting the medial prefrontal and anterior cingulate cortices for methamphetamine use disorder: a randomised, double-blind, sham-controlled study. Gen Psychiatr 2023; 36:e101149. [PMID: 37781340 PMCID: PMC10533780 DOI: 10.1136/gpsych-2023-101149] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/25/2023] [Indexed: 10/03/2023] Open
Affiliation(s)
- Di Zhao
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ningning Zeng
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hang-Bin Zhang
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Zhang
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiatong Shan
- Department of Arts and Sciences, New York University Shanghai, Shanghai, China
| | - Huichun Luo
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Abraham Zangen
- Department of Life Sciences, Ben-Gurion University, Beer Sheva, Israel
| | - Ti-Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
19
|
Pagano R, Salamian A, Skonieczna E, Wojtas B, Gielniewski B, Harda Z, Cały A, Havekes R, Abel T, Radwanska K. Molecular fingerprints in the hippocampus of alcohol seeking during withdrawal. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554622. [PMID: 37662388 PMCID: PMC10473700 DOI: 10.1101/2023.08.24.554622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Alcohol use disorder (AUD) is characterized by excessive alcohol seeking and use. Here, we investigated the molecular correlates of impaired extinction of alcohol seeking using a multidimentional mouse model of AUD. We distinguished AUD-prone and AUD-resistant mice, based on the presence of ≥ 2 or < 2 criteria of AUD and utilized RNA sequencing to identify genes that were differentially expressed in the hippocampus and amygdala of mice meeting ≥ 2 or < 2 criteria, as these brain regions are implicated in alcohol motivation, seeking, consumption and the cognitive inflexibility characteristic of AUD. Our findings revealed dysregulation of the genes associated with the actin cytoskeleton, including actin binding molecule cofilin, and impaired synaptic transmission in the hippocampi of mice meeting ≥ 2 criteria. Overexpression of cofilin in the polymorphic layer of the dentate gyrus (PoDG) inhibited ML-DG synapses, increased motivation to seek alcohol and impaired extinction of alcohol seeking, resembling the phenotype observed in mice meeting ≥ 2 criteria. Overall, our study uncovers a novel mechanism linking increased hippocampal cofilin expression with the AUD phenotype.
Collapse
Affiliation(s)
- Roberto Pagano
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Ahmad Salamian
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Edyta Skonieczna
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Bartosz Wojtas
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Bartek Gielniewski
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Zofia Harda
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
- current address: Department Molecular Neuropharmacology, Maj Institute of Pharmacology of Polish Academy of Sciences, Krakow, Poland
| | - Anna Cały
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Robbert Havekes
- Neurobiology expertise group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Ted Abel
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Kasia Radwanska
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| |
Collapse
|
20
|
Chuong V, Farokhnia M, Khom S, Pince CL, Elvig SK, Vlkolinsky R, Marchette RC, Koob GF, Roberto M, Vendruscolo LF, Leggio L. The glucagon-like peptide-1 (GLP-1) analogue semaglutide reduces alcohol drinking and modulates central GABA neurotransmission. JCI Insight 2023; 8:e170671. [PMID: 37192005 PMCID: PMC10371247 DOI: 10.1172/jci.insight.170671] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/10/2023] [Indexed: 05/17/2023] Open
Abstract
Growing evidence indicates that the glucagon-like peptide-1 (GLP-1) system is involved in the neurobiology of addictive behaviors, and GLP-1 analogues may be used for the treatment of alcohol use disorder (AUD). Here, we examined the effects of semaglutide, a long-acting GLP-1 analogue, on biobehavioral correlates of alcohol use in rodents. A drinking-in-the-dark procedure was used to test the effects of semaglutide on binge-like drinking in male and female mice. We also tested the effects of semaglutide on binge-like and dependence-induced alcohol drinking in male and female rats, as well as acute effects of semaglutide on spontaneous inhibitory postsynaptic currents (sIPSCs) from central amygdala (CeA) and infralimbic cortex (ILC) neurons. Semaglutide dose-dependently reduced binge-like alcohol drinking in mice; a similar effect was observed on the intake of other caloric/noncaloric solutions. Semaglutide also reduced binge-like and dependence-induced alcohol drinking in rats. Semaglutide increased sIPSC frequency in CeA and ILC neurons from alcohol-naive rats, suggesting enhanced GABA release, but had no overall effect on GABA transmission in alcohol-dependent rats. In conclusion, the GLP-1 analogue semaglutide decreased alcohol intake across different drinking models and species and modulated central GABA neurotransmission, providing support for clinical testing of semaglutide as a potentially novel pharmacotherapy for AUD.
Collapse
Affiliation(s)
- Vicky Chuong
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program (NIDA IRP) and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research (NIAAA DICBR), NIH, Baltimore and Bethesda, Maryland, USA
- Neurobiology of Addiction Section, NIDA IRP, NIH, Baltimore, Maryland, USA
| | - Mehdi Farokhnia
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program (NIDA IRP) and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research (NIAAA DICBR), NIH, Baltimore and Bethesda, Maryland, USA
| | - Sophia Khom
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Claire L. Pince
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program (NIDA IRP) and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research (NIAAA DICBR), NIH, Baltimore and Bethesda, Maryland, USA
- Neurobiology of Addiction Section, NIDA IRP, NIH, Baltimore, Maryland, USA
| | - Sophie K. Elvig
- Neurobiology of Addiction Section, NIDA IRP, NIH, Baltimore, Maryland, USA
| | - Roman Vlkolinsky
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | | | - George F. Koob
- Neurobiology of Addiction Section, NIDA IRP, NIH, Baltimore, Maryland, USA
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Leandro F. Vendruscolo
- Stress and Addiction Neuroscience Unit, NIDA IRP and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, Maryland, USA
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program (NIDA IRP) and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research (NIAAA DICBR), NIH, Baltimore and Bethesda, Maryland, USA
| |
Collapse
|
21
|
Chang VN, Peters J. Neural circuits controlling choice behavior in opioid addiction. Neuropharmacology 2023; 226:109407. [PMID: 36592884 PMCID: PMC9898219 DOI: 10.1016/j.neuropharm.2022.109407] [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: 08/01/2022] [Revised: 12/23/2022] [Accepted: 12/25/2022] [Indexed: 12/31/2022]
Abstract
As the opioid epidemic presents an ever-expanding public health threat, there is a growing need to identify effective new treatments for opioid use disorder (OUD). OUD is characterized by a behavioral misallocation in choice behavior between opioids and other rewards, as opioid use leads to negative consequences, such as job loss, family neglect, and potential overdose. Preclinical models of addiction that incorporate choice behavior, as opposed to self-administration of a single drug reward, are needed to understand the neural circuits governing opioid choice. These choice models recapitulate scenarios that humans suffering from OUD encounter in their daily lives. Indeed, patients with substance use disorders (SUDs) exhibit a propensity to choose drug under certain conditions. While most preclinical addiction models have focused on relapse as the outcome measure, our data suggest that choice is an independent metric of addiction severity, perhaps relating to loss of cognitive control over choice, as opposed to excessive motivational drive to seek drugs during relapse. In this review, we examine both preclinical and clinical literature on choice behavior for drugs, with a focus on opioids, and the neural circuits that mediate drug choice versus relapse. We argue that preclinical models of opioid choice are needed to identify promising new avenues for OUD therapy that are translationally relevant. Both forward and reverse translation will be necessary to identify novel treatment interventions. This article is part of the Special Issue on "Opioid-induced changes in addiction and pain circuits".
Collapse
Affiliation(s)
- Victoria N Chang
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Jamie Peters
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, 80045, USA; Department of Pharmacology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
22
|
Stefaniuk M, Pawłowska M, Barański M, Nowicka K, Zieliński Z, Bijoch Ł, Legutko D, Majka P, Bednarek S, Jermakow N, Wójcik D, Kaczmarek L. Global brain c-Fos profiling reveals major functional brain networks rearrangements after alcohol reexposure. Neurobiol Dis 2023; 178:106006. [PMID: 36682503 DOI: 10.1016/j.nbd.2023.106006] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023] Open
Abstract
Many fundamental questions on alcohol use disorder (AUD) are frequently difficult to address by examining a single brain structure, but should be viewed from the whole brain perspective. c-Fos is a marker of neuronal activation. Global brain c-Fos profiling in rodents represents a promising platform to study brain functional networks rearrangements in AUD. We used a mouse model of alcohol drinking in IntelliCage. We trained mice to voluntarily drink alcohol, next subjected them to withdrawal and alcohol reexposure. We have developed a dedicated image computational workflow to identify c-Fos-positive cells in three-dimensional images obtained after whole-brain optical clearing and imaging in the light-sheet microscope. We provide a complete list of 169 brain structures with annotated c-Fos expression. We analyzed functional networks, brain modularity and engram index. Brain c-Fos levels in animals reexposed to alcohol were different from both control and binge drinking animals. Structures involved in reward processing, decision making and characteristic for addictive behaviors, such as precommissural nucleus, nucleus Raphe, parts of colliculus and tecta stood out particularly. Alcohol reexposure leads to a massive change of brain modularity including a formation of numerous smaller functional modules grouping structures involved in addiction development. Binge drinking can lead to substantial functional remodeling in the brain. We provide a list of structures that can be used as a target in pharmacotherapy but also point to the networks and modules that can hold therapeutic potential demonstrated by a clinical trial in patients.
Collapse
Affiliation(s)
- Marzena Stefaniuk
- Laboratory of Neurobiology, Nencki Institute, BRAINCITY, Warsaw, Poland.
| | - Monika Pawłowska
- Laboratory of Neurobiology, Nencki Institute, BRAINCITY, Warsaw, Poland; Institute of Experimental Physics, Section of Optics, Warsaw University, Warsaw, Poland
| | - Marcin Barański
- Laboratory of Neurobiology, Nencki Institute, BRAINCITY, Warsaw, Poland
| | - Klaudia Nowicka
- Laboratory of Neurobiology, Nencki Institute, BRAINCITY, Warsaw, Poland
| | | | - Łukasz Bijoch
- Laboratory of Neurobiology, Nencki Institute, BRAINCITY, Warsaw, Poland; Laboratory of Neuronal Plasticity, Nencki Institute, BRAINCITY, Warsaw, Poland
| | - Diana Legutko
- Laboratory of Neurobiology, Nencki Institute, BRAINCITY, Warsaw, Poland
| | - Piotr Majka
- Laboratory of Neuroinformatics, Nencki Institute, Warsaw, Poland
| | - Sylwia Bednarek
- Laboratory of Neuroinformatics, Nencki Institute, Warsaw, Poland
| | - Natalia Jermakow
- Laboratory of Neuroinformatics, Nencki Institute, Warsaw, Poland
| | - Daniel Wójcik
- Laboratory of Neuroinformatics, Nencki Institute, Warsaw, Poland
| | - Leszek Kaczmarek
- Laboratory of Neurobiology, Nencki Institute, BRAINCITY, Warsaw, Poland
| |
Collapse
|
23
|
Valyear MD, LeCocq MR, Brown A, Villaruel FR, Segal D, Chaudhri N. Learning processes in relapse to alcohol use: lessons from animal models. Psychopharmacology (Berl) 2023; 240:393-416. [PMID: 36264342 DOI: 10.1007/s00213-022-06254-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 10/02/2022] [Indexed: 11/29/2022]
Abstract
RATIONALE Alcohol use is reliably preceded by discrete and contextual stimuli which, through diverse learning processes, acquire the capacity to promote alcohol use and relapse to alcohol use. OBJECTIVE We review contemporary extinction, renewal, reinstatement, occasion setting, and sex differences research within a conditioning framework of relapse to alcohol use to inform the development of behavioural and pharmacological therapies. KEY FINDINGS Diverse learning processes and corresponding neurobiological substrates contribute to relapse to alcohol use. Results from animal models indicate that cortical, thalamic, accumbal, hypothalamic, mesolimbic, glutamatergic, opioidergic, and dopaminergic circuitries contribute to alcohol relapse through separable learning processes. Behavioural therapies could be improved by increasing the endurance and generalizability of extinction learning and should incorporate whether discrete cues and contexts influence behaviour through direct excitatory conditioning or occasion setting mechanisms. The types of learning processes that most effectively influence responding for alcohol differ in female and male rats. CONCLUSION Sophisticated conditioning experiments suggest that diverse learning processes are mediated by distinct neural circuits and contribute to relapse to alcohol use. These experiments also suggest that gender-specific behavioural and pharmacological interventions are a way towards efficacious therapies to prevent relapse to alcohol use.
Collapse
Affiliation(s)
- Milan D Valyear
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montréal, QC, Canada. .,Department of Psychology, McGill University, 1205 Ave. Dr. Penfield, Room N8/5, Montréal, QC, H3A 1B1, Canada.
| | - Mandy R LeCocq
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montréal, QC, Canada
| | - Alexa Brown
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montréal, QC, Canada
| | - Franz R Villaruel
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montréal, QC, Canada
| | - Diana Segal
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montréal, QC, Canada
| | - Nadia Chaudhri
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montréal, QC, Canada
| |
Collapse
|
24
|
Brown A, Chaudhri N. Optogenetic stimulation of infralimbic cortex projections to the paraventricular thalamus attenuates context-induced renewal. Eur J Neurosci 2023; 57:762-779. [PMID: 36373226 DOI: 10.1111/ejn.15862] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 10/11/2022] [Accepted: 10/27/2022] [Indexed: 11/16/2022]
Abstract
Contexts associated with prior reinforcement can renew extinguished conditioned responding. The prelimbic (PL) and infralimbic (IL) cortices are thought to mediate the expression and suppression of conditioned responding, respectively. Evidence suggests that PL inputs to the paraventricular nucleus of the thalamus (PVT) drive the expression of cue-induced reinstatement of drug seeking and that IL inputs to the PVT mediate fear extinction retrieval. However, the role of these projections in renewal of appetitive Pavlovian conditioned responding is unknown. We trained male and female Long-Evans rats to associate a conditioned stimulus (CS; 10 s white noise) with delivery of a 10% sucrose unconditioned stimulus (US; .2 ml/CS) to a fluid port in a distinct context (Context A). We then extinguished responding by presenting the CS without the US in a different context (Context B). At test, rats were returned to Context A, and optogenetic stimulation was delivered to either the IL-to-PVT or PL-to-PVT pathway during CS presentations. Optically stimulating the IL-to-PVT, but not the PL-to-PVT pathway, attenuated ABA renewal of CS port entries, and this effect was similar in males and females. Further, rats self-administered optical stimulation of the IL-to-PVT but not the PL-to-PVT pathway suggesting that activation of the IL-to-PVT pathway is reinforcing. The effectiveness of optical stimulation parameters to activate neurons in the IL, PL and PVT was confirmed using Fos immunohistochemistry. These findings provide evidence for novel neural mechanisms in renewal of responding to a sucrose-predictive CS, as well as more generally in contextual processing and appetitive associative learning.
Collapse
Affiliation(s)
- Alexa Brown
- Center for Studies in Behavioural Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| | - Nadia Chaudhri
- Center for Studies in Behavioural Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| |
Collapse
|
25
|
Brown A, Villaruel FR, Chaudhri N. Neural correlates of recall and extinction in a rat model of appetitive Pavlovian conditioning. Behav Brain Res 2023; 440:114248. [PMID: 36496079 DOI: 10.1016/j.bbr.2022.114248] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/30/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022]
Abstract
Extinction is a fundamental form of inhibitory learning that is important for adapting to changing environmental contingencies. While numerous studies have investigated the neural correlates of extinction using Pavlovian fear conditioning and appetitive operant reward-seeking procedures, less is known about the neural circuitry mediating the extinction of appetitive Pavlovian responding. Here, we aimed to generate an extensive brain activation map of extinction learning in a rat model of appetitive Pavlovian conditioning. Male Long-Evans rats were trained to associate a conditioned stimulus (CS; 20 s white noise) with the delivery of a 10% sucrose unconditioned stimulus (US; 0.3 ml/CS) to a fluid port. Control groups also received CS presentations, but sucrose was delivered either during the inter-trial interval or in the home-cage. After conditioning, 1 or 6 extinction sessions were conducted in which the CS was presented but sucrose was withheld. We performed Fos immunohistochemistry and network connectivity analyses on a set of cortical, striatal, thalamic, and amygdalar brain regions. Neural activity in the prelimbic cortex, ventral orbitofrontal cortex, nucleus accumbens core, and paraventricular nucleus of the thalamus was greater during recall relative to extinction. Conversely, prolonged extinction following 6 sessions induced increased neural activity in the infralimbic cortex, medial orbitofrontal cortex, and nucleus accumbens shell compared to home-cage controls. All these structures were similarly recruited during recall on the first extinction session. These findings provide novel evidence for the contribution of brain areas and neural networks that are differentially involved in the recall versus extinction of appetitive Pavlovian conditioned responding.
Collapse
Affiliation(s)
- Alexa Brown
- Center for Studies in Behavioural Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada.
| | - Franz R Villaruel
- Center for Studies in Behavioural Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada
| | - Nadia Chaudhri
- Center for Studies in Behavioural Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada
| |
Collapse
|
26
|
The Recruitment of a Neuronal Ensemble in the Central Nucleus of the Amygdala During the First Extinction Episode Has Persistent Effects on Extinction Expression. Biol Psychiatry 2023; 93:300-308. [PMID: 36336498 DOI: 10.1016/j.biopsych.2022.07.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/30/2022] [Accepted: 07/29/2022] [Indexed: 01/21/2023]
Abstract
BACKGROUND Adaptive behavior depends on the delicate and dynamic balance between acquisition and extinction memories. Disruption of this balance, particularly when the extinction of memory loses control over behavior, is the root of treatment failure of maladaptive behaviors such as substance abuse or anxiety disorders. Understanding this balance requires a better understanding of the underlying neurobiology and its contribution to behavioral regulation. METHODS We microinjected Daun02 in Fos-lacZ transgenic rats following a single extinction training episode to delete extinction-recruited neuronal ensembles in the basolateral amygdala (BLA) and central nucleus of the amygdala (CN) and examined their contribution to behavior in an appetitive Pavlovian task. In addition, we used immunohistochemistry and neuronal staining methods to identify the molecular markers of activated neurons in the BLA and CN during extinction learning or retrieval. RESULTS CN neurons were preferentially engaged following extinction, and deletion of these extinction-activated ensembles in the CN but not the BLA impaired the retrieval of extinction despite additional extinction training and promoted greater levels of behavioral restoration in spontaneous recovery and reinstatement. Disrupting extinction processing in the CN in turn increased activity in the BLA. Our results also show a specific role for CN PKCδ+ neurons in behavioral inhibition but not during initial extinction learning. CONCLUSIONS We showed that the initial extinction-recruited CN ensemble is critical to the acquisition-extinction balance and that greater behavioral restoration does not mean weaker extinction contribution. These findings provide a novel avenue for thinking about the neural mechanisms of extinction and for developing treatments for cue-triggered appetitive behaviors.
Collapse
|
27
|
Arnold ME, Butts AN, Erlenbach TR, Amico KN, Schank JR. Sex differences in neuronal activation during aversion-resistant alcohol consumption. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:240-250. [PMID: 36575056 PMCID: PMC9992309 DOI: 10.1111/acer.15006] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 12/15/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND One of the DSM-5 criteria for Alcohol Use Disorder is continued alcohol consumption despite negative consequences. This has been modeled in mice using adulteration of alcohol solution with the bitter tastant quinine. Mice that continue to consume alcohol despite this adulteration are considered aversion resistant. The limited number of studies dissecting the underlying neuronal mechanisms of aversion-resistant drinking behaviors used only male subjects. We have previously shown that female mice are more resistant to quinine adulteration of alcohol than males. Our aim here is to identify potential sex differences in neuronal activation that may underlie this behavior. METHODS Male and female C57BL/6J mice were allowed continuous access to 20% alcohol in a two-bottle choice procedure. To test aversion-resistance, the alcohol was adulterated with increasing concentrations (0.03, 0.1, and 0.2 mM) of quinine hydrochloride. After consumption rates were calculated, brains were extracted to examine neuronal activation using Fos immunohistochemistry. RESULTS We found that female mice suppressed their intake to a lesser extent than males when the alcohol solution was adulterated with quinine. Our Fos staining revealed three regions of interest that exhibit a sex difference during quinine-adulterated alcohol drinking: the ventromedial prefrontal cortex (vmPFC), the posterior insular cortex (PIC), and the ventral tegmental area (VTA). Both the vmPFC and the PIC exhibited higher neuronal activation in males during quinine-adulterated alcohol consumption. However, females showed higher Fos activation in the VTA during quinine-adulterated alcohol consumption. CONCLUSIONS Females more readily exhibit aversion-resistant alcohol intake than their male counterparts and exhibit some differences in neuronal activation patterns. We conclude that there are sex differences in neurocircuitry that may underlie compulsive drinking behaviors.
Collapse
Affiliation(s)
- Miranda E Arnold
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Arielle N Butts
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Theresa R Erlenbach
- Department of Genetics, Franklin College of Arts and Science, University of Georgia, Athens, Georgia, USA
| | - Kristen N Amico
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Jesse R Schank
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
28
|
Griffin WC, Lopez MF, Woodward JJ, Becker HC. Alcohol dependence and the ventral hippocampal influence on alcohol drinking in male mice. Alcohol 2023; 106:44-54. [PMID: 36328184 PMCID: PMC9868110 DOI: 10.1016/j.alcohol.2022.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/26/2022] [Accepted: 10/26/2022] [Indexed: 11/07/2022]
Abstract
Examining neural circuits underlying persistent, heavy drinking provides insight into the neurobiological mechanisms driving alcohol use disorder. Facilitated by its connectivity with other parts of the brain such as the nucleus accumbens (NAc), the ventral hippocampus (vHC) supports many behaviors, including those related to reward seeking and addiction. These studies used a well-established mouse model of alcohol (ethanol) dependence. After surgery to infuse DREADD-expressing viruses (hM4Di, hM3Dq, or mCherry-only) into the vHC and position guide cannula above the NAc, male C57BL/6J mice were treated in the CIE drinking model that involved repeated cycles of chronic intermittent alcohol (CIE) vapor or air (CTL) exposure alternating with weekly test drinking cycles in which mice were offered alcohol (15% v/v) 2 h/day. Additionally, smaller groups of mice were evaluated for either cFos expression or glutamate release using microdialysis procedures. In CIE mice expressing inhibitory (hM4Di) DREADDs in the vHC, drinking increased as expected, but CNO (3 mg/kg intraperitoneally [i.p.]) given 30 min before testing did not alter alcohol intake. However, in CTL mice expressing hM4Di, CNO significantly increased alcohol drinking (∼30%; p < 0.05) to levels similar to the CIE mice. The vHC-NAc pathway was targeted by infusing CNO into the NAc (3 or 10 μM/side) 30 min before testing. CNO activation of the pathway in mice expressing excitatory (hM3Dq) DREADDs selectively reduced consumption in CIE mice back to CTL levels (∼35-45%; p < 0.05) without affecting CTL alcohol intake. Lastly, activating the vHC-NAc pathway increased cFos expression and evoked significant glutamate release from the vHC terminals in the NAc. These data indicate that reduced activity of the vHC increases alcohol consumption and that targeted, increased activity of the vHC-NAc pathway attenuates excessive drinking associated with alcohol dependence. Thus, these findings indicate that the vHC and its glutamatergic projections to the NAc are involved in excessive alcohol drinking.
Collapse
Affiliation(s)
- William C Griffin
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Science, Medical University of South Carolina, Charleston, SC, United States.
| | - Marcelo F Lopez
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Science, Medical University of South Carolina, Charleston, SC, United States
| | - John J Woodward
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Science, Medical University of South Carolina, Charleston, SC, United States; Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Howard C Becker
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Science, Medical University of South Carolina, Charleston, SC, United States; Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States; Ralph H. Johnson VA Medical Center, Charleston, SC 29425-0742, United States
| |
Collapse
|
29
|
Domi E, Barchiesi R, Barbier E. Epigenetic Dysregulation in Alcohol-Associated Behaviors: Preclinical and Clinical Evidence. Curr Top Behav Neurosci 2023. [PMID: 36717533 DOI: 10.1007/7854_2022_410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Alcohol use disorder (AUD) is characterized by loss of control over intake and drinking despite harmful consequences. At a molecular level, AUD is associated with long-term neuroadaptations in key brain regions that are involved in reward processing and decision-making. Over the last decades, a great effort has been made to understand the neurobiological basis underlying AUD. Epigenetic mechanisms have emerged as an important mechanism in the regulation of long-term alcohol-induced gene expression changes. Here, we review the literature supporting a role for epigenetic processes in AUD. We particularly focused on the three most studied epigenetic mechanisms: DNA methylation, Histone modification and non-coding RNAs. Clinical studies indicate an association between AUD and DNA methylation both at the gene and global levels. Using behavioral paradigms that mimic some of the characteristics of AUD, preclinical studies demonstrate that changes in epigenetic mechanisms can functionally impact alcohol-associated behaviors. While many studies support a therapeutic potential for targeting epigenetic enzymes, more research is needed to fully understand their role in AUD. Identification of brain circuits underlying alcohol-associated behaviors has made major advances in recent years. However, there are very few studies that investigate how epigenetic mechanisms can affect these circuits or impact the neuronal ensembles that promote alcohol-associated behaviors. Studies that focus on the role of circuit-specific and cell-specific epigenetic changes for clinically relevant alcohol behaviors may provide new insights on the functional role of epigenetic processes in AUD.
Collapse
Affiliation(s)
- Esi Domi
- Department of Biomedical and Clinical Sciences, Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
- School of Pharmacy, Pharmacology Unit, Center for Neuroscience, University of Camerino, Camerino, Italy
| | - Riccardo Barchiesi
- Department of Neuroscience, Waggoner Center for Alcohol and Alcohol Addiction Research, University of Texas at Austin, Austin, TX, USA
| | - Estelle Barbier
- Department of Biomedical and Clinical Sciences, Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden.
| |
Collapse
|
30
|
Fredriksson I, Tsai PJ, Shekara A, Duan Y, Applebey SV, Minier-Toribio A, Batista A, Chow JJ, Altidor L, Barbier E, Cifani C, Li X, Reiner DJ, Rubio FJ, Hope BT, Yang Y, Bossert JM, Shaham Y. Role of ventral subiculum neuronal ensembles in incubation of oxycodone craving after electric barrier-induced voluntary abstinence. SCIENCE ADVANCES 2023; 9:eadd8687. [PMID: 36630511 PMCID: PMC9833671 DOI: 10.1126/sciadv.add8687] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 12/14/2022] [Indexed: 06/17/2023]
Abstract
High relapse rate is a key feature of opioid addiction. In humans, abstinence is often voluntary due to negative consequences of opioid seeking. To mimic this human condition, we recently introduced a rat model of incubation of oxycodone craving after electric barrier-induced voluntary abstinence. Incubation of drug craving refers to time-dependent increases in drug seeking after cessation of drug self-administration. Here, we used the activity marker Fos, muscimol-baclofen (GABAa + GABAb receptor agonists) global inactivation, Daun02-selective inactivation of putative relapse-associated neuronal ensembles, and fluorescence-activated cell sorting of Fos-positive cells and quantitative polymerase chain reaction to demonstrate a key role of vSub neuronal ensembles in incubation of oxycodone craving after voluntary abstinence, but not homecage forced abstinence. We also used a longitudinal functional magnetic resonance imaging method and showed that functional connectivity changes in vSub-related circuits predict opioid relapse after abstinence induced by adverse consequences of opioid seeking.
Collapse
Affiliation(s)
- Ida Fredriksson
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, USA
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
| | - Pei-Jung Tsai
- Neuroimaging Research Branch, IRP/NIDA/NIH, Baltimore, MD, USA
| | | | - Ying Duan
- Neuroimaging Research Branch, IRP/NIDA/NIH, Baltimore, MD, USA
| | | | | | - Ashley Batista
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, USA
| | - Jonathan J. Chow
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, USA
| | - Lindsay Altidor
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, USA
| | - Estelle Barbier
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
| | - Carlo Cifani
- School of Pharmacy, University of Camerino, Camerino, Italy
| | - Xuan Li
- Department of Psychology, University of Maryland College Park, College Park, MD, USA
| | - David J. Reiner
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, USA
| | - F. Javier Rubio
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, USA
| | - Bruce T. Hope
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, USA
| | - Yihong Yang
- Neuroimaging Research Branch, IRP/NIDA/NIH, Baltimore, MD, USA
| | | | - Yavin Shaham
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, USA
| |
Collapse
|
31
|
Keefer SE, Petrovich GD. Necessity and recruitment of cue-specific neuronal ensembles within the basolateral amygdala during appetitive reversal learning. Neurobiol Learn Mem 2022; 194:107663. [PMID: 35870716 PMCID: PMC10326893 DOI: 10.1016/j.nlm.2022.107663] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/14/2022] [Accepted: 07/17/2022] [Indexed: 11/28/2022]
Abstract
Through Pavlovian appetitive conditioning, environmental cues can become predictors of food availability. Over time, however, the food, and thus the value of the associated cues, can change based on environmental variations. This change in outcome necessitates updating of the value of the cue to appropriately alter behavioral responses to these cues. The basolateral amygdala (BLA) is critical in updating the outcomes of learned cues. However, it is unknown if the same BLA neuronal ensembles that are recruited in the initial associative memory are required when the new cue-outcome association is formed during reversal learning. The current study used the Daun02 inactivation method that enables selective targeting and disruption of activated neuronal ensembles in Fos-lacZ transgenic rats. Rats were implanted with bilateral cannulas that target the BLA and underwent appetitive discriminative conditioning in which rats had to discriminate between two auditory stimuli. One stimulus (CS+) co-terminated with food delivery, and the other stimulus was unrewarded (CS-; counterbalanced). Rats were then tested for CS+ or CS- memory retrieval and infused with either Daun02 or a vehicle solution into the BLA to inactivate either CS+ or CS- neuronal ensembles that were activated during that test. To assess if the same neuronal ensembles are necessary to update the value of the new association when the outcomes are changed, rats underwent reversal learning: the CS+ was no longer followed by food (reversal CS-, rCS-), and the CS- was now followed by food (reversal CS+; rCS+). The group that received Daun02 following CS+ session showed a decrease in conditioned responding and increased latency to the rCS- (previously CS+) during the first session of reversal learning, specifically during the first trial. This indicates that the neuronal ensemble that was activated during the recall of the CS+ memory was the same neuronal ensemble needed for learning the new outcome of the same CS, now rCS-. Additionally, the group that received Daun02 following CS- session was slower to respond to the rCS+ (previously CS-) during reversal learning. This indicates that the neuronal ensemble that was activated during the recall of the CS- memory was the same neuronal ensemble needed for learning the new outcome of the same CS. These results demonstrate that different neuronal ensembles within the BLA mediate memory recall of CS+ and CS- cues and reactivation of each cue-specific neuronal ensemble is necessary to update the value of that specific cue to respond appropriately during reversal learning. These results also indicate substantial plasticity within the BLA for behavioral flexibility as both groups eventually showed similar terminal levels of reversal learning.
Collapse
Affiliation(s)
- Sara E Keefer
- Department of Psychology and Neuroscience, Boston College, 140 Commonwealth Avenue, Chestnut Hill, MA 02467, USA.
| | - Gorica D Petrovich
- Department of Psychology and Neuroscience, Boston College, 140 Commonwealth Avenue, Chestnut Hill, MA 02467, USA
| |
Collapse
|
32
|
Körber C, Sommer WH. From ensembles to meta-ensembles: Specific reward encoding by correlated network activity. Front Behav Neurosci 2022; 16:977474. [PMID: 36177094 PMCID: PMC9513968 DOI: 10.3389/fnbeh.2022.977474] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/15/2022] [Indexed: 11/29/2022] Open
Abstract
Neuronal ensembles are local, sparsely distributed populations of neurons that are reliably re-activated by a specific stimulus, context or task. Such discrete cell populations can be defined either functionally, by electrophysiological recordings or in vivo calcium imaging, or anatomically, using the expression of markers such as the immediate early gene cFos. A typical example of tasks that involve the formation of neuronal ensembles is reward learning, such as the cue-reward pairing during operant conditioning. These ensembles are re-activated during cue-presentation and increasing evidence suggests that this re-activation is the neurophysiological basis for the execution of reward-seeking behavior. Whilst the pursuit of rewards is a common daily activity, it is also related to the consumption of drugs, such as alcohol, and may result in problematic behaviors including addiction. Recent research has identified neuronal ensembles in several reward-related brain regions that control distinct aspects of a conditioned response, e.g., contextual information about the availability of a specific reward or the actions needed to retrieve this reward under the given circumstances. Here, we review studies using the activity marker cFos to identify and characterize neuronal ensembles related to alcohol and non-drug rewards with a special emphasis on the discrimination between different rewards by meta-ensembles, i.e., by dynamic co-activation of multiple ensembles across different brain areas.
Collapse
Affiliation(s)
- Christoph Körber
- Department of Functional Neuroanatomy, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Wolfgang H Sommer
- Medical Faculty Mannheim, Institute of Psychopharmacology, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| |
Collapse
|
33
|
Harel M, Perini I, Kämpe R, Alyagon U, Shalev H, Besser I, Sommer WH, Heilig M, Zangen A. Repetitive Transcranial Magnetic Stimulation in Alcohol Dependence: A Randomized, Double-Blind, Sham-Controlled Proof-of-Concept Trial Targeting the Medial Prefrontal and Anterior Cingulate Cortices. Biol Psychiatry 2022; 91:1061-1069. [PMID: 35067356 DOI: 10.1016/j.biopsych.2021.11.020] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 11/10/2021] [Accepted: 11/10/2021] [Indexed: 11/02/2022]
Abstract
BACKGROUND Alcohol addiction is associated with a high disease burden, and treatment options are limited. In a proof-of-concept study, we used deep repetitive transcranial magnetic stimulation (dTMS) to target circuitry associated with the pathophysiology of alcohol addiction. We evaluated clinical outcomes and explored associated neural signatures using functional magnetic resonance imaging. METHODS This was a double-blind, randomized, sham-controlled trial. A total of 51 recently abstinent treatment-seeking patients with alcohol use disorder (moderate to severe) were randomized to sham or active dTMS, using an H7 coil targeting midline frontocortical areas, including the medial prefrontal and anterior cingulate cortices. Treatment included 15 sessions over 3 weeks, followed by five sessions over 3 months of follow-up. Each session delivered 100 trains of 30 pulses at 10 Hz. The primary predefined outcome was reduction in percentage of heavy drinking days, obtained using timeline follow-back interviews. Secondary analyses included self-reports of craving, ethyl glucuronide in urine, and brain imaging measures. RESULTS Both craving after treatment and percentage of heavy drinking days during follow-up were significantly lower in the active versus sham control group (percentage of heavy drinking days = 2.9 ± 0.8% vs. 10.6 ± 1.9%, p = .037). Active dTMS was associated with decreased resting-state functional connectivity of the dorsal anterior cingulate cortex with the caudate nucleus and decreased connectivity of the medial prefrontal cortex to the subgenual anterior cingulate cortex. CONCLUSIONS We provide initial proof-of-concept for dTMS targeting midline frontocortical structures as a treatment for alcohol addiction. These data strongly support a rationale for a full-scale confirmatory multicenter trial. Therapeutic benefits of dTMS appear to be associated with persistent changes in brain network activity.
Collapse
Affiliation(s)
- Maayan Harel
- Department of Life Sciences, Ben-Gurion University, Beer Sheva, Israel; Zlotowski Center for Neuroscience, Ben-Gurion University, Beer Sheva, Israel
| | - Irene Perini
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University Hospital, Linköping, Sweden
| | - Robin Kämpe
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University Hospital, Linköping, Sweden
| | - Uri Alyagon
- Department of Life Sciences, Ben-Gurion University, Beer Sheva, Israel; Zlotowski Center for Neuroscience, Ben-Gurion University, Beer Sheva, Israel
| | - Hadar Shalev
- Zlotowski Center for Neuroscience, Ben-Gurion University, Beer Sheva, Israel; Department of Psychiatry, Ben-Gurion University and Soroka Medical Center, Beer Sheva, Israel
| | - Itay Besser
- Zlotowski Center for Neuroscience, Ben-Gurion University, Beer Sheva, Israel; Department of Psychiatry, Ben-Gurion University and Soroka Medical Center, Beer Sheva, Israel
| | - Wolfgang H Sommer
- Institute of Psychopharmacology, Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim, Mannheim, Germany; Bethanien Hospital for Psychiatry, Psychosomatics, and Psychotherapy, Greifswald, Germany
| | - Markus Heilig
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University Hospital, Linköping, Sweden; Department of Psychiatry, Linköping University Hospital, Linköping, Sweden.
| | - Abraham Zangen
- Department of Life Sciences, Ben-Gurion University, Beer Sheva, Israel; Zlotowski Center for Neuroscience, Ben-Gurion University, Beer Sheva, Israel.
| |
Collapse
|
34
|
Flores-Ramirez FJ, Matzeu A, Sánchez-Marín L, Martin-Fardon R. Blockade of corticotropin-releasing factor-1 receptors in the infralimbic cortex prevents stress-induced reinstatement of alcohol seeking in male Wistar rats: Evidence of interaction between CRF 1 and orexin receptor signaling. Neuropharmacology 2022; 210:109046. [PMID: 35341789 PMCID: PMC9176217 DOI: 10.1016/j.neuropharm.2022.109046] [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: 10/06/2021] [Revised: 03/11/2022] [Accepted: 03/21/2022] [Indexed: 10/18/2022]
Abstract
Alcohol use dysregulates responsivity to stress, which is mediated by corticotropin-releasing factor (CRF). With repeated cycles of alcohol use, the hypothalamic-pituitary-adrenal axis becomes hyporesponsive, rendering individuals vulnerable to the reinstatement of alcohol-seeking behavior during stressful episodes. Orexin (Orx; also called hypocretin) plays a well-established role in regulating diverse physiological processes, including stress, and interacts with CRF. The infralimbic cortex (IL) is a CRF-rich region. Anatomical evidence suggests that CRF and Orx interact in this area. To test the behavioral implication of CRF and Orx transmission in the IL during the stress-induced reinstatement of alcohol-seeking behavior, male Wistar rats were trained to self-administer 10% alcohol for 3 weeks. The rats then underwent two weeks of extinction training (identical to the alcohol self-administration sessions, but alcohol was withheld). The day after the last extinction session, the rats received a bilateral intra-IL injection of the CRF1 receptor antagonist CP154,526 (0.6 μg/0.5 μl/side), the dual Orx receptor antagonist TCS1102 (15 μg/0.5 μl/side), or their combination and then were tested for the footshock stress-induced reinstatement of alcohol-seeking behavior. CP154,526 significantly prevented reinstatement, but TCS1102 did not produce such an effect. Interestingly, the co-administration of TCS1102 and CP154,526 reversed the effect of CP154,526 alone, and footshock stress induced a significant increase in Crhr1 and Hcrtr2 mRNA expression in the IL. These results demonstrate a functional interaction between Orx receptor and CRF1 receptor signaling and suggest that CRF1 receptor antagonism may ameliorate stress-induced alcohol-seeking behavior.
Collapse
Affiliation(s)
| | - Alessandra Matzeu
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Laura Sánchez-Marín
- Unidad Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga, Hospital Regional Universitario de Málaga/Universidad de Málaga, Málaga, Spain
| | - Rémi Martin-Fardon
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
35
|
Pérez-Ramírez Ú, López-Madrona VJ, Pérez-Segura A, Pallarés V, Moreno A, Ciccocioppo R, Hyytiä P, Sommer WH, Moratal D, Canals S. Brain Network Allostasis after Chronic Alcohol Drinking Is Characterized by Functional Dedifferentiation and Narrowing. J Neurosci 2022; 42:4401-4413. [PMID: 35437279 PMCID: PMC9145238 DOI: 10.1523/jneurosci.0389-21.2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/25/2022] [Accepted: 03/30/2022] [Indexed: 11/21/2022] Open
Abstract
Alcohol use disorder (AUD) causes complex alterations in the brain that are poorly understood. The heterogeneity of drinking patterns and the high incidence of comorbid factors compromise mechanistic investigations in AUD patients. Here we used male Marchigian Sardinian alcohol-preferring (msP) rats, a well established animal model of chronic alcohol drinking, and a combination of longitudinal resting-state fMRI and manganese-enhanced MRI to provide objective measurements of brain connectivity and activity, respectively. We found that 1 month of chronic alcohol drinking changed the correlation between resting-state networks. The change was not homogeneous, resulting in the reorganization of pairwise interactions and a shift in the equilibrium of functional connections. We identified two fundamentally different forms of network reorganization. First is functional dedifferentiation, which is defined as a regional increase in neuronal activity and overall correlation, with a concomitant decrease in preferential connectivity between specific networks. Through this mechanism, occipital cortical areas lost their specific interaction with sensory-insular cortex, striatal, and sensorimotor networks. Second is functional narrowing, which is defined as an increase in neuronal activity and preferential connectivity between specific brain networks. Functional narrowing strengthened the interaction between striatal and prefrontocortical networks, involving the anterior insular, cingulate, orbitofrontal, prelimbic, and infralimbic cortices. Importantly, these two types of alterations persisted after alcohol discontinuation, suggesting that dedifferentiation and functional narrowing rendered persistent network states. Our results support the idea that chronic alcohol drinking, albeit at moderate intoxicating levels, induces an allostatic change in the brain functional connectivity that propagates into early abstinence.SIGNIFICANCE STATEMENT Excessive consumption of alcohol is positioned among the top five risk factors for disease and disability. Despite this priority, the transformations that the nervous system undergoes from an alcohol-naive state to a pathologic alcohol drinking are not well understood. In our study, we use an animal model with proven translational validity to study this transformation longitudinally. The results show that shortly after chronic alcohol consumption there is an increase in redundant activity shared by brain structures, and the specific communication shrinks to a set of pathways. This functional dedifferentiation and narrowing are not reversed immediately after alcohol withdrawal but persist during early abstinence. We causally link chronic alcohol drinking with an early and abstinence-persistent retuning of the functional equilibrium of the brain.
Collapse
Affiliation(s)
- Úrsula Pérez-Ramírez
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, E-46022 Valencia, Spain
| | - Víctor J López-Madrona
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, 03550 Sant Joan d'Alacant, Spain
| | - Andrés Pérez-Segura
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, 03550 Sant Joan d'Alacant, Spain
| | - Vicente Pallarés
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, 03550 Sant Joan d'Alacant, Spain
| | - Andrea Moreno
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, 03550 Sant Joan d'Alacant, Spain
| | | | - Petri Hyytiä
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Wolfgang H Sommer
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159 Mannheim, Germany
| | - David Moratal
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, E-46022 Valencia, Spain
| | - Santiago Canals
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, 03550 Sant Joan d'Alacant, Spain
| |
Collapse
|
36
|
Hamel L, Cavdaroglu B, Yeates D, Nguyen D, Riaz S, Patterson D, Khan N, Kirolos N, Roper K, Ha QA, Ito R. Cortico-Striatal Control over Adaptive Goal-Directed Responding Elicited by Cues Signaling Sucrose Reward or Punishment. J Neurosci 2022; 42:3811-3822. [PMID: 35351827 PMCID: PMC9087743 DOI: 10.1523/jneurosci.2175-21.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 11/21/2022] Open
Abstract
The medial prefrontal cortex (mPFC) and nucleus accumbens (NAc) have been associated with the expression of adaptive and maladaptive behavior elicited by fear-related and drug-associated cues. However, reported effects of mPFC manipulations on cue-elicited natural reward-seeking and inhibition thereof have been varied, with few studies examining cortico-striatal contributions in tasks that require adaptive responding to cues signaling reward and punishment within the same session. The current study aimed to better elucidate the role of mPFC and NAc subdivisions, and their functional connectivity in cue-elicited adaptive responding using a novel discriminative cue responding task. Male Long-Evans rats learned to lever-press on a VR5 schedule for a discriminative cue signaling reward, and to avoid pressing the same lever in the presence of another cue signaling punishment. Postacquisition, prelimbic (PL) and infralimbic (IL) areas of the mPFC, NAc core, shell, PL-core, or IL-shell circuits were pharmacologically or chemogenetically inhibited while animals performed under (1) nonreinforced (extinction) conditions, where the appetitive and aversive cues were presented in alternating trials alone or as a compound stimulus; and (2) reinforced conditions, whereby cued responding was accompanied by associated outcomes. PL and IL inactivation attenuated nonreinforced and reinforced goal-directed cue responding, whereas NAc core and shell inactivation impaired nonreinforced responding for the appetitive, but not aversive cue. Furthermore, PL-core and IL-shell inhibition disinhibited nonreinforced but not reinforced cue responding. Our findings implicate the mPFC as a site of confluence of motivationally significant cues and outcomes, and in the regulation of nonreinforced cue responding via downstream NAc targets.SIGNIFICANCE STATEMENT The ability to discriminate and respond appropriately to environmental cues that signal availability of reward or punishment is essential for survival. The medial prefrontal cortex (mPFC) and nucleus accumbens (NAc) have been implicated in adaptive and maladaptive behavior elicited by fear-related and drug-associated cues. However, less is known about the role they play in orchestrating adaptive responses to natural reward and punishment cues within the same behavioral task. Here, using a novel discriminative cue responding task combined with pharmacological or chemogenetic inhibition of mPFC, NAc and mPFC-NAc circuits, we report that mPFC is critically involved in responding to changing cued response-outcomes, both when the responses are reinforced, and nonreinforced. Furthermore, the mPFC coordinates nonreinforced discriminative cue responding by suppressing inappropriate responding via downstream NAc targets.
Collapse
Affiliation(s)
- Laurie Hamel
- Department of Psychology (Scarborough), University of Toronto, Toronto, Ontario, M1C 1A4, Canada
| | - Bilgehan Cavdaroglu
- Department of Psychology (Scarborough), University of Toronto, Toronto, Ontario, M1C 1A4, Canada
| | - Dylan Yeates
- Department of Psychology (Scarborough), University of Toronto, Toronto, Ontario, M1C 1A4, Canada
| | - David Nguyen
- Department of Psychology (Scarborough), University of Toronto, Toronto, Ontario, M1C 1A4, Canada
| | - Sadia Riaz
- Department of Psychology (Scarborough), University of Toronto, Toronto, Ontario, M1C 1A4, Canada
| | - Dylan Patterson
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, M5S 3G5 Canada
| | - Nisma Khan
- Department of Psychology (Scarborough), University of Toronto, Toronto, Ontario, M1C 1A4, Canada
| | - Nardin Kirolos
- Department of Psychology (Scarborough), University of Toronto, Toronto, Ontario, M1C 1A4, Canada
| | - Katherine Roper
- Department of Psychology (Scarborough), University of Toronto, Toronto, Ontario, M1C 1A4, Canada
| | - Quynh An Ha
- Department of Psychology (Scarborough), University of Toronto, Toronto, Ontario, M1C 1A4, Canada
| | - Rutsuko Ito
- Department of Psychology (Scarborough), University of Toronto, Toronto, Ontario, M1C 1A4, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, M5S 3G5 Canada
| |
Collapse
|
37
|
Howland JG, Ito R, Lapish CC, Villaruel FR. The rodent medial prefrontal cortex and associated circuits in orchestrating adaptive behavior under variable demands. Neurosci Biobehav Rev 2022; 135:104569. [PMID: 35131398 PMCID: PMC9248379 DOI: 10.1016/j.neubiorev.2022.104569] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 12/17/2021] [Accepted: 02/01/2022] [Indexed: 11/28/2022]
Abstract
Emerging evidence implicates rodent medial prefrontal cortex (mPFC) in tasks requiring adaptation of behavior to changing information from external and internal sources. However, the computations within mPFC and subsequent outputs that determine behavior are incompletely understood. We review the involvement of mPFC subregions, and their projections to the striatum and amygdala in two broad types of tasks in rodents: 1) appetitive and aversive Pavlovian and operant conditioning tasks that engage mPFC-striatum and mPFC-amygdala circuits, and 2) foraging-based tasks that require decision making to optimize reward. We find support for region-specific function of the mPFC, with dorsal mPFC and its projections to the dorsomedial striatum supporting action control with higher cognitive demands, and ventral mPFC engagement in translating affective signals into behavior via discrete projections to the ventral striatum and amygdala. However, we also propose that defined mPFC subdivisions operate as a functional continuum rather than segregated functional units, with crosstalk that allows distinct subregion-specific inputs (e.g., internal, affective) to influence adaptive behavior supported by other subregions.
Collapse
Affiliation(s)
- John G Howland
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada.
| | - Rutsuko Ito
- Department of Psychology, University of Toronto-Scarborough, Toronto, ON, Canada.
| | - Christopher C Lapish
- Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA.
| | - Franz R Villaruel
- Department of Psychology, Concordia University, Montreal, QC, Canada.
| |
Collapse
|
38
|
Hasanpour Razmanjani N, Reisi P. Effects of selective orexin receptor-2 and cannabinoid receptor-1 antagonists on the response of medial prefrontal cortex neurons to tramadol. Synapse 2022; 76:e22232. [PMID: 35313383 DOI: 10.1002/syn.22232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 02/19/2022] [Accepted: 03/14/2022] [Indexed: 11/07/2022]
Abstract
Tramadol is widely used to control pain in various diseases, but the relevant mechanisms are less known despite the severe risks of abuse. The medial prefrontal cortex (mPFC) is one of the critical centers of the reward system. Studies have shown that orexins and endocannabinoids are likely to play an important role in addiction. In this study, the effect of orexin receptor-2 (OX2R) and endocannabinoid receptor-1 (CB1R) blockade on the neuronal activity of mPFC was investigated in response to tramadol in male rats. Tramadol was injected intraperitoneally, and its effects on the firing of mPFC pyramidal neurons were investigated using in vivo extracellular single-unit recording. Tramadol affected the pyramidal neuronal activity of the mPFC. AM251 (18 nmol/4 μl), as a selective CB1R antagonist, and TCS-OX2-29 (50 nmol/4 μl), as a selective OX2R antagonist, individually or simultaneously were microinjected into the lateral ventricle of the brain (intracerebroventricular, ICV). The results showed that the ratio of neurons with the excitatory/inhibitory or no responses was significantly changed by tramadol (p < .05). These changes were prevented by blockade of CB1Rs alone or blockade of OX2Rs and CB1Rs simultaneously (p < .05). However, blockade of these receptors in the vehicle group had no significant effect on neuronal activity. The findings of this study indicate the potential role of orexin and endocannabinoid systems in mediating the effects of tramadol in mPFC and the possible interaction between the two systems via OX2 and CB1 receptors. However, further studies are needed to identify these effects by examining intracellular signaling.
Collapse
Affiliation(s)
| | - Parham Reisi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
39
|
Gobin C, Sortman B, Rakela S, Quintana-Feliciano R, Warren BL. Fos-expressing neuronal ensembles in rat infralimbic cortex encode initial and maintained oxycodone seeking in rats. Addict Biol 2022; 27:e13148. [PMID: 35229934 PMCID: PMC10167745 DOI: 10.1111/adb.13148] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/11/2021] [Accepted: 01/10/2022] [Indexed: 12/26/2022]
Abstract
Neuronal ensembles within the infralimbic cortex (IL) and their projections to the nucleus accumbens (NAc) mediate opiate seeking in well-trained rats. However, it is unclear how early this circuitry is recruited during oxycodone self-administration. Here, we used retrograde labelling (CTb) and immunohistochemistry to identify NAc-projecting neurons in the IL that were activated during initial oxycodone seeking. Next, we sought to determine the role of IL neuronal ensembles in initial oxycodone self-administration. We used the Daun02 procedure in male and female Fos-LacZ rats to chemogenetically inactivate IL Fos-expressing neurons at different time points in oxycodone self-administration training: immediately after meeting criteria for acquisition of behaviour and following nine daily sessions with increasing schedules of reinforcement (FR1, FR2 and FR3) in which rats demonstrated stable oxycodone intake under increasing effort to self-administer. We found that Daun02 infusions attenuated oxycodone seeking at both the initial learning and well-trained time points. These results suggest that IL neuronal ensembles are formed during initial learning of oxycodone self-administration and required for the maintenance and expression of oxycodone seeking.
Collapse
Affiliation(s)
- Christina Gobin
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida, USA
| | - Bo Sortman
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida, USA
| | - Samantha Rakela
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida, USA
| | | | - Brandon L Warren
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
40
|
Sinha R, Fogelman N, Wemm S, Angarita G, Seo D, Hermes G. Alcohol withdrawal symptoms predict corticostriatal dysfunction that is reversed by prazosin treatment in alcohol use disorder. Addict Biol 2022; 27:e13116. [PMID: 34856641 PMCID: PMC9872962 DOI: 10.1111/adb.13116] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/31/2021] [Accepted: 11/03/2021] [Indexed: 01/27/2023]
Abstract
Chronic alcohol use increases risk of alcohol withdrawal symptoms (AW) and disrupts stress biology and resilient coping, thereby promoting excessive alcohol intake. Chronic alcohol intake and multiple alcohol detoxifications are known to impair brain medial prefrontal cortex (mPFC) and striatal functioning, regions involved in regulating stress, craving and alcohol intake. In two related studies, we examined whether AW predicts this functional brain pathology and whether Prazosin versus Placebo treatment may reverse these effects. In Study 1, patients with Alcohol Use Disorder (AUD) (N = 45) with varying AW levels at treatment entry were assessed to examine AW effects on corticostriatal responses to stress, alcohol cue and neutral visual images with functional magnetic resonance imaging (fMRI). In Study 2, 23 AUD patients entering a 12-week randomised controlled trial (RCT) of Prazosin, an alpha1 adrenergic antagonist that decreased withdrawal-related alcohol intake in laboratory animals, participated in two fMRI sessions at pretreatment and also at week 9-10 of chronic treatment (Placebo: N = 13; Prazosin: N = 10) to assess Prazosin treatment effects on alcohol-related cortico-striatal dysfunction. Study 1 results indicated that higher AW predicted greater disruption in brain mPFC and striatal response to stress and alcohol cues (p < 0.001, family-wise error [FWE] correction) and also subsequently greater heavy drinking days (HDD) in early treatment (p < 0.01). In Study 2, Prazosin versus Placebo treatment reversed mPFC-striatal dysfunction (p < 0.001, FWE), which in turn predicted fewer drinking days (p < 0.01) during the 12-week treatment period. These results indicate that AW is a significant predictor of alcohol-related prefrontal-striatal dysfunction, and Prazosin treatment reversed these effects that in turn contributed to improved alcohol treatment outcomes.
Collapse
Affiliation(s)
- Rajita Sinha
- Yale Stress Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Nia Fogelman
- Yale Stress Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Stephanie Wemm
- Yale Stress Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Gustavo Angarita
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Dongju Seo
- Yale Stress Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Gretchen Hermes
- Yale Stress Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
41
|
Jessen K, Slaker Bennett ML, Liu S, Olsen CM. Comparison of prefrontal cortex sucrose seeking ensembles engaged in multiple seeking sessions: Context is key. J Neurosci Res 2022; 100:1008-1029. [PMID: 35137974 PMCID: PMC8940716 DOI: 10.1002/jnr.25025] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 12/31/2021] [Accepted: 01/17/2022] [Indexed: 12/11/2022]
Abstract
Encoding of memories, including those associated with prior drug or reward, is thought to take place within distinct populations of neurons, termed ensembles. Neuronal ensembles for drug- and reward-seeking have been identified in regions of the medial prefrontal cortex, but much of our understanding of these ensembles is based on experiments that take place in a single reward-associated environment and measure ensemble encoding over short durations of time. In contrast, reward seeking behavior is evident across different reward-associated environments and persists over time. Using TetTag mice and Fos immunohistochemistry, we examined the relationship between persistent sucrose-seeking and ensemble encoding in mice that undergo seeking sessions in the same or different sucrose self-administration contexts 2 weeks apart. We found that prelimbic (PrL) and anterior cingulate cortex ensembles tagged in the first seeking session were highly sensitive to the context in which a second seeking session took place: reactivation of these ensembles was reduced in the same context but elevated in a distinct sucrose self-administration context. Correlational analyses revealed that ensemble reactivation in the PrL was proportional to the persistence of sucrose seeking behavior across sessions in differing ways in female mice. In the same context, reactivation was proportional to the persistence of non-reinforced operant responses, whereas in a distinct context, reactivation was proportional to the persistence of non-reinforced head entries into the sucrose receptacle. This study underlines the importance of the medial prefrontal cortex importance in maintaining a reward-seeking ensemble over time and identifies context-dependent changes in behavioral correlates of ensemble reactivation.
Collapse
Affiliation(s)
- Kristen Jessen
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Megan L Slaker Bennett
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Department of Neuroscience, Wisconsin Lutheran College, Milwaukee, Wisconsin, USA
| | - Shuai Liu
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Christopher M Olsen
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
42
|
Guarque-Chabrera J, Gil-Miravet I, Olucha-Bordonau F, Melchor-Eixea I, Miquel M. When the front fails, the rear wins. Cerebellar correlates of prefrontal dysfunction in cocaine-induced memory in male rats. Prog Neuropsychopharmacol Biol Psychiatry 2022; 112:110429. [PMID: 34416354 DOI: 10.1016/j.pnpbp.2021.110429] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/04/2021] [Accepted: 08/11/2021] [Indexed: 01/03/2023]
Abstract
Reciprocal pathways connecting the cerebellum to the prefrontal cortex provide a biological and functional substrate to modulate cognitive functions. Dysfunction of both medial prefrontal cortex (mPFC) and cerebellum underlie the phenotypes of several neuropsychiatric disorders that exhibit comorbidity with substance use disorder (SUD). In people with SUD, cue-action-reward associations appears to be particularly strong and salient, acting as powerful motivational triggers for craving and relapse. Studies of cue reactivity in human with SUD have shown cerebellar activations when drug-related cues are presented. Our preclinical research showed that cocaine-induced conditioned preference increases neural activity and upregulates perineuronal nets (PNNs) around Golgi interneurons in the posterior cerebellar cortex. In the present investigation, we aimed at evaluating cerebellar signatures of conditioned preference for cocaine when drug learning is established under mPFC impairment. We used lidocaine to temporarily inactivate in male rats either the Prelimbic (PL) or the Infralimbic (IL) cortices during cocaine-induced conditioning. The inactivation of the IL, but not the PL, encouraged the acquisition of preference for cocaine-related cues, increased posterior cerebellar cortex activity, and upregulated the expression of PNNs around Golgi interneurons. Moreover, IL impairment not only increased vGluT2- and vGAT-related activity around Golgi cells but also regulated PNNs differently on subpopulations of Golgi cells, increasing the number of neurogranin+ PNN-expressing Golgi cells. Our findings suggest that IL dysfunction may facilitate the acquisition of cocaine-induced memory and cerebellar drug-related learning hallmarks. Overall, IL perturbation during cocaine-induced Pavlovian learning increased cerebellar activity and drug effects. Importantly, cerebellum involvement requires a contingent experience with the drug, and it is not the effect of a mere inactivation of IL cortex.
Collapse
Affiliation(s)
- Julian Guarque-Chabrera
- Área de Psicobiología, Universitat Jaume I, Castellón de la Plana, Comunitat Valenciana 12071, Spain.
| | - Isis Gil-Miravet
- Área de Psicobiología, Universitat Jaume I, Castellón de la Plana, Comunitat Valenciana 12071, Spain.
| | | | - Ignasi Melchor-Eixea
- Área de Psicobiología, Universitat Jaume I, Castellón de la Plana, Comunitat Valenciana 12071, Spain.
| | - Marta Miquel
- Área de Psicobiología, Universitat Jaume I, Castellón de la Plana, Comunitat Valenciana 12071, Spain.
| |
Collapse
|
43
|
Nett KE, LaLumiere RT. Infralimbic cortex functioning across motivated behaviors: Can the differences be reconciled? Neurosci Biobehav Rev 2021; 131:704-721. [PMID: 34624366 PMCID: PMC8642304 DOI: 10.1016/j.neubiorev.2021.10.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 09/10/2021] [Accepted: 10/02/2021] [Indexed: 10/20/2022]
Abstract
The rodent infralimbic cortex (IL) is implicated in higher order executive functions such as reward seeking and flexible decision making. However, the precise nature of its role in these processes is unclear. Early evidence indicated that the IL promotes the extinction and ongoing inhibition of fear conditioning and cocaine seeking. However, evidence spanning other behavioral domains, such as natural reward seeking and habit-based learning, suggests a more nuanced understanding of IL function. As techniques have advanced and more studies have examined IL function, identifying a unifying explanation for its behavioral function has become increasingly difficult. Here, we discuss evidence of IL function across motivated behaviors, including associative learning, drug seeking, natural reward seeking, and goal-directed versus habit-based behaviors, and emphasize how context-specific encoding and heterogeneous IL neuronal populations may underlie seemingly conflicting findings in the literature. Together, the evidence suggests that a major IL function is to facilitate the encoding and updating of contingencies between cues and behaviors to guide subsequent behaviors.
Collapse
Affiliation(s)
- Kelle E Nett
- Interdisciplinary Neuroscience Program, University of Iowa, Iowa City, IA 52242, United States.
| | - Ryan T LaLumiere
- Interdisciplinary Neuroscience Program, University of Iowa, Iowa City, IA 52242, United States; Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA 52242, United States; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, United States
| |
Collapse
|
44
|
Madangopal R, Ramsey LA, Weber SJ, Brenner MB, Lennon VA, Drake OR, Komer LE, Tunstall BJ, Bossert JM, Shaham Y, Hope BT. Inactivation of the infralimbic cortex decreases discriminative stimulus-controlled relapse to cocaine seeking in rats. Neuropsychopharmacology 2021; 46:1969-1980. [PMID: 34162997 PMCID: PMC8429767 DOI: 10.1038/s41386-021-01067-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 06/07/2021] [Accepted: 06/09/2021] [Indexed: 01/13/2023]
Abstract
Persistent susceptibility to cue-induced relapse is a cardinal feature of addiction. Discriminative stimuli (DSs) are one type of drug-associated cue that signal drug availability (DS+) or unavailability (DS-) and control drug seeking prior to relapse. We previously established a trial-based procedure in rats to isolate DSs from context, conditioned stimuli, and other drug-associated cues during cocaine self-administration and demonstrated DS-controlled cocaine seeking up to 300 abstinence days. The behavioral and neural mechanisms underlying trial-based DS-control of drug seeking have rarely been investigated. Here we show that following discrimination training in our trial-based procedure, the DS+ and DS- independently control the expression and suppression of cocaine seeking during abstinence. Using microinjections of GABAA + GABAB receptor agonists (muscimol + baclofen) in medial prefrontal cortex, we report that infralimbic, but not prelimbic, subregion of medial prefrontal cortex is critical to persistent DS-controlled relapse to cocaine seeking after prolonged abstinence, but not DS-guided discriminated cocaine seeking or DS-controlled cocaine self-admininstration. Finally, using ex vivo whole-cell recordings from pyramidal neurons in the medial prefrontal cortex, we demonstrate that the disruption of DS-controlled cocaine seeking following infralimbic cortex microinjections of muscimol+baclofen is likely a result of suppression of synaptic transmission in the region via a presynaptic mechanism of action.
Collapse
Affiliation(s)
- Rajtarun Madangopal
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Leslie A Ramsey
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Sophia J Weber
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Megan B Brenner
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Veronica A Lennon
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Olivia R Drake
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Lauren E Komer
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, USA
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Brendan J Tunstall
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, USA
- Department of Pharmacology, Addiction Science, and Toxicology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Jennifer M Bossert
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Yavin Shaham
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Bruce T Hope
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, USA.
| |
Collapse
|
45
|
Heinsbroek JA, Giannotti G, Mandel MR, Josey M, Aston-Jones G, James MH, Peters J. A common limiter circuit for opioid choice and relapse identified in a rodent addiction model. Nat Commun 2021; 12:4788. [PMID: 34373454 PMCID: PMC8352904 DOI: 10.1038/s41467-021-25080-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 07/19/2021] [Indexed: 12/16/2022] Open
Abstract
Activity in numerous brain regions drives heroin seeking, but no circuits that limit heroin seeking have been identified. Furthermore, the neural circuits controlling opioid choice are unknown. In this study, we examined the role of the infralimbic cortex (IL) to nucleus accumbens shell (NAshell) pathway during heroin choice and relapse. This model yielded subpopulations of heroin versus food preferring rats during choice, and choice was unrelated to subsequent relapse rates to heroin versus food cues, suggesting that choice and relapse are distinct behavioral constructs. Supporting this, inactivation of the IL with muscimol produced differential effects on opioid choice versus relapse. A pathway-specific chemogenetic approach revealed, however, that the IL-NAshell pathway acts as a common limiter of opioid choice and relapse. Furthermore, dendritic spines in IL-NAshell neurons encode distinct aspects of heroin versus food reinforcement. Thus, opioid choice and relapse share a common addiction-limiting circuit in the IL-NAshell pathway.
Collapse
Affiliation(s)
- Jasper A Heinsbroek
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Giuseppe Giannotti
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Mitchel R Mandel
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Megan Josey
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Gary Aston-Jones
- Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA
| | - Morgan H James
- Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA.,Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Jamie Peters
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA. .,Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
46
|
Joo YH, Kim JH, Kim HK, Son YD, Cumming P, Kim JH. Functional Analysis of Brain Imaging Suggests Changes in the Availability of mGluR5 and Altered Connectivity in the Cerebral Cortex of Long-Term Abstaining Males with Alcohol Dependence: A Preliminary Study. Life (Basel) 2021; 11:life11060506. [PMID: 34070900 PMCID: PMC8228527 DOI: 10.3390/life11060506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/23/2021] [Accepted: 05/27/2021] [Indexed: 11/16/2022] Open
Abstract
Direct in vivo evidence of altered metabotropic glutamate receptor-5 (mGluR5) availability in alcohol-related disorders is lacking. We performed [11C]ABP688 positron emission tomography (PET) and resting-state functional magnetic resonance imaging (rs-fMRI) in prolonged abstinent subjects with alcohol dependence to examine alterations of mGluR5 availability, and to investigate their functional significance relating to neural systems-level changes. Twelve prolonged abstinent male subjects with alcohol dependence (median abstinence duration: six months) and ten healthy male controls underwent [11C]ABP688 PET imaging and 3-Tesla MRI. For mGluR5 availability, binding potential (BPND) was calculated using the simplified reference tissue model with cerebellar gray matter as the reference region. The initial region-of-interest (ROI)-based analysis yielded no significant group differences in mGluR5 availability. The voxel-based analysis revealed significantly lower [11C]ABP688 BPND in the middle temporal and inferior parietal cortices, and higher BPND in the superior temporal cortex in the alcohol dependence group compared with controls. Functional connectivity analysis of the rs-fMRI data employed seed regions identified from the quantitative [11C]ABP688 PET analysis, which revealed significantly altered functional connectivity from the inferior parietal cortex seed to the occipital pole and dorsal visual cortex in the alcohol dependence group compared with the control group. To our knowledge, this is the first report on the combined analysis of mGluR5 PET imaging and rs-fMRI in subjects with alcohol dependence. These preliminary results suggest the possibility of region-specific alterations of mGluR5 availability in vivo and related functional connectivity perturbations in prolonged abstinent subjects.
Collapse
Affiliation(s)
- Yo-Han Joo
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (Y.-H.J.); (J.-H.K.); (H.-K.K.)
| | - Jeong-Hee Kim
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (Y.-H.J.); (J.-H.K.); (H.-K.K.)
- Department of Biomedical Engineering, College of Health Science, Gachon University, Incheon 21936, Korea
| | - Hang-Keun Kim
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (Y.-H.J.); (J.-H.K.); (H.-K.K.)
- Department of Biomedical Engineering, College of Health Science, Gachon University, Incheon 21936, Korea
- Gachon Advanced Institute for Health Science and Technology, Graduate School, Gachon University, Incheon 21565, Korea
| | - Young-Don Son
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (Y.-H.J.); (J.-H.K.); (H.-K.K.)
- Department of Biomedical Engineering, College of Health Science, Gachon University, Incheon 21936, Korea
- Gachon Advanced Institute for Health Science and Technology, Graduate School, Gachon University, Incheon 21565, Korea
- Correspondence: (Y.-D.S.); or (J.-H.K.); Tel.: +82-32-820-4416 (Y.-D.S.); +82-32-460-2696 (J.-H.K.)
| | - Paul Cumming
- Department of Nuclear Medicine, Inselspital, University of Bern, CH-3010 Bern, Switzerland;
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Jong-Hoon Kim
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (Y.-H.J.); (J.-H.K.); (H.-K.K.)
- Gachon Advanced Institute for Health Science and Technology, Graduate School, Gachon University, Incheon 21565, Korea
- Gil Medical Center, Department of Psychiatry, Gachon University College of Medicine, Gachon University, Incheon 21565, Korea
- Correspondence: (Y.-D.S.); or (J.-H.K.); Tel.: +82-32-820-4416 (Y.-D.S.); +82-32-460-2696 (J.-H.K.)
| |
Collapse
|
47
|
Kane L, Venniro M, Quintana‐Feliciano R, Madangopal R, Rubio FJ, Bossert JM, Caprioli D, Shaham Y, Hope BT, Warren BL. Fos-expressing neuronal ensemble in rat ventromedial prefrontal cortex encodes cocaine seeking but not food seeking in rats. Addict Biol 2021; 26:e12943. [PMID: 32683756 DOI: 10.1111/adb.12943] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/02/2020] [Accepted: 07/07/2020] [Indexed: 12/30/2022]
Abstract
Neuronal ensembles in ventromedial prefrontal cortex (vmPFC) play a role in both cocaine and palatable food seeking. However, it is unknown whether similar or different vmPFC neuronal ensembles mediate food and cocaine seeking. Here, we used the Daun02 inactivation procedure to assess whether the neuronal ensembles mediating food and cocaine seeking can be functionally distinguished. We trained male and female Fos-LacZ rats to self-administer palatable food pellets and cocaine on alternating days for 18 days. We then exposed the rats to a brief nonreinforced food- or cocaine-seeking test to induce Fos and β-gal in neuronal ensembles associated with food or cocaine seeking, respectively and infused Daun02 into vmPFC to ablate the β-gal-expressing ensembles. Two days later, we tested the rats for food or cocaine seeking under extinction conditions. Although inactivation of the food-seeking ensemble did not influence food or cocaine seeking, inactivation of the cocaine-seeking ensemble reduced cocaine seeking but not food seeking. Results indicate that the neuronal ensemble activated by cocaine seeking in vmPFC is functionally separate from the ensemble activated by food seeking.
Collapse
Affiliation(s)
- Louisa Kane
- Behavioral Neuroscience Branch IRP/NIDA/NIH/DHHS Baltimore Maryland USA
| | - Marco Venniro
- Behavioral Neuroscience Branch IRP/NIDA/NIH/DHHS Baltimore Maryland USA
| | - Richard Quintana‐Feliciano
- Behavioral Neuroscience Branch IRP/NIDA/NIH/DHHS Baltimore Maryland USA
- Department of Pharmacodynamics University of Florida Gainesville Florida USA
| | | | - F. Javier Rubio
- Behavioral Neuroscience Branch IRP/NIDA/NIH/DHHS Baltimore Maryland USA
| | | | - Daniele Caprioli
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia) Rome Italy
- Department of Physiology and Pharmacology Sapienza University of Rome Rome Italy
| | - Yavin Shaham
- Behavioral Neuroscience Branch IRP/NIDA/NIH/DHHS Baltimore Maryland USA
| | - Bruce T. Hope
- Behavioral Neuroscience Branch IRP/NIDA/NIH/DHHS Baltimore Maryland USA
| | - Brandon L. Warren
- Behavioral Neuroscience Branch IRP/NIDA/NIH/DHHS Baltimore Maryland USA
- Department of Pharmacodynamics University of Florida Gainesville Florida USA
| |
Collapse
|
48
|
Li M, Cabrera-Garcia D, Salling MC, Au E, Yang G, Harrison NL. Alcohol reduces the activity of somatostatin interneurons in the mouse prefrontal cortex: A neural basis for its disinhibitory effect? Neuropharmacology 2021; 188:108501. [PMID: 33636191 DOI: 10.1016/j.neuropharm.2021.108501] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/01/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022]
Abstract
The prefrontal cortex (PFC) is involved in executive ("top-down") control of behavior and its function is especially susceptible to the effects of alcohol, leading to behavioral disinhibition that is associated with alterations in decision making, response inhibition, social anxiety and working memory. The circuitry of the PFC involves a complex interplay between pyramidal neurons (PNs) and several subclasses of inhibitory interneurons (INs), including somatostatin (SST)-expressing INs. Using in vivo calcium imaging, we showed that alcohol dose-dependently altered network activity in layers 2/3 of the prelimbic subregion of the mouse PFC. Low doses of alcohol (1 g/kg, intraperitoneal, i.p.) caused moderate activation of SST INs and weak inhibition of PNs. At moderate to high doses, alcohol (2-3 g/kg) strongly inhibited the activity of SST INs in vivo, and this effect may result in disinhibition, as the activity of a subpopulation of PNs was simultaneously enhanced. In contrast, recordings in brain slices using ex vivo electrophysiology revealed no direct effect of alcohol on the excitability of either SST INs or PNs over a range of concentrations (20 and 50 mM) consistent with the blood alcohol levels reached in the in vivo experiments. This dose-dependent effect of alcohol on SST INs in vivo may reveal a neural basis for the disinhibitory effect of alcohol in the PFC mediated by other neurons within or external to the PFC circuitry.
Collapse
Affiliation(s)
- Miao Li
- Columbia University, Department of Anesthesiology, 630 West 168th Street, New York, NY, 10032, USA
| | - David Cabrera-Garcia
- Columbia University, Department of Anesthesiology, 630 West 168th Street, New York, NY, 10032, USA
| | - Michael C Salling
- Louisiana State University, Department of Anatomy, New Orleans, LA, 1901 Perdido Street, New Orleans, LA, 70112, USA
| | - Edmund Au
- Columbia University, Department of Pathology & Cell Biology and Rehabilitative Medicine and Regeneration, Columbia Translational Neuroscience Initiative Scholar, 630 West 168th Street, New York, NY, 10032, USA
| | - Guang Yang
- Columbia University, Department of Anesthesiology, 630 West 168th Street, New York, NY, 10032, USA.
| | - Neil L Harrison
- Columbia University, Department of Anesthesiology, 630 West 168th Street, New York, NY, 10032, USA; Columbia University, Department of Molecular Pharmacology and Therapeutics, 630 West 168th Street, New York, NY, 10032, USA.
| |
Collapse
|
49
|
Salery M, Godino A, Nestler EJ. Drug-activated cells: From immediate early genes to neuronal ensembles in addiction. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 90:173-216. [PMID: 33706932 DOI: 10.1016/bs.apha.2020.09.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Beyond their rapid rewarding effects, drugs of abuse can durably alter an individual's response to their environment as illustrated by the compulsive drug seeking and risk of relapse triggered by drug-associated stimuli. The persistence of these associations even long after cessation of drug use demonstrates the enduring mark left by drugs on brain reward circuits. However, within these circuits, neuronal populations are differently affected by drug exposure and growing evidence indicates that relatively small subsets of neurons might be involved in the encoding and expression of drug-mediated associations. The identification of sparse neuronal populations recruited in response to drug exposure has benefited greatly from the study of immediate early genes (IEGs) whose induction is critical in initiating plasticity programs in recently activated neurons. In particular, the development of technologies to manipulate IEG-expressing cells has been fundamental to implicate broadly distributed neuronal ensembles coincidently activated by either drugs or drug-associated stimuli and to then causally establish their involvement in drug responses. In this review, we summarize the literature regarding IEG regulation in different learning paradigms and addiction models to highlight their role as a marker of activity and plasticity. As the exploration of neuronal ensembles in addiction improves our understanding of drug-associated memory encoding, it also raises several questions regarding the cellular and molecular characteristics of these discrete neuronal populations as they become incorporated in drug-associated neuronal ensembles. We review recent efforts towards this goal and discuss how they will offer a more comprehensive understanding of addiction pathophysiology.
Collapse
Affiliation(s)
- Marine Salery
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Arthur Godino
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
50
|
Wandres M, Pfarr S, Molnár B, Schöllkopf U, Ercsey-Ravasz M, Sommer WH, Körber C. Alcohol and sweet reward are encoded by distinct meta-ensembles. Neuropharmacology 2021; 195:108496. [PMID: 33582149 DOI: 10.1016/j.neuropharm.2021.108496] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/30/2021] [Accepted: 02/08/2021] [Indexed: 01/14/2023]
Abstract
Cue-reward associations form distinct memories that can drive appetitive behaviors and cravings for both drugs and natural rewards. It is still unclear how such memories are encoded in the brain's reward system. We trained rats to concurrently self-administer either alcohol or a sweet saccharin solution as drug or natural rewards, respectively. Memory recall due to cue exposure reactivated reward-associated functional ensembles in reward-related brain regions, marked by a neural cFos response. While the local ensembles activated by cue presentation for either reward consisted of similar numbers of neurons, using advanced statistical network theory, we found robust reward-specific co-activation patterns across brain regions. Interestingly, the resulting meta-ensemble networks differed by the most influential regions, which in case of saccharin comprised the prefrontal cortex, while for alcohol seeking control shifted to insular cortex with strong involvement of the amygdala. Our results support the view of memory representation as a differential co-activation of local neuronal ensembles. This article is part of the special issue on 'Neurocircuitry Modulating Drug and Alcohol Abuse'.
Collapse
Affiliation(s)
- Miriam Wandres
- Institute of Anatomy and Cell Biology, Department of Functional Neuroanatomy, Heidelberg University, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany
| | - Simone Pfarr
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany
| | - Botond Molnár
- Faculty of Mathematics and Informatics, Babeş-Bolyai University, Cluj-Napoca, Romania; Faculty of Physics, Babeş-Bolyai University, Cluj-Napoca, Romania; Transylvanian Institute of Neuroscience, Network Science Lab, Cluj-Napoca, Romania
| | - Ursula Schöllkopf
- Institute of Anatomy and Cell Biology, Department of Functional Neuroanatomy, Heidelberg University, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany
| | - Maria Ercsey-Ravasz
- Faculty of Physics, Babeş-Bolyai University, Cluj-Napoca, Romania; Transylvanian Institute of Neuroscience, Network Science Lab, Cluj-Napoca, Romania
| | - Wolfgang H Sommer
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany; Department of Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany.
| | - Christoph Körber
- Institute of Anatomy and Cell Biology, Department of Functional Neuroanatomy, Heidelberg University, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany.
| |
Collapse
|