51
|
Kumaresan V, Lim Y, Juneja P, Tipton AE, de Guglielmo G, Carrette LLG, Kallupi M, Maturin L, Liu Y, George O, Zhang H. Abstinence from Escalation of Cocaine Intake Changes the microRNA Landscape in the Cortico-Accumbal Pathway. Biomedicines 2023; 11:1368. [PMID: 37239038 PMCID: PMC10216163 DOI: 10.3390/biomedicines11051368] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 05/28/2023] Open
Abstract
Cocaine administration alters the microRNA (miRNA) landscape in the cortico-accumbal pathway. These changes in miRNA can play a major role in the posttranscriptional regulation of gene expression during withdrawal. This study aimed to investigate the changes in microRNA expression in the cortico-accumbal pathway during acute withdrawal and protracted abstinence following escalated cocaine intake. Small RNA sequencing (sRNA-seq) was used to profile miRNA transcriptomic changes in the cortico-accumbal pathway [infralimbic- and prelimbic-prefrontal cortex (IL and PL) and nucleus accumbens (NAc)] of rats with extended access to cocaine self-administration followed by an 18-h withdrawal or a 4-week abstinence. An 18-h withdrawal led to differential expression (fold-change > 1.5 and p < 0.05) of 21 miRNAs in the IL, 18 miRNAs in the PL, and two miRNAs in the NAc. The mRNAs potentially targeted by these miRNAs were enriched in the following pathways: gap junctions, neurotrophin signaling, MAPK signaling, and cocaine addiction. Moreover, a 4-week abstinence led to differential expression (fold-change > 1.5 and p < 0.05) of 23 miRNAs in the IL, seven in the PL, and five miRNAs in the NAc. The mRNAs potentially targeted by these miRNAs were enriched in pathways including gap junctions, cocaine addiction, MAPK signaling, glutamatergic synapse, morphine addiction, and amphetamine addiction. Additionally, the expression levels of several miRNAs differentially expressed in either the IL or the NAc were significantly correlated with addiction behaviors. Our findings highlight the impact of acute and protracted abstinence from escalated cocaine intake on miRNA expression in the cortico-accumbal pathway, a key circuit in addiction, and suggest developing novel biomarkers and therapeutic approaches to prevent relapse by targeting abstinence-associated miRNAs and their regulated mRNAs.
Collapse
Affiliation(s)
- Vidhya Kumaresan
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA;
| | - Yolpanhchana Lim
- Department of Psychiatry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA; (Y.L.); (P.J.); (Y.L.)
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| | - Poorva Juneja
- Department of Psychiatry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA; (Y.L.); (P.J.); (Y.L.)
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| | - Allison E. Tipton
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA;
| | - Giordano de Guglielmo
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; (G.d.G.); (L.L.G.C.); (M.K.); (L.M.); (O.G.)
| | - Lieselot L. G. Carrette
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; (G.d.G.); (L.L.G.C.); (M.K.); (L.M.); (O.G.)
| | - Marsida Kallupi
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; (G.d.G.); (L.L.G.C.); (M.K.); (L.M.); (O.G.)
| | - Lisa Maturin
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; (G.d.G.); (L.L.G.C.); (M.K.); (L.M.); (O.G.)
| | - Ying Liu
- Department of Psychiatry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA; (Y.L.); (P.J.); (Y.L.)
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| | - Olivier George
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; (G.d.G.); (L.L.G.C.); (M.K.); (L.M.); (O.G.)
| | - Huiping Zhang
- Department of Psychiatry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA; (Y.L.); (P.J.); (Y.L.)
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
52
|
Lewandowski SI, Hodebourg R, Wood SK, Carter JS, Nelson KH, Kalivas PW, Reichel CM. Matrix metalloproteinase activity during methamphetamine cued relapse. Addict Biol 2023; 28:e13279. [PMID: 37186441 PMCID: PMC10506177 DOI: 10.1111/adb.13279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/23/2023] [Accepted: 03/27/2023] [Indexed: 05/17/2023]
Abstract
Relapse to drug seeking involves transient synaptic remodelling that occurs in response to drug-associated cues. This remodelling includes activation of matrix metalloproteinases (MMPs) to initiate catalytic signalling in the extracellular matrix in the nucleus accumbens core (NAcore). We hypothesized that MMP activity would be increased in the NAcore during cue-induced methamphetamine (meth) seeking in a rat model of meth use and relapse. Male and female rats had indwelling jugular catheters and bilateral intracranial cannula targeting the NAcore surgically implanted. Following recovery, rats underwent meth or saline self-administration (6 h/day for 15 days) in which active lever responding was paired with a light + tone stimulus complex, followed by home cage abstinence. Testing occurred after 7 or 30 days of abstinence. On test day, rats were microinjected with a fluorescein isothiocyanate (FITC)-quenched gelatin substrate that fluoresces following cleavage by MMP-2,9, allowing for the quantification of gelatinase activity during cued-relapse testing. MMP-2,9 activity was significantly increased in the NAcore by meth cues presentation after 7 and 30 days of abstinence, indicating that remodelling by MMPs occurs during presentation of meth associated cues. Surprisingly, although cue-induced seeking increased between Days 7 and 30, MMP-2,9 activity did not increase. These findings indicate that although MMP activation is elicited during meth cue-induced seeking, MMP activation did not parallel the meth seeking that occurs during extended drug abstinence.
Collapse
Affiliation(s)
- Stacia I. Lewandowski
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - Ritchy Hodebourg
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - Samuel K. Wood
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - Jordan S. Carter
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - Katherine H. Nelson
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - Peter W. Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - Carmela M. Reichel
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| |
Collapse
|
53
|
Zanda MT, Floris G, Daws SE. Orbitofrontal cortex microRNAs support long-lasting heroin seeking behavior in male rats. Transl Psychiatry 2023; 13:117. [PMID: 37031193 PMCID: PMC10082780 DOI: 10.1038/s41398-023-02423-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 03/23/2023] [Accepted: 03/29/2023] [Indexed: 04/10/2023] Open
Abstract
Recovery from opioid use disorder (OUD) and maintenance of abstinence from opioid use is hampered by perseverant drug cravings that may persist for months after cessation of drug use. Drug cravings can intensify during the abstinence period, a phenomenon referred to as the 'incubation of craving' that has been well-described in preclinical studies. We previously reported that animals that self-administered heroin at a dosage of 0.075 mg/kg/infusion (HH) paired with discrete drug cues displayed robust incubation of heroin craving behavior after 21 days (D) of forced abstinence, an effect that was not observed with a lower dosage (0.03 mg/kg/infusion; HL). Here, we sought to elucidate molecular mechanisms underlying long-term heroin seeking behavior by profiling microRNA (miRNA) pathways in the orbitofrontal cortex (OFC), a brain region that modulates incubation of heroin seeking. miRNAs are small noncoding RNAs with long half-lives that have emerged as critical regulators of drug seeking behavior but their expression in the OFC has not been examined in any drug exposure paradigm. We employed next generation sequencing to detect OFC miRNAs differentially expressed after 21D of forced abstinence between HH and HL animals, and proteomics analysis to elucidate miRNA-dependent translational neuroadaptations. We identified 55 OFC miRNAs associated with incubation of heroin craving, including miR-485-5p, which was significantly downregulated following 21D forced abstinence in HH but not HL animals. We bidirectionally manipulated miR-485-5p in the OFC to demonstrate that miR-485-5p can regulate long-lasting heroin seeking behavior after extended forced abstinence. Proteomics analysis identified 45 proteins selectively regulated in the OFC of HH but not HL animals that underwent 21D forced abstinence, of which 7 were putative miR-485-5p target genes. Thus, the miR-485-5p pathway is dysregulated in animals with a phenotype of persistent heroin craving behavior and OFC miR-485-5p pathways may function to support long-lasting heroin seeking.
Collapse
Affiliation(s)
- Mary Tresa Zanda
- Center for Substance Abuse Research, Temple University, Philadelphia, PA, USA
- Department of Neural Sciences, Temple University, Philadelphia, PA, USA
| | - Gabriele Floris
- Center for Substance Abuse Research, Temple University, Philadelphia, PA, USA
- Department of Neural Sciences, Temple University, Philadelphia, PA, USA
| | - Stephanie E Daws
- Center for Substance Abuse Research, Temple University, Philadelphia, PA, USA.
- Department of Neural Sciences, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
54
|
Brown AR, Branthwaite HE, Farahbakhsh ZZ, Mukerjee S, Melugin PR, Song K, Noamany H, Siciliano CA. Structured tracking of alcohol reinforcement (STAR) for basic and translational alcohol research. Mol Psychiatry 2023; 28:1585-1598. [PMID: 36849824 PMCID: PMC10208967 DOI: 10.1038/s41380-023-01994-4] [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: 01/05/2022] [Revised: 01/26/2023] [Accepted: 02/07/2023] [Indexed: 03/01/2023]
Abstract
There is inherent tension between methodologies developed to address basic research questions in model species and those intended for preclinical to clinical translation: basic investigations require flexibility of experimental design as hypotheses are rapidly tested and revised, whereas preclinical models emphasize standardized protocols and specific outcome measures. This dichotomy is particularly relevant in alcohol research, which spans a diverse range of basic sciences in addition to intensive efforts towards understanding the pathophysiology of alcohol use disorder (AUD). To advance these goals there is a great need for approaches that facilitate synergy across basic and translational areas of nonhuman alcohol research. In male and female mice, we establish a modular alcohol reinforcement paradigm: Structured Tracking of Alcohol Reinforcement (STAR). STAR provides a robust platform for quantitative assessment of AUD-relevant behavioral domains within a flexible framework that allows direct crosstalk between translational and mechanistically oriented studies. To achieve cross-study integration, despite disparate task parameters, a straightforward multivariate phenotyping analysis is used to classify subjects based on propensity for heightened alcohol consumption and insensitivity to punishment. Combining STAR with extant preclinical alcohol models, we delineate longitudinal phenotype dynamics and reveal putative neuro-biomarkers of heightened alcohol use vulnerability via neurochemical profiling of cortical and brainstem tissues. Together, STAR allows quantification of time-resolved biobehavioral processes essential for basic research questions simultaneous with longitudinal phenotyping of clinically relevant outcomes, thereby providing a framework to facilitate cohesion and translation in alcohol research.
Collapse
Affiliation(s)
- Alex R Brown
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Hannah E Branthwaite
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Zahra Z Farahbakhsh
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Snigdha Mukerjee
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Patrick R Melugin
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Keaton Song
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Habiba Noamany
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Cody A Siciliano
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
55
|
Towers EB, Williams IL, Qillawala EI, Rissman EF, Lynch WJ. Sex/Gender Differences in the Time-Course for the Development of Substance Use Disorder: A Focus on the Telescoping Effect. Pharmacol Rev 2023; 75:217-249. [PMID: 36781217 PMCID: PMC9969523 DOI: 10.1124/pharmrev.121.000361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 10/05/2022] [Accepted: 10/25/2022] [Indexed: 12/14/2022] Open
Abstract
Sex/gender effects have been demonstrated for multiple aspects of addiction, with one of the most commonly cited examples being the "telescoping effect" where women meet criteria and/or seek treatment of substance use disorder (SUD) after fewer years of drug use as compared with men. This phenomenon has been reported for multiple drug classes including opioids, psychostimulants, alcohol, and cannabis, as well as nonpharmacological addictions, such as gambling. However, there are some inconsistent reports that show either no difference between men and women or opposite effects and a faster course to addiction in men than women. Thus, the goals of this review are to evaluate evidence for and against the telescoping effect in women and to determine the conditions/populations for which the telescoping effect is most relevant. We also discuss evidence from preclinical studies, which strongly support the validity of the telescoping effect and show that female animals develop addiction-like features (e.g., compulsive drug use, an enhanced motivation for the drug, and enhanced drug-craving/vulnerability to relapse) more readily than male animals. We also discuss biologic factors that may contribute to the telescoping effect, such as ovarian hormones, and its neurobiological basis focusing on the mesolimbic dopamine reward pathway and the corticomesolimbic glutamatergic pathway considering the critical roles these pathways play in the rewarding/reinforcing effects of addictive drugs and SUD. We conclude with future research directions, including intervention strategies to prevent the development of SUD in women. SIGNIFICANCE STATEMENT: One of the most widely cited gender/sex differences in substance use disorder (SUD) is the "telescoping effect," which reflects an accelerated course in women versus men for the development and/or seeking treatment for SUD. This review evaluates evidence for and against a telescoping effect drawing upon data from both clinical and preclinical studies. We also discuss the contribution of biological factors and underlying neurobiological mechanisms and highlight potential targets to prevent the development of SUD in women.
Collapse
Affiliation(s)
- Eleanor Blair Towers
- Psychiatry and Neurobehavioral Sciences (E.B.T., I.L.W., E.I.Q., W.J.L.) and Medical Scientist Training Program (E.B.T.), University of Virginia, Charlottesville, Virginia, and Center for Human Health and the Environment and Program in Genetics, North Carolina State University, Raleigh, North Carolina (E.F.R.)
| | - Ivy L Williams
- Psychiatry and Neurobehavioral Sciences (E.B.T., I.L.W., E.I.Q., W.J.L.) and Medical Scientist Training Program (E.B.T.), University of Virginia, Charlottesville, Virginia, and Center for Human Health and the Environment and Program in Genetics, North Carolina State University, Raleigh, North Carolina (E.F.R.)
| | - Emaan I Qillawala
- Psychiatry and Neurobehavioral Sciences (E.B.T., I.L.W., E.I.Q., W.J.L.) and Medical Scientist Training Program (E.B.T.), University of Virginia, Charlottesville, Virginia, and Center for Human Health and the Environment and Program in Genetics, North Carolina State University, Raleigh, North Carolina (E.F.R.)
| | - Emilie F Rissman
- Psychiatry and Neurobehavioral Sciences (E.B.T., I.L.W., E.I.Q., W.J.L.) and Medical Scientist Training Program (E.B.T.), University of Virginia, Charlottesville, Virginia, and Center for Human Health and the Environment and Program in Genetics, North Carolina State University, Raleigh, North Carolina (E.F.R.)
| | - Wendy J Lynch
- Psychiatry and Neurobehavioral Sciences (E.B.T., I.L.W., E.I.Q., W.J.L.) and Medical Scientist Training Program (E.B.T.), University of Virginia, Charlottesville, Virginia, and Center for Human Health and the Environment and Program in Genetics, North Carolina State University, Raleigh, North Carolina (E.F.R.)
| |
Collapse
|
56
|
Namba MD, Phillips MN, Chen PJ, Blass BE, Olive MF, Neisewander JL. HIV gp120 impairs nucleus accumbens neuroimmune function and dopamine D3 receptor-mediated inhibition of cocaine seeking in male rats. ADDICTION NEUROSCIENCE 2023; 5:100062. [PMID: 36909738 PMCID: PMC9997483 DOI: 10.1016/j.addicn.2023.100062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Cocaine Use Disorders (CUDs) are associated with an increased risk of human immunodeficiency virus (HIV) infection. Cocaine and the HIV envelope protein gp120 each induce distinct deficits to mesocorticolimbic circuit function and motivated behavior; however, little is known regarding how they interact to dysregulate these functions or how such interactions impact pharmacotherapeutic efficacy. We have previously shown that the selective, weak partial agonist of the dopamine D3 receptor (D3R), MC-25-41, attenuates cocaine-seeking behavior in male rats. Here, we sought to characterize changes in striatal neuroimmune function in gp120-exposed rats across abstinence from operant access to cocaine (0.75 mg/kg, i.v.) or sucrose (45 mg/pellet), and to examine the impact of gp120 exposure on MC-25-41-reduced cocaine seeking. After establishing a history of cocaine or sucrose self-administration, rats received intracerebroventricular gp120 infusions daily the first 5 days of abstinence and were sacrificed either on day 6 or after 21 days of forced abstinence and a cue-induced cocaine seeking test. We demonstrated that MC-25-41 treatment attenuated cue-induced cocaine seeking among control rats but not gp120-exposed rats. Moreover, postmortem analysis of nucleus accumbens (NAc) core neuroimmune function indicated cocaine abstinence- and gp120-induced impairments, and the expression of several immune factors within the NAc core significantly correlated with cocaine-seeking behavior. We conclude that cocaine abstinence dysregulates striatal neuroimmune function and interacts with gp120 to inhibit the effectiveness of a D3R partial agonist in reducing cocaine seeking. These findings highlight the need to consider comorbidities, such as immune status, when evaluating the efficacy of novel pharmacotherapeutics.
Collapse
Affiliation(s)
- Mark D Namba
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Megan N Phillips
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Peng-Jen Chen
- Department of Pharmaceutical Sciences, Moulder Center for Drug Discovery Research, School of Pharmacy, Temple University, Philadelphia, PA, USA
| | - Benjamin E Blass
- Department of Pharmaceutical Sciences, Moulder Center for Drug Discovery Research, School of Pharmacy, Temple University, Philadelphia, PA, USA
| | - M Foster Olive
- Department of Psychology, Arizona State University, Tempe, AZ, USA
| | | |
Collapse
|
57
|
Borges C, Inigo F, Quteishat N, Charles J, Ah-Yen E, U S. Acute food deprivation-induced relapse to heroin seeking after short and long punishment-imposed abstinence in male rats. Psychopharmacology (Berl) 2023; 240:595-607. [PMID: 35951079 DOI: 10.1007/s00213-022-06207-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/28/2022] [Indexed: 11/24/2022]
Abstract
RATIONAL Stress is a major trigger for drug relapse in humans and animal models, even after prolonged abstinence. However, animal models for stress-induced relapse were criticized for the lack of predictive and face validity. OBJECTIVES Here we investigated the effect of acute food deprivation stress in a novel stress-induced relapse model using voluntary, punishment-imposed abstinence from heroin. We also performed a detailed characterization of the development of punishment-imposed abstinence. METHODS Male rats were trained to self-administered heroin (0.1 mg/kg/infusion) for 2 weeks, using the seeking-taking chained schedule. Pressing the 'seeking' lever led to the insertion of the 'taking' lever and pressing the take lever resulted in heroin infusion. Following self-administration training, rats were exposed to 8 or 21 days of heroin-seeking punishment. During punishment, 30% of the completed seek links resulted in a mild escalating footshock instead of take lever presentation. Next, rats were tested for heroin seeking under extinction conditions after 24 h of food deprivation and sated conditions. RESULTS Probabilistic punishment of seeking lever responses resulted in gradual suppression of heroin seeking and taking. Exposure to food-deprivation stress induced a robust relapse to heroin seeking after short and long punishment-imposed abstinence periods, without significant effects of time, i.e., no incubation of heroin seeking. Individual differences were observed in the development of punishment-induced abstinence and stress-induced relapse. CONCLUSIONS These results suggest that stress is a reliable trigger to relapse even after a prolonged period of punishment-induced, voluntary abstinence.
Collapse
Affiliation(s)
- C Borges
- Department of Psychology, Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Quebec, Canada
| | - F Inigo
- Department of Psychology, Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Quebec, Canada
| | - N Quteishat
- Department of Psychology, Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Quebec, Canada
| | - J Charles
- Department of Psychology, Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Quebec, Canada
| | - E Ah-Yen
- Department of Psychology, Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Quebec, Canada
| | - Shalev U
- Department of Psychology, Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Quebec, Canada.
| |
Collapse
|
58
|
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
|
59
|
Funke JR, Hwang EK, Wunsch AM, Baker R, Engeln KA, Murray CH, Milovanovic M, Caccamise AJ, Wolf ME. Persistent Neuroadaptations in the Nucleus Accumbens Core Accompany Incubation of Methamphetamine Craving in Male and Female Rats. eNeuro 2023; 10:ENEURO.0480-22.2023. [PMID: 36792361 PMCID: PMC10016192 DOI: 10.1523/eneuro.0480-22.2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/06/2023] [Accepted: 01/10/2023] [Indexed: 02/17/2023] Open
Abstract
Relapse is a major problem in treating methamphetamine use disorder. "Incubation of craving" during abstinence is a rat model for persistence of vulnerability to craving and relapse. While methamphetamine incubation has previously been demonstrated in male and female rats, it has not been demonstrated after withdrawal periods greater than 51 d and most mechanistic work used males. Here, we address both gaps. First, although methamphetamine intake was higher in males during self-administration training (6 h/d × 10 d), incubation was similar in males and females, with "incubated" craving persisting through withdrawal day (WD)100. Second, using whole-cell patch-clamp recordings in medium spiny neurons (MSNs) of the nucleus accumbens (NAc) core, we assessed synaptic levels of calcium-permeable AMPA receptors (CP-AMPARs), as their elevation is required for expression of incubation in males. In both sexes, compared with saline-self-administering controls, CP-AMPAR levels were significantly higher in methamphetamine rats across withdrawal, although this was less pronounced in WD100-135 rats than WD15-35 or WD40-75 methamphetamine rats. We also examined membrane properties and NMDA receptor (NMDAR) transmission. In saline controls, MSNs from males exhibited lower excitability than females. This difference was eliminated after incubation because of increased excitability of MSNs from males. NMDAR transmission did not differ between sexes and was not altered after incubation. In conclusion, incubation persists for longer than previously described and equally persistent CP-AMPAR plasticity in NAc core occurs in both sexes. Thus, abstinence-related synaptic plasticity in NAc is similar in males and females although other methamphetamine-related behaviors and neuroadaptations show differences.
Collapse
Affiliation(s)
- Jonathan R Funke
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97212
| | - Eun-Kyung Hwang
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97212
| | - Amanda M Wunsch
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97212
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064
| | - Raines Baker
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97212
| | - Kimberley A Engeln
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97212
| | - Conor H Murray
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064
| | - Mike Milovanovic
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064
| | - Aaron J Caccamise
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064
| | - Marina E Wolf
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97212
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064
| |
Collapse
|
60
|
Wang W, Xie X, Zhuang X, Huang Y, Tan T, Gangal H, Huang Z, Purvines W, Wang X, Stefanov A, Chen R, Rodriggs L, Chaiprasert A, Yu E, Vierkant V, Hook M, Huang Y, Darcq E, Wang J. Striatal μ-opioid receptor activation triggers direct-pathway GABAergic plasticity and induces negative affect. Cell Rep 2023; 42:112089. [PMID: 36796365 PMCID: PMC10404641 DOI: 10.1016/j.celrep.2023.112089] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 12/27/2022] [Accepted: 01/26/2023] [Indexed: 02/17/2023] Open
Abstract
Withdrawal from chronic opioid use often causes hypodopaminergic states and negative affect, which may drive relapse. Direct-pathway medium spiny neurons (dMSNs) in the striatal patch compartment contain μ-opioid receptors (MORs). It remains unclear how chronic opioid exposure and withdrawal impact these MOR-expressing dMSNs and their outputs. Here, we report that MOR activation acutely suppressed GABAergic striatopallidal transmission in habenula-projecting globus pallidus neurons. Notably, withdrawal from repeated morphine or fentanyl administration potentiated this GABAergic transmission. Furthermore, intravenous fentanyl self-administration enhanced GABAergic striatonigral transmission and reduced midbrain dopaminergic activity. Fentanyl-activated striatal neurons mediated contextual memory retrieval required for conditioned place preference tests. Importantly, chemogenetic inhibition of striatal MOR+ neurons rescued fentanyl withdrawal-induced physical symptoms and anxiety-like behaviors. These data suggest that chronic opioid use triggers GABAergic striatopallidal and striatonigral plasticity to induce a hypodopaminergic state, which may promote negative emotions and relapse.
Collapse
Affiliation(s)
- Wei Wang
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA; Interdisciplinary Faculty of Toxicology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Xueyi Xie
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| | - Xiaowen Zhuang
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| | - Yufei Huang
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA; Institute for Neuroscience, Texas A&M University, College Station, TX 77843, USA
| | - Tao Tan
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| | - Himanshu Gangal
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA; Institute for Neuroscience, Texas A&M University, College Station, TX 77843, USA
| | - Zhenbo Huang
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| | - William Purvines
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA; Institute for Neuroscience, Texas A&M University, College Station, TX 77843, USA
| | - Xuehua Wang
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| | - Alexander Stefanov
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA; Institute for Neuroscience, Texas A&M University, College Station, TX 77843, USA
| | - Ruifeng Chen
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA; Interdisciplinary Faculty of Toxicology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Lucas Rodriggs
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| | - Anita Chaiprasert
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| | - Emily Yu
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| | - Valerie Vierkant
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| | - Michelle Hook
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA; Institute for Neuroscience, Texas A&M University, College Station, TX 77843, USA
| | - Yun Huang
- Institute of Biosciences and Technology, Department of Translational Medical Sciences, College of Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Emmanuel Darcq
- Department of Psychiatry, University of Strasbourg, INSERM U1114, 67084 Strasbourg Cedex, France
| | - Jun Wang
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA; Interdisciplinary Faculty of Toxicology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA; Institute for Neuroscience, Texas A&M University, College Station, TX 77843, USA; Institute of Biosciences and Technology, Department of Translational Medical Sciences, College of Medicine, Texas A&M University, Houston, TX 77030, USA.
| |
Collapse
|
61
|
Martínez-Rivera A, Hao J, Rice R, Inturrisi CE, Rajadhyaksha AM. Ca v1.3 L-type Ca 2+ channel-activated CaMKII/ERK2 pathway in the ventral tegmental area is required for cocaine conditioned place preference. Neuropharmacology 2023; 224:109368. [PMID: 36481277 PMCID: PMC9796157 DOI: 10.1016/j.neuropharm.2022.109368] [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] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 11/23/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
We have previously demonstrated that pharmacological blockade of ventral tegmental area (VTA) Cav1.3 L-type calcium channels (LTCCs) using Cav1.2 dihydropyridine insensitive (Cav1.2DHP-/-) mutant mice attenuates cocaine conditioned place preference (CPP). However, the molecular mechanisms by which Cav1.3 channels mediate the effects of cocaine in the VTA remain largely unknown. In this study using Cav1.2DHP-/- male mice, we find that cocaine place preference increases CaM kinase IIα, ERK2, and CREB phosphorylation in the VTA, proteins strongly linked to cocaine behaviors. To further explore the causal role of these intracellular signaling proteins in cocaine preference, the CaM kinase II inhibitor, KN93 was directly injected into the VTA of male mice before each cocaine conditioning session. We found that KN93 attenuates conditioned preference for cocaine compared to vehicle treated mice and decreased VTA ERK2 and CREB phosphorylation. Additionally, blockade of the ERK pathway with the MEK inhibitor, U0126 or knockdown of ERK2 using siRNA, attenuated cocaine preference and VTA CREB phosphorylation but not CaMKIIα phosphorylation, suggesting that ERK is activated downstream of CaMKIIα. Examination of postsynaptic density (PSD) GluA1 subunit of AMPA receptors in the nucleus accumbens (NAc) that we have previously shown to be upregulated following long withdrawal periods, was blunted by KN93, U0126 and ERK2 siRNA when examined 30 days following cocaine CPP. Taken together, these findings demonstrate that Cav1.3 channels in the VTA are required for cocaine reward behavior and activation of the CaMKIIα/ERK/CREB signaling pathway in the VTA is necessary for long-lasting changes in the NAc. This article is part of the Special Issue on 'L-type calcium channel mechanisms in neuropsychiatric disorders'.
Collapse
Affiliation(s)
- Arlene Martínez-Rivera
- Department of Pediatrics, Division of Pediatric Neurology, Weill Cornell Medicine, New York, NY, USA; Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| | - Jin Hao
- Department of Pediatrics, Division of Pediatric Neurology, Weill Cornell Medicine, New York, NY, USA
| | - Richard Rice
- Department of Pediatrics, Division of Pediatric Neurology, Weill Cornell Medicine, New York, NY, USA
| | | | - Anjali M Rajadhyaksha
- Department of Pediatrics, Division of Pediatric Neurology, Weill Cornell Medicine, New York, NY, USA; Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
62
|
Chiariello R, McCarthy C, Glaeser BL, Shah AS, Budde MD, Stemper BD, Olsen CM. Chronicity of repeated blast traumatic brain injury associated increase in oxycodone seeking in rats. Behav Brain Res 2023; 438:114181. [PMID: 36330906 PMCID: PMC9993345 DOI: 10.1016/j.bbr.2022.114181] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/18/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
Abstract
Numerous epidemiological studies have found co-morbidity between non-severe traumatic brain injury (TBI) and substance misuse in both civilian and military populations. Preclinical studies have also identified this relationship for some misused substances. We have previously demonstrated that repeated blast traumatic brain injury (rbTBI) increased oxycodone seeking without increasing oxycodone self-administration, suggesting that the neurological sequelae of traumatic brain injury can elevate opioid misuse liability. Here, we determined the chronicity of this effect by testing different durations of time between injury and oxycodone self-administration and durations of abstinence. We found that the subchronic (four weeks), but not the acute (three days) or chronic (four months) duration between injury and oxycodone self-administration was associated with increased drug seeking and re-acquisition of self-administration following a 10-day abstinence. Examination of other abstinence durations (two days, four weeks, or four months) revealed no effect of rbTBI on drug seeking at any of the abstinence durations tested. Together, these data indicate that there is a window of vulnerability after TBI when oxycodone self-administration is associated with elevated drug seeking and relapse-related behaviors.
Collapse
Affiliation(s)
- Rachel Chiariello
- Department of Neurosurgery, Medical College of Wisconsin, United States; Clement J. Zablocki Veterans Affairs Medical Center, United States
| | - Cassandra McCarthy
- Department of Neurosurgery, Medical College of Wisconsin, United States; Clement J. Zablocki Veterans Affairs Medical Center, United States
| | - Breanna L Glaeser
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, United States; Neuroscience Research Center, Medical College of Wisconsin, United States
| | - Alok S Shah
- Department of Neurosurgery, Medical College of Wisconsin, United States; Clement J. Zablocki Veterans Affairs Medical Center, United States
| | - Matthew D Budde
- Department of Neurosurgery, Medical College of Wisconsin, United States; Clement J. Zablocki Veterans Affairs Medical Center, United States; Neuroscience Research Center, Medical College of Wisconsin, United States
| | - Brian D Stemper
- Clement J. Zablocki Veterans Affairs Medical Center, United States; Neuroscience Research Center, Medical College of Wisconsin, United States; Joint Department of Biomedical Engineering, Marquette University and Medical College of Wisconsin, United States
| | - Christopher M Olsen
- Department of Neurosurgery, Medical College of Wisconsin, United States; Department of Pharmacology and Toxicology, Medical College of Wisconsin, United States; Neuroscience Research Center, Medical College of Wisconsin, United States.
| |
Collapse
|
63
|
Domi A, Domi E, Lagstrom O, Gobbo F, Jerlhag E, Adermark L. Abstinence-Induced Nicotine Seeking Relays on a Persistent Hypoglutamatergic State within the Amygdalo-Striatal Neurocircuitry. eNeuro 2023; 10:ENEURO.0468-22.2023. [PMID: 36754627 PMCID: PMC9946069 DOI: 10.1523/eneuro.0468-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/21/2022] [Accepted: 01/06/2023] [Indexed: 02/10/2023] Open
Abstract
Nicotine robustly sustains smoking behavior by acting as a primary reinforcer and by enhancing the incentive salience of the nicotine-associated stimuli. The motivational effects produced by environmental cues associated with nicotine delivery can progressively manifest during abstinence resulting in reinstatement of nicotine seeking. However, how the activity in reward neuronal circuits is transformed during abstinence-induced nicotine seeking is not yet fully understood. In here we used a contingent nicotine and saline control self-administration model to disentangle the contribution of cue-elicited seeking responding for nicotine after drug abstinence in male Wistar rats. Using ex vivo electrophysiological recordings and a network analysis approach, we defined temporal and brain-region specific amygdalo-striatal glutamatergic alterations that occur during nicotine abstinence. The results from this study provide critical evidence indicating a persistent hypoglutamatergic state within the amygdalo-striatal neurocircuitry over protracted nicotine abstinence. During abstinence-induced nicotine seeking, electrophysiological recordings showed progressive neuroadaptations in dorsal and ventral striatum already at 14-d abstinence while neuroadaptations in subregions of the amygdala emerged only after 28-d abstinence. The observed neuroadaptations pointed to a brain network involving the amygdala and the dorsolateral striatum (DLS) to be implied in cue-induced reinstatement of nicotine seeking. Together these data suggest long-lasting neuroadaptations that might reflect neuroplastic changes responsible to abstinence-induced nicotine craving. Neurophysiological transformations were detected within a time window that allows therapeutic intervention advancing clinical development of preventive strategies in nicotine addiction.
Collapse
Affiliation(s)
- Ana Domi
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 413 90, Sweden
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 413 45, Sweden
| | - Esi Domi
- School of Pharmacy, Pharmacology Unit, Center for Neuroscience, University of Camerino, Camerino 62032, Italy
| | - Oona Lagstrom
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 413 90, Sweden
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 413 45, Sweden
| | - Francesco Gobbo
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9JZ, United Kingdom
| | - Elisabet Jerlhag
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 413 90, Sweden
| | - Louise Adermark
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 413 90, Sweden
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 413 45, Sweden
| |
Collapse
|
64
|
Qin M, Guan J, Huang Y, Lin H, Cui D, Yao T, Tian C, Wong M, Shi Z, Yuan T. A novel model of drug cue-induced behaviours in rhesus macaque subjected to chronic ketamine exposure. Gen Psychiatr 2023; 36:e100902. [PMID: 36760345 PMCID: PMC9884838 DOI: 10.1136/gpsych-2022-100902] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 01/09/2023] [Indexed: 01/28/2023] Open
Affiliation(s)
- Meng Qin
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Jiaoqiong Guan
- Department of Rehabilitation Medicine, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology School of Medicine, Guangzhou, Guangdong, China,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
| | - Yichun Huang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - He Lin
- The Third Research Institute of Ministry of Public Security, Shanghai, China
| | - Ding Cui
- Department of Neurosciences, Laboratory for Neuro- and Psychophysiology, KU Leuven, Leuven, Belgium
| | - Tao Yao
- Department of Neurosciences, Laboratory for Neuro- and Psychophysiology, KU Leuven, Leuven, Belgium
| | - Chaoyang Tian
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, Hainan, China
| | - Mali Wong
- Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Zhe Shi
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Tifei 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, Jiangsu, China
| |
Collapse
|
65
|
Moschak TM, Sloand TJ, Carelli RM. Prelimbic Cortex Activity during a Distress Tolerance Task Predicts Cocaine-Seeking Behavior in Male, But Not Female Rats. J Neurosci 2023; 43:647-655. [PMID: 36639895 PMCID: PMC9888504 DOI: 10.1523/jneurosci.1718-22.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/04/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022] Open
Abstract
Distress tolerance (DT) is defined as the ability to persist in challenging goal-directed behavior in the face of stress, and individuals with low DT exhibit heightened drug-seeking behavior. However, no preclinical studies have examined the neurobiology underlying this phenomenon. To assess this, in vivo electrophysiology was used in Long Evans male and female rats during a DT task to record neural activity in the prelimbic cortex (PrL), a brain region implicated in drug-seeking. Rats were first assessed for DT, defined as the amount of time elapsed before rats quit seeking reward in an increasingly difficult operant task. Subsequently, rats underwent 2 weeks of self-administration for either water/saline or cocaine for 6 h/day. Animals then began a 1 month period of experimenter-imposed abstinence to induce heightened drug-seeking behavior. On day 28 of abstinence, DT and neural activity were reassessed; and on day 30, cocaine-seeking behavior was examined under extinction. Males had significantly higher DT than females and exhibited significantly more phasic PrL activity during the DT task. Furthermore, in male rats with a history of cocaine, PrL activity shifted to track DT; and this change in activity significantly correlated with the change in DT. Additionally, male (but not female) rats with low DT after 28 d of abstinence had significantly heightened drug-seeking behavior. Finally, PrL activity during the DT task predicted cocaine-seeking behavior. Collectively, these data demonstrate an important role for the PrL in DT in males, and link this neural activity and behavior to drug-seeking, particularly in males.SIGNIFICANCE STATEMENT Distress tolerance (DT) is defined as the ability to persist in challenging goal-directed behavior in the face of stress, and individuals with low DT exhibit heightened drug-seeking. Here, we investigated the role of the prelimbic cortex (PrL) in DT and its relationship to cocaine-seeking in male and female rats. We found that males had significantly higher DT than females and exhibited significantly more PrL activity during the DT task. Furthermore, male (but not female) rats with low DT after 28 d of abstinence had significantly heightened drug-seeking behavior. Finally, PrL activity during the DT task predicted cocaine-seeking. These data demonstrate an important role for the PrL in DT and link this neural activity and behavior to drug-seeking in males.
Collapse
Affiliation(s)
- Travis M Moschak
- Department of Biological Sciences, University of Texas at El Paso, El Paso, Texas 79902
| | - T Joseph Sloand
- Department of Psychology & Neuroscience, University of North Carolina Chapel Hill, Chapel Hill, North Carolina 27599
| | - Regina M Carelli
- Department of Psychology & Neuroscience, University of North Carolina Chapel Hill, Chapel Hill, North Carolina 27599
| |
Collapse
|
66
|
Preventing incubation of drug craving to treat drug relapse: from bench to bedside. Mol Psychiatry 2023; 28:1415-1429. [PMID: 36646901 DOI: 10.1038/s41380-023-01942-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 12/24/2022] [Accepted: 01/06/2023] [Indexed: 01/17/2023]
Abstract
In 1986, Gawin and Kleber reported a progressive increase in cue-induced drug craving in individuals with cocaine use disorders during prolonged abstinence. After years of controversy, as of 2001, this phenomenon was confirmed in rodent studies using self-administration model, and defined as the incubation of drug craving. The intensification of cue-induced drug craving after withdrawal exposes abstinent individuals to a high risk of relapse, which urged us to develop effective interventions to prevent incubated craving. Substantial achievements have been made in deciphering the neural mechanisms, with potential implications for reducing drug craving and preventing the relapse. The present review discusses promising drug targets that have been well investigated in animal studies, including some neurotransmitters, neuropeptides, neurotrophic factors, and epigenetic markers. We also discuss translational exploitation and challenges in the field of the incubation of drug craving, providing insights into future investigations and highlighting the potential of pharmacological interventions, environment-based interventions, and neuromodulation techniques.
Collapse
|
67
|
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
|
68
|
Browne CJ, Futamura R, Minier-Toribio A, Hicks EM, Ramakrishnan A, Martínez-Rivera F, Estill M, Godino A, Parise EM, Torres-Berrío A, Cunningham AM, Hamilton PJ, Walker DM, Huckins LM, Hurd YL, Shen L, Nestler EJ. Transcriptional signatures of heroin intake and seeking throughout the brain reward circuit. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.11.523688. [PMID: 36711574 PMCID: PMC9882165 DOI: 10.1101/2023.01.11.523688] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Opioid use disorder (OUD) looms as one of the most severe medical crises currently facing society. More effective therapeutics for OUD requires in-depth understanding of molecular changes supporting drug-taking and relapse. Recent efforts have helped advance these aims, but studies have been limited in number and scope. Here, we develop a brain reward circuit-wide atlas of opioid-induced transcriptional regulation by combining RNA sequencing (RNAseq) and heroin self-administration in male mice modeling multiple OUD-relevant conditions: acute heroin exposure, chronic heroin intake, context-induced drug-seeking following prolonged abstinence, and heroin-primed drug-seeking (i.e., "relapse"). Bioinformatics analysis of this rich dataset identified numerous patterns of molecular changes, transcriptional regulation, brain-region-specific involvement in various aspects of OUD, and both region-specific and pan-circuit biological domains affected by heroin. Integrating RNAseq data with behavioral outcomes using factor analysis to generate an "addiction index" uncovered novel roles for particular brain regions in promoting addiction-relevant behavior, and implicated multi-regional changes in affected genes and biological processes. Comparisons with RNAseq and genome-wide association studies from humans with OUD reveal convergent molecular regulation that are implicated in drug-taking and relapse, and point to novel gene candidates with high therapeutic potential for OUD. These results outline broad molecular reprogramming that may directly promote the development and maintenance of OUD, and provide a foundational resource to the field for future research into OUD mechanisms and treatment strategies.
Collapse
Affiliation(s)
- Caleb J Browne
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Rita Futamura
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Angélica Minier-Toribio
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Emily M Hicks
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Dept. of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Freddyson Martínez-Rivera
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Molly Estill
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Arthur Godino
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Eric M Parise
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Angélica Torres-Berrío
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ashley M Cunningham
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Peter J Hamilton
- Dept. of Anatomy & Neurobiology, Virginia Commonwealth University School of Medicine
| | - Deena M Walker
- Dept. of Behavioral Neuroscience, Oregon Health and Science University
| | - Laura M. Huckins
- Dept. of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai
| | - Yasmin L Hurd
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Dept. of Anatomy & Neurobiology, Virginia Commonwealth University School of Medicine
- Dept. of Behavioral Neuroscience, Oregon Health and Science University
| | - Li Shen
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Dept. of Anatomy & Neurobiology, Virginia Commonwealth University School of Medicine
- Dept. of Behavioral Neuroscience, Oregon Health and Science University
| |
Collapse
|
69
|
Persistent increase of accumbens cocaine ensemble excitability induced by IRK downregulation after withdrawal mediates the incubation of cocaine craving. Mol Psychiatry 2023; 28:448-462. [PMID: 36481931 PMCID: PMC9812793 DOI: 10.1038/s41380-022-01884-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 10/30/2022] [Accepted: 11/10/2022] [Indexed: 12/13/2022]
Abstract
The incubation phenomenon, cue-induced drug craving progressively increasing over prolonged withdrawal, accounts for persistent relapse, leading to a dilemma in the treatment of cocaine addiction. The role of neuronal ensembles activated by initial cocaine experience in the incubation phenomenon was unclear. In this study, with cocaine self-administration (SA) models, we found that neuronal ensembles in the nucleus accumbens shell (NAcSh) showed increasing activation induced by cue-induced drug-seeking after 30-day withdrawal. Inhibition or activation of NAcSh cocaine-ensembles suppressed or promoted craving for cocaine, demonstrating a critical role of NAcSh cocaine-ensembles in incubation for cocaine craving. NAcSh cocaine-ensembles showed a specific increase of membrane excitability and a decrease of inward rectifying channels Kir2.1 currents after 30-day withdrawal. Overexpression of Kir2.1 in NAcSh cocaine-ensembles restored neuronal membrane excitability and suppressed cue-induced drug-seeking after 30-day withdrawal. Expression of dominant-negative Kir2.1 in NAcSh cocaine-ensembles enhanced neuronal membrane excitability and accelerated incubation of cocaine craving. Our results provide a cellular mechanism that the downregulation of Kir2.1 functions in NAcSh cocaine-ensembles induced by prolonged withdrawal mediates the enhancement of ensemble membrane excitability, leading to incubation of cocaine craving.
Collapse
|
70
|
Slivicki RA, Earnest T, Chang YH, Pareta R, Casey E, Li JN, Tooley J, Abiraman K, Vachez YM, Wolf DK, Sackey JT, Kumar Pitchai D, Moore T, Gereau RW, Copits BA, Kravitz AV, Creed MC. Oral oxycodone self-administration leads to features of opioid misuse in male and female mice. Addict Biol 2023; 28:e13253. [PMID: 36577735 DOI: 10.1111/adb.13253] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 11/18/2022]
Abstract
Use of prescription opioids, particularly oxycodone, is an initiating factor driving the current opioid epidemic. There are several challenges with modelling oxycodone abuse. First, prescription opioids including oxycodone are orally self-administered and have different pharmacokinetics and dynamics than morphine or fentanyl, which have been more commonly used in rodent research. This oral route of administration determines the pharmacokinetic profile, which then influences the establishment of drug-reinforcement associations in animals. Moreover, the pattern of intake and the environment in which addictive drugs are self-administered are critical determinants of the levels of drug intake, of behavioural sensitization and of propensity to relapse behaviour. These are all important considerations when modelling prescription opioid use, which is characterized by continuous drug access in familiar environments. Thus, to model features of prescription opioid use and the transition to abuse, we designed an oral, homecage-based oxycodone self-administration paradigm. Mice voluntarily self-administer oxycodone in this paradigm without any taste modification such as sweeteners, and the majority exhibit preference for oxycodone, escalation of intake, physical signs of dependence and reinstatement of seeking after withdrawal. In addition, a subset of animals demonstrate drug taking that is resistant to aversive consequences. This model is therefore translationally relevant and useful for studying the neurobiological substrates of prescription opioid abuse.
Collapse
Affiliation(s)
- Richard A Slivicki
- Washington University Pain Center, Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Tom Earnest
- Washington University Pain Center, Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Yu-Hsuan Chang
- Washington University Pain Center, Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Rajesh Pareta
- Washington University Pain Center, Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Eric Casey
- Department of Psychiatry, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jun-Nan Li
- Washington University Pain Center, Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jessica Tooley
- Washington University Pain Center, Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Kavitha Abiraman
- Washington University Pain Center, Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Yvan M Vachez
- Washington University Pain Center, Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Drew K Wolf
- Washington University Pain Center, Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jason T Sackey
- Washington University Pain Center, Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | | | | | - Robert W Gereau
- Washington University Pain Center, Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri, USA.,Department of Neuroscience, Washington University in St. Louis, St. Louis, Missouri, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Bryan A Copits
- Washington University Pain Center, Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri, USA.,Department of Neuroscience, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Alexxai V Kravitz
- Washington University Pain Center, Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri, USA.,Department of Psychiatry, Washington University in St. Louis, St. Louis, Missouri, USA.,Department of Neuroscience, Washington University in St. Louis, St. Louis, Missouri, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Meaghan C Creed
- Washington University Pain Center, Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri, USA.,Department of Psychiatry, Washington University in St. Louis, St. Louis, Missouri, USA.,Department of Neuroscience, Washington University in St. Louis, St. Louis, Missouri, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
71
|
Guo R, Vaughan DT, Rojo ALA, Huang YH. Sleep-mediated regulation of reward circuits: implications in substance use disorders. Neuropsychopharmacology 2023; 48:61-78. [PMID: 35710601 PMCID: PMC9700806 DOI: 10.1038/s41386-022-01356-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/22/2022] [Accepted: 05/27/2022] [Indexed: 12/11/2022]
Abstract
Our modern society suffers from both pervasive sleep loss and substance abuse-what may be the indications for sleep on substance use disorders (SUDs), and could sleep contribute to the individual variations in SUDs? Decades of research in sleep as well as in motivated behaviors have laid the foundation for us to begin to answer these questions. This review is intended to critically summarize the circuit, cellular, and molecular mechanisms by which sleep influences reward function, and to reveal critical challenges for future studies. The review also suggests that improving sleep quality may serve as complementary therapeutics for treating SUDs, and that formulating sleep metrics may be useful for predicting individual susceptibility to SUDs and other reward-associated psychiatric diseases.
Collapse
Affiliation(s)
- Rong Guo
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Allen Institute, Seattle, WA, 98109, USA
| | - Dylan Thomas Vaughan
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- The Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
| | - Ana Lourdes Almeida Rojo
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- The Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
| | - Yanhua H Huang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA.
- The Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
72
|
The Protective Effect of Social Reward on Opioid and Psychostimulant Reward and Relapse: Behavior, Pharmacology, and Brain Regions. J Neurosci 2022; 42:9298-9314. [PMID: 36517252 PMCID: PMC9794371 DOI: 10.1523/jneurosci.0931-22.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 10/27/2022] [Accepted: 10/31/2022] [Indexed: 12/30/2022] Open
Abstract
Until recently, most modern neuroscience research on addiction using animal models did not incorporate manipulations of social factors. Social factors play a critical role in human addiction: social isolation and exclusion can promote drug use and relapse, while social connections and inclusion tend to be protective. Here, we discuss the state of the literature on social factors in animal models of opioid and psychostimulant preference, self-administration, and relapse. We first summarize results from rodent studies on behavioral, pharmacological, and circuit mechanisms of the protective effect of traditional experimenter-controlled social interaction procedures on opioid and psychostimulant conditioned place preference, self-administration, and relapse. Next, we summarize behavioral and brain-mechanism results from studies using newer operant social-interaction procedures that inhibit opioid and psychostimulant self-administration and relapse. We conclude by discussing how the reviewed studies point to future directions for the addiction field and other neuroscience and psychiatric fields, and their implications for mechanistic understanding of addiction and development of new treatments.SIGNIFICANCE STATEMENT In this review, we propose that incorporating social factors into modern neuroscience research on addiction could improve mechanistic accounts of addiction and help close gaps in translating discovery to treatment. We first summarize rodent studies on behavioral, pharmacological, and circuit mechanisms of the protective effect of both traditional experimenter-controlled and newer operant social-interaction procedures. We then discuss potential future directions and clinical implications.
Collapse
|
73
|
Guo R, Wang Y, Yan R, Chen B, Ding W, Gorczyca MT, Ozsoy S, Cai L, Hines RL, Tseng GC, Allocca G, Dong Y, Fang J, Huang YH. Rapid Eye Movement Sleep Engages Melanin-Concentrating Hormone Neurons to Reduce Cocaine Seeking. Biol Psychiatry 2022; 92:880-894. [PMID: 35953320 PMCID: PMC9872495 DOI: 10.1016/j.biopsych.2022.06.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 06/03/2022] [Accepted: 06/06/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND Persistent sleep disruptions following withdrawal from abused drugs may hold keys to battle drug relapse. It is posited that there may be sleep signatures that predict relapse propensity, identifying which may open new avenues for treating substance use disorders. METHODS We trained male rats (approximately postnatal day 56) to self-administer cocaine. After long-term drug withdrawal (approximately postnatal day 100), we examined the correlations between the intensity of cocaine seeking and key sleep features. To test for causal relationships, we then used behavioral, chemogenetic, or optogenetic methods to selectively increase rapid eye movement sleep (REMS) and measured behavioral and electrophysiological outcomes to probe for cellular and circuit mechanisms underlying REMS-mediated regulation of cocaine seeking. RESULTS A selective set of REMS features was preferentially associated with the intensity of cue-induced cocaine seeking after drug withdrawal. Moreover, selectively increasing REMS time and continuity by environmental warming attenuated a withdrawal time-dependent intensification of cocaine seeking, or incubation of cocaine craving, suggesting that REMS may benefit withdrawal. Warming increased the activity of lateral hypothalamic melanin-concentrating hormone (MCH) neurons selectively during prolonged REMS episodes and counteracted cocaine-induced synaptic accumulation of calcium-permeable AMPA receptors in the nucleus accumbens-a critical substrate for incubation. Finally, the warming effects were partly mimicked by chemogenetic or optogenetic stimulations of MCH neurons during sleep, or intra-accumbens infusions of MCH peptide during the rat's inactive phase. CONCLUSIONS REMS may encode individual vulnerability to relapse, and MCH neuron activities can be selectively targeted during REMS to reduce drug relapse.
Collapse
Affiliation(s)
- Rong Guo
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yao Wang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rongzhen Yan
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bo Chen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wanqiao Ding
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael T Gorczyca
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sahin Ozsoy
- Somnivore Pty. Ltd., Bacchus Marsh, Victoria, Australia
| | - Li Cai
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rachel L Hines
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - George C Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Giancarlo Allocca
- Somnivore Pty. Ltd., Bacchus Marsh, Victoria, Australia; Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Yan Dong
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jidong Fang
- Department of Psychiatry and Behavioral Health, Penn State College of Medicine, Hershey, Pennsylvania
| | - Yanhua H Huang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
74
|
Olaniran A, Garcia KT, Burke MAM, Lin H, Venniro M, Li X. Operant social seeking to a novel peer after social isolation is associated with activation of nucleus accumbens shell in rats. Psychopharmacology (Berl) 2022:10.1007/s00213-022-06280-9. [PMID: 36449074 PMCID: PMC10227185 DOI: 10.1007/s00213-022-06280-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/08/2022] [Indexed: 12/03/2022]
Abstract
RATIONALE AND OBJECTIVE Deprivation of social interaction promotes social reward seeking in rodents, assessed primarily by the conditioned place preference procedure. Here, we used an operant social procedure in rats and examined the effect of the housing condition (pair-housing vs. single-housing) during or after social self-administration on social reward seeking. METHODS We first trained paired-housed or single-housed rats to gain access to an age- and sex-matched novel peer. On post-training day 1 (PTD1), we tested both groups for social seeking without the presence of the novel peer. Next, we divided each group into pair-housing or single-housing conditions and tested all four groups (pair-pair, pair-single, single-pair, and single-single) for social seeking on post-training day 12 (PTD12). Finally, we analyzed Fos expression in the striatum associated with social seeking on PTD12. RESULT Single-housed rats earned more social rewards during social self-administration than pair-housed rats. Social isolation during social self-administration also promoted social seeking on PTD1 and PTD12, regardless of their housing conditions after social self-administration training. Additionally, in pair-housed rats, social isolation during the post-training period led to a time-dependent increase of social seeking on PTD12 compared with PTD1. Finally, the Fos analyses revealed an increase of Fos expression in NAc shell of single-single rats after social seeking test on PTD12 compared with pair-pair rats. CONCLUSION Our data suggest that social isolation promotes operant social self-administration and social seeking. In addition, neuronal activation of NAc shell is associated with social seeking after social isolation.
Collapse
Affiliation(s)
- Adedayo Olaniran
- Department of Psychology, University of Maryland College Park, College Park, MD, 20742, USA
| | - Kristine T Garcia
- Department of Psychology, University of Maryland College Park, College Park, MD, 20742, USA
| | - Megan A M Burke
- Department of Psychology, University of Maryland College Park, College Park, MD, 20742, USA
| | - Hongyu Lin
- Department of Psychology, University of Maryland College Park, College Park, MD, 20742, USA
| | - Marco Venniro
- Department of Neurobiology and Anatomy, University of Maryland Baltimore, Baltimore, MD, 21201, USA.
| | - Xuan Li
- Department of Psychology, University of Maryland College Park, College Park, MD, 20742, USA.
| |
Collapse
|
75
|
Madangopal R, Szelenyi ER, Nguyen J, Brenner MB, Drake OR, Pham DQ, Shekara A, Jin M, Choong JJ, Heins C, Komer LE, Weber SJ, Hope BT, Shaham Y, Golden SA. Incubation of palatable food craving is associated with brain-wide neuronal activation in mice. Proc Natl Acad Sci U S A 2022; 119:e2209382119. [PMID: 36603188 PMCID: PMC9659381 DOI: 10.1073/pnas.2209382119] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/14/2022] [Indexed: 11/06/2022] Open
Abstract
Studies using rodent models have shown that relapse to drug or food seeking increases progressively during abstinence, a behavioral phenomenon termed "incubation of craving." Mechanistic studies of incubation of craving have focused on specific neurobiological targets within preselected brain areas. Recent methodological advances in whole-brain immunohistochemistry, clearing, and imaging now allow unbiased brain-wide cellular resolution mapping of regions and circuits engaged during learned behaviors. However, these whole-brain imaging approaches were developed for mouse brains, while incubation of drug craving has primarily been studied in rats, and incubation of food craving has not been demonstrated in mice. Here, we established a mouse model of incubation of palatable food craving and examined food reward seeking after 1, 15, and 60 abstinence days. We then used the neuronal activity marker Fos with intact-brain mapping procedures to identify corresponding patterns of brain-wide activation. Relapse to food seeking was significantly higher after 60 abstinence days than after 1 or 15 days. Using unbiased ClearMap analysis, we identified increased activation of multiple brain regions, particularly corticostriatal structures, following 60 but not 1 or 15 abstinence days. We used orthogonal SMART2 analysis to confirm these findings within corticostriatal and thalamocortical subvolumes and applied expert-guided registration to investigate subdivision and layer-specific activation patterns. Overall, we 1) identified brain-wide activity patterns during incubation of food seeking using complementary analytical approaches and 2) provide a single-cell resolution whole-brain atlas that can be used to identify functional networks and global architecture underlying the incubation of food craving.
Collapse
Affiliation(s)
- Rajtarun Madangopal
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Eric R. Szelenyi
- Department of Biological Structure, University of Washington, Seattle, WA 98195
- Center of Excellence in Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195
| | - Joseph Nguyen
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Megan B. Brenner
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Olivia R. Drake
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Diana Q. Pham
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Aniruddha Shekara
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Michelle Jin
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Jia Jie Choong
- Department of Biological Structure, University of Washington, Seattle, WA 98195
- Center of Excellence in Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195
- Department of Electrical and Computer Engineering, University of Washington, Seattle, WA 98195
| | - Conor Heins
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Lauren E. Komer
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Sophia J. Weber
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Bruce T. Hope
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Yavin Shaham
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Sam A. Golden
- Department of Biological Structure, University of Washington, Seattle, WA 98195
- Center of Excellence in Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195
| |
Collapse
|
76
|
Darrag HM, Almuhanna HT, Hakami EH, Alhojaily SM. Analysis of Volatile Secondary Metabolites in Ocimum basilicum Cell Suspensions: Inhibition, In Silico Molecular Docking, and an ADMET Analysis against Proteolytic Enzymes of Rhynchophorus ferrugineus. PLANTS (BASEL, SWITZERLAND) 2022; 11:2949. [PMID: 36365402 PMCID: PMC9655874 DOI: 10.3390/plants11212949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 06/16/2023]
Abstract
Our study's overarching goal was to determine which O. basilicum cell suspensions approach yielded the most insecticidal and R. ferrugineus-inhibitory volatile secondary metabolites. After inoculation with Verticillium dahliae as an activator, the growth kinetics were measured, and the extract was identified using GC-MS. Validation was achieved for the insecticidal efficacy of a volatile extract, the pure phenolic content against larva and adult R. ferrugineus, and the inhibitory effect on proteases (in vivo and in vitro). The volatile extract achieved an LC50 of 1229 µg/mL and an LD50 of 13.8 µg/larva. The LC50 values for β-bergamotene, α-eudesmol, β-farnesene, linalool, 1,8-cineole, eugenol, α-guaiene, and β-caryophyllene were 1294, 1312, 1356, 1398, 1426, 1459, 1491, and 1523 g/mL, respectively. The LD50 activities of α-eudesmol, linalool, 1,8-cineole, eugenol, and nerol were 12.4, 13.7, 13.9, 14.2, and 15.6 g/larva, respectively. Active volatile extract of O. basilicum inhibited trypsin proteinase, elastase, cysteine, overall protease, and metalloprotease activity with IC50 values of 89.4, 101.7, 394.7, 112.4, and 535.2 µg/mL and 178.5, 192.4, 547.3, 208.3, and 924.8 µg/mL, in vitro and in vivo, respectively. There was evidence of action against total proteases (in vitro) with IC50 values of 78.9, 81.2, 88.6, 90.7, 91.5, 97.6, 107.4, and 176.3 µg/mL for β-bergamotene, α-eudesmol, β-farnesene, linalool, 1,8-cineole, eugenol, α-guaiene, and β-caryophyllene, respectively. Total proteases (in vivo) are inhibited by the α-eudesmol, linalool, 1,8-cineole, eugenol, nerol, and (E)-β-ocimene, with IC50 values of 162.3, 192.7, 193.1, 201.4, 248.6, and 273.2 µg/mL, respectively. ADMET and molecular docking modeling were the only two methods used to conduct in-depth computational analyses of compounds. The study recommended using an efficient cell suspension method to produce a volatile extract rich in useful secondary metabolites that may be utilized as a bio-insecticide.
Collapse
Affiliation(s)
- Hossam Moustafa Darrag
- Department of Research and Training, Research and Training Station, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- Pesticide Chemistry and Technology Department, Faculty of Agriculture, Alexandria University, Alexandria 21545, Egypt
| | - Hani Taher Almuhanna
- Research and Training Station, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | | | - Sameer M. Alhojaily
- Research and Training Station, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| |
Collapse
|
77
|
Kuhn BN, Cannella N, Crow AD, Roberts AT, Lunerti V, Allen C, Nall RW, Hardiman G, Woods LCS, Chung D, Ciccocioppo R, Kalivas PW. Novelty-induced locomotor behavior predicts heroin addiction vulnerability in male, but not female, rats. Psychopharmacology (Berl) 2022; 239:3605-3620. [PMID: 36112154 PMCID: PMC9632364 DOI: 10.1007/s00213-022-06235-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 09/02/2022] [Indexed: 11/28/2022]
Abstract
RATIONALE The ongoing rise in opioid use disorder (OUD) has made it imperative to better model the individual variation within the human population that contributes to OUD vulnerability. Using animal models that capture such variation can be a useful tool. Individual variation in novelty-induced locomotion is predictive of substance use disorder (SUD) propensity. In this model, rats are characterized as high-responders (HR) or low-responders (LR) using a median split based on distance travelled during a locomotor test, and HR rats are generally found to exhibit a more SUD vulnerable behavioral phenotype. OBJECTIVES The HR/LR model has commonly been used to assess behaviors in male rats using psychostimulants, with limited knowledge of the predictive efficacy of this model in females or the use of an opioid as the reward. In the current study, we assessed several behaviors across the different phases of drug addiction (heroin taking, refraining, and seeking) in over 500 male and female heterogeneous stock rats run at two geographically separate locations. Rats were characterized as HRs or LRs within each sex for analysis. RESULTS Overall, females exhibit a more OUD vulnerable phenotype relative to males. Additionally, the HR/LR model was predictive of OUD-like behaviors in male, but not female rats. Furthermore, phenotypes did not differ in anxiety-related behaviors, reacquisition of heroin-taking, or punished heroin-taking behavior in either sex. CONCLUSIONS These results emphasize the importance of assessing females in models of individual variation in SUD and highlight limitations in using the HR/LR model to assess OUD propensity.
Collapse
Affiliation(s)
- Brittany N Kuhn
- Department of Neuroscience, Medical University of South Carolina, 173 Ashley Avenue, BSB 403-MSC 510, Charleston, SC, 29425, USA.
| | | | - Ayteria D Crow
- Department of Neuroscience, Medical University of South Carolina, 173 Ashley Avenue, BSB 403-MSC 510, Charleston, SC, 29425, USA
| | - Analyse T Roberts
- Department of Neuroscience, Medical University of South Carolina, 173 Ashley Avenue, BSB 403-MSC 510, Charleston, SC, 29425, USA
| | | | - Carter Allen
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Rusty W Nall
- Department of Psychology, Jacksonville State University, Jacksonville, AL, USA
| | - Gary Hardiman
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | | | - Dongjun Chung
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | | | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, 173 Ashley Avenue, BSB 403-MSC 510, Charleston, SC, 29425, USA
| |
Collapse
|
78
|
Wong B, Zimbelman AR, Milovanovic M, Wolf ME, Stefanik MT. GluA2-lacking AMPA receptors in the nucleus accumbens core and shell contribute to the incubation of oxycodone craving in male rats. Addict Biol 2022; 27:e13237. [PMID: 36301206 PMCID: PMC10655598 DOI: 10.1111/adb.13237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 07/31/2022] [Accepted: 09/22/2022] [Indexed: 01/24/2023]
Abstract
One of the most challenging issues in the treatment of substance use disorder, including misuse of opioids such as oxycodone, is persistent vulnerability to relapse, often triggered by cues or contexts previously associated with drug use. In rats, cue-induced craving progressively intensifies ('incubates') during withdrawal from extended-access self-administration of several classes of misused drugs, including the psychostimulants cocaine and methamphetamine. For these psychostimulants, incubation is associated with strengthening of excitatory synapses in the nucleus accumbens (NAc) through incorporation of α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors that lack the GluA2 subunit and are therefore Ca2+ -permeable (CP-AMPARs). Once CP-AMPAR upregulation occurs, their stimulation is required for expression of incubation. It is not known if a similar mechanism contributes to incubation of oxycodone craving. Using male rats, we established that incubation occurs by withdrawal day (WD) 15 and persists through WD30. Then, using cell-surface biotinylation, we found that surface levels of the AMPAR subunit GluA1 but not GluA2 are elevated in NAc core and shell of oxycodone rats on WD15, although this wanes by WD30. Next, using intra-NAc injection of the selective CP-AMPAR antagonist Naspm before a seeking test, we demonstrate that CP-AMPAR blockade in either subregion decreases oxycodone seeking on WD15 or WD30 (after incubation), but not WD1, and has no effect in saline self-administering animals. The Naspm results suggest CP-AMPARs persist in synapses through WD30 even if total cell surface levels wane. These results suggest that a common neurobiological mechanism contributes to expression of incubation of craving for oxycodone and psychostimulants.
Collapse
Affiliation(s)
- Benjamin Wong
- Department of Psychology and Neuroscience, North Central College, Naperville, Illinois, USA
| | - Alexa R. Zimbelman
- Department of Psychology and Neuroscience, North Central College, Naperville, Illinois, USA
| | - Mike Milovanovic
- Department of Neuroscience, Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Marina E. Wolf
- Department of Neuroscience, Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
| | - Michael T. Stefanik
- Department of Psychology and Neuroscience, North Central College, Naperville, Illinois, USA
- Department of Neuroscience, Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| |
Collapse
|
79
|
Muacevic A, Adler JR, Natarajan B, Mitra S, Tango T, Ahmed A, Bansode S, Almushawah AA. Current Treatments of Post-traumatic Stress Disorder and Amygdala Ablation as a Potential Cutting-Edge Therapy in Its Refractory Cases. Cureus 2022; 14:e31943. [PMID: 36582576 PMCID: PMC9794924 DOI: 10.7759/cureus.31943] [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] [Accepted: 11/27/2022] [Indexed: 11/29/2022] Open
Abstract
Post-traumatic stress disorder (PTSD)is a very common psychiatric disorder occurring in an individual of any age, gender, or race who underwent trauma, with women being twice more at risk than men. It is generally seen more in American Indians, United States Latinos, and African American ethnic groups. A patient is diagnosed with PTSD if the symptoms of intrusion, avoidance, changes in cognition and emotions, arousal, and mood reactivity changes persist for more than a month and cause the individual severe difficulty in their everyday cognitive and psychological functioning. The psychological treatment includes numerous therapies including trauma-focused therapies such as cognitive behavioral therapy, cognitive processing therapy, prolonged exposure therapy, eye movement desensitization and reprocessing, and non-trauma-focused therapies such as relaxation techniques, interpersonal therapy, and mindfulness. Various pharmacological measures have also been tried with mixed results such as selective serotonin reuptake inhibitors, benzodiazepines, adrenergic drugs, atypical antipsychotics, and mood stabilizers like lithium and valproate. As numerous studies have proven, PTSD is linked with right-side stimulation of the amygdala. The purpose of this article is to highlight the use of extremely selective laser ablation of the amygdala-hippocampal unit as a successful surgical intervention for medically unresponsive PTSD and as a revolutionary solution and prospective cutting-edge therapy in the near future.
Collapse
|
80
|
Samson KR, Xu W, Kortagere S, España RA. Intermittent access to oxycodone decreases dopamine uptake in the nucleus accumbens core during abstinence. Addict Biol 2022; 27:e13241. [PMID: 36301217 PMCID: PMC10262085 DOI: 10.1111/adb.13241] [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: 02/25/2022] [Revised: 07/29/2022] [Accepted: 09/27/2022] [Indexed: 01/24/2023]
Abstract
A major obstacle in treating opioid use disorder is the persistence of drug seeking or craving during periods of abstinence, which is believed to contribute to relapse. Dopamine transmission in the mesolimbic pathway is posited to contribute to opioid reinforcement, but the processes by which dopamine influences drug seeking have not been completely elucidated. To examine whether opioid seeking during abstinence is associated with alterations in dopamine transmission, female and male rats self-administered oxycodone under an intermittent access schedule of reinforcement. Following self-administration, rats underwent a forced abstinence period, and cue-induced seeking tests were conducted to assess oxycodone seeking. One day following the final seeking test, rats were sacrificed to perform ex vivo fast scan cyclic voltammetry and western blotting in the nucleus accumbens. Rats displayed reduced dopamine uptake rate on abstinence day 2 and abstinence day 15, compared to oxycodone-naïve rats. Further, on abstinence day 15, rats had reduced phosphorylation of the dopamine transporter. Additionally, local application of oxycodone to the nucleus accumbens reduced dopamine uptake in oxycodone-naïve rats and in rats during oxycodone abstinence, on abstinence day 2 and abstinence day 15. These observations suggest that abstinence from oxycodone results in dysfunctional dopamine transmission, which may contribute to sustained oxycodone seeking during abstinence.
Collapse
Affiliation(s)
- Kyle R. Samson
- Drexel University College of Medicine, Department of Neurobiology and Anatomy
| | - Wei Xu
- Drexel University College of Medicine, Department of Microbiology and Immunology
| | - Sandhya Kortagere
- Drexel University College of Medicine, Department of Microbiology and Immunology
| | - Rodrigo A. España
- Drexel University College of Medicine, Department of Neurobiology and Anatomy
| |
Collapse
|
81
|
Abstract
Relapse is a defining feature of smoking and a significant challenge in cessation management. Elucidation of novel factors underlying relapse may inform future treatments. Cotinine, the major metabolite of nicotine, has been shown to support intravenous self-administration in rats, implicating it as one potential factor contributing to nicotine reinforcement. However, it remains unknown whether cotinine would induce relapse-like behaviors. The current study investigated relapse to cotinine seeking in two relapse models, the reinstatement of drug seeking and incubation of drug craving models. In the reinstatement model, rats were trained to self-administer cotinine, underwent extinction of cotinine-associated responses, and were tested for cue-, drug-, or stress-induced reinstatement. Conditioned cues associated with cotinine self-administration, cotinine (1-2 mg/kg), or the pharmacological stressor yohimbine (1.25-2.5 mg/kg) induced reinstatement of cotinine seeking. Female rats displayed more pronounced cue-induced, but not drug- or stress-induced reinstatement than male rats. In the incubation of the craving model, rats were trained to self-administer cotinine and underwent forced withdrawal in home cages. Rats were tested for cue-induced cotinine-seeking on both withdrawal day 1 and withdrawal day 18. Rats exhibited greater cue-induced cotinine-seeking on withdrawal day 18 compared to withdrawal day 1, with no difference between male and female rats. These findings indicate that cotinine induces sex-specific relapse to drug seeking in rats, suggesting that cotinine may contribute to relapse.
Collapse
Affiliation(s)
- Xiaoying Tan
- Department of Anesthesiology & Perioperative Medicine
| | | | - Zheng-Ming Ding
- Department of Anesthesiology & Perioperative Medicine
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
82
|
Edson H, Ball KT. Sex-dependent effects of chronic stress on punished cocaine self-administration and cue-induced relapse to cocaine seeking. Physiol Behav 2022; 257:113972. [PMID: 36170943 DOI: 10.1016/j.physbeh.2022.113972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/18/2022] [Accepted: 09/23/2022] [Indexed: 10/14/2022]
Abstract
We tested the effects of chronic stress on cocaine relapse after drug-reinforced responding was suppressed by punishment, an animal model of human relapse after self-imposed abstinence due to the negative consequences of drug use. Male rats displayed greater resistance to punishment than females, but daily stress decreased this resistance. Only female rats with a history of chronic stress displayed increased responding for cocaine cues from abstinence Day 1 to Day 8. Thus, the effects of chronic stress on punished cocaine self-administration and cue-induced relapse are dependent on biological sex, and may have implications for more targeted treatments for cocaine addiction.
Collapse
Affiliation(s)
- Hunter Edson
- Department of Psychology, Bloomsburg University of Pennsylvania, 400 E. 2nd St., Bloomsburg, PA 17815, USA
| | - Kevin T Ball
- Department of Psychology, Bloomsburg University of Pennsylvania, 400 E. 2nd St., Bloomsburg, PA 17815, USA.
| |
Collapse
|
83
|
Mayberry HL, Bavley CC, Karbalaei R, Peterson DR, Bongiovanni AR, Ellis AS, Downey SH, Toussaint AB, Wimmer ME. Transcriptomics in the nucleus accumbens shell reveal sex- and reinforcer-specific signatures associated with morphine and sucrose craving. Neuropsychopharmacology 2022; 47:1764-1775. [PMID: 35190706 PMCID: PMC9372067 DOI: 10.1038/s41386-022-01289-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 12/16/2022]
Abstract
Incubation of craving is a well-documented phenomenon referring to the intensification of drug craving over extended abstinence. The neural adaptations that occur during forced abstinence following chronic drug taking have been a topic of intense study. However, little is known about the transcriptomic changes occurring throughout this window of time. To define gene expression changes associated with morphine consumption and extended abstinence, male and female rats underwent 10 days of morphine self-administration. Separate drug-naive rats self-administered sucrose in order to compare opioid-induced changes from those associated with natural, non-drug rewards. After one or 30 days of forced abstinence, rats were tested for craving, or nucleus accumbens shell tissue was dissected for RNA sequencing. Morphine consumption was predictive of drug seeking after extended (30 days) but not brief (1 day) abstinence in both sexes. Extended abstinence was also associated with robust sex- and reinforcer-specific changes in gene expression, suggesting sex differences underlying incubation of morphine and sucrose seeking respectively. Importantly, these changes in gene expression occurred without re-exposure to drug-paired cues, indicating that chronic morphine causes long-lasting changes in gene expression that prime the system for increased craving. These findings lay the groundwork for identifying specific therapeutic targets for curbing opioid craving without impacting the natural reward system in males and females.
Collapse
Affiliation(s)
- Hannah L Mayberry
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Charlotte C Bavley
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Reza Karbalaei
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Drew R Peterson
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Angela R Bongiovanni
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Alexandra S Ellis
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Sara H Downey
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Andre B Toussaint
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Mathieu E Wimmer
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
84
|
Farrell MR, Ye Q, Xie Y, Esteban JSD, Mahler SV. Ventral pallidum GABA neurons bidirectionally control opioid relapse across rat behavioral models. ADDICTION NEUROSCIENCE 2022; 3:100026. [PMID: 36156918 PMCID: PMC9494709 DOI: 10.1016/j.addicn.2022.100026] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Opioid addiction is a chronic, relapsing disorder. Whether addicted individuals are forced to abstain or they decide themselves to quit using drugs, relapse rates are high-especially upon encountering contexts and stimuli associated with prior opioid use. Rodents similarly show context- and cue-induced reinstatement of drug seeking following abstinence, and intriguingly, the neural circuits underlying these relapse-like behaviors differ when abstinence is involuntarily imposed, responding is extinguished, or animals decide themselves to cease taking drug. Here, we employ two complementary rat behavioral models of relapse-like behavior for the highly reinforcing opioid drug remifentanil, and asked whether GABAergic neurons in the ventral pallidum (VPGABA) control opioid seeking under these behavioral conditions. Specifically, we asked how chemogenetically stimulating VPGABA neurons with clozapine-N-oxide (CNO) influences the ability of contextual or discrete remifentanil-paired cues to reinstate drug seeking following either voluntary abstinence (punishment-induced; GroupPunish), or extinction training (GroupExt). In GroupPunish rats, we also chemogenetically inhibited VPGABA neurons, and examined spontaneous VP activity (Fos) during cued reinstatement. In both GroupPunish and GroupExt rats, stimulating Gq-signaling in VPGABA neurons augmented remifentanil reinstatement in a cue- and context-dependent manner. Conversely, engaging inhibitory Gi-signaling in VPGABA neurons in GroupPunish suppressed cue-induced reinstatement, and cue-triggered seeking was correlated with Fos expression in rostral, but not caudal VP. Neither stimulating nor inhibiting VPGABA neurons influenced unpunished remifentanil self-administration. We conclude that VPGABA neurons bidirectionally control opioid seeking regardless of the specific relapse model employed, highlighting their fundamental role in opioid relapse-like behavior across behavioral models, and potentially across species.
Collapse
Affiliation(s)
- Mitchell R. Farrell
- University of California, Irvine Department of Neurobiology and Behavior, 1203 McGaugh Hall Irvine, CA, 92697, USA
| | - Qiying Ye
- University of California, Irvine Department of Neurobiology and Behavior, 1203 McGaugh Hall Irvine, CA, 92697, USA
| | - Yiyan Xie
- University of California, Irvine Department of Neurobiology and Behavior, 1203 McGaugh Hall Irvine, CA, 92697, USA
| | - Jeanine Sandra D. Esteban
- University of California, Irvine Department of Neurobiology and Behavior, 1203 McGaugh Hall Irvine, CA, 92697, USA
| | - Stephen V. Mahler
- University of California, Irvine Department of Neurobiology and Behavior, 1203 McGaugh Hall Irvine, CA, 92697, USA
| |
Collapse
|
85
|
Fulton SL, Mitra S, Lepack AE, Martin JA, Stewart AF, Converse J, Hochstetler M, Dietz DM, Maze I. Histone H3 dopaminylation in ventral tegmental area underlies heroin-induced transcriptional and behavioral plasticity in male rats. Neuropsychopharmacology 2022; 47:1776-1783. [PMID: 35094023 PMCID: PMC9372029 DOI: 10.1038/s41386-022-01279-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 01/10/2022] [Accepted: 01/14/2022] [Indexed: 12/14/2022]
Abstract
Persistent transcriptional events in ventral tegmental area (VTA) and other reward relevant brain regions contribute to enduring behavioral adaptations that characterize substance use disorder. Recent data from our laboratory indicate that aberrant accumulation of the newly discovered histone post-translational modification (PTM), H3 dopaminylation at glutamine 5 (H3Q5dop), contributes significantly to cocaine-seeking behavior following prolonged periods of abstinence. It remained unclear, however, whether this modification is important for relapse vulnerability in the context of other drugs of abuse, such as opioids. Here, we showed that H3Q5dop plays a critical role in heroin-mediated transcriptional plasticity in midbrain regions, particularly the VTA. In rats undergoing abstinence from heroin self-administration (SA), we found acute and persistent accumulation of H3Q5dop in VTA. Attenuation of H3Q5dop during abstinence induced persistent changes in gene expression programs associated with neuronal signaling and dopaminergic function in heroin abstinence and led to reduced heroin-seeking behavior. Interestingly, the observed changes in molecular pathways after heroin SA showed significant yet reversed overlap with the same genes altered in cocaine SA. These findings establish an essential role for H3Q5dop, and its downstream transcriptional consequences, in heroin-induced functional plasticity in VTA.
Collapse
Affiliation(s)
- Sasha L. Fulton
- grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Swarup Mitra
- grid.273335.30000 0004 1936 9887Department of Pharmacology and Toxicology, Program in Neuroscience, University at Buffalo, Buffalo, NY 14214 USA
| | - Ashley E. Lepack
- grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Jennifer A. Martin
- grid.273335.30000 0004 1936 9887Department of Pharmacology and Toxicology, Program in Neuroscience, University at Buffalo, Buffalo, NY 14214 USA
| | - Andrew F. Stewart
- grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Jacob Converse
- grid.273335.30000 0004 1936 9887Department of Pharmacology and Toxicology, Program in Neuroscience, University at Buffalo, Buffalo, NY 14214 USA
| | - Mason Hochstetler
- grid.273335.30000 0004 1936 9887Department of Pharmacology and Toxicology, Program in Neuroscience, University at Buffalo, Buffalo, NY 14214 USA
| | - David M. Dietz
- grid.273335.30000 0004 1936 9887Department of Pharmacology and Toxicology, Program in Neuroscience, University at Buffalo, Buffalo, NY 14214 USA
| | - Ian Maze
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Howard Hughes Medical Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
86
|
Jing MY, Ding XY, Han X, Zhao TY, Luo MM, Wu N, Li J, Song R. Activation of mesocorticolimbic dopamine projections initiates cue-induced reinstatement of reward seeking in mice. Acta Pharmacol Sin 2022; 43:2276-2288. [PMID: 35217811 PMCID: PMC9433452 DOI: 10.1038/s41401-022-00866-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 01/13/2022] [Indexed: 12/28/2022] Open
Abstract
Drug addiction is characterized by relapse when addicts are re-exposed to drug-associated environmental cues, but the neural mechanisms underlying cue-induced relapse are unclear. In the present study we investigated the role of a specific dopaminergic (DA) pathway from ventral tegmental area (VTA) to nucleus accumbens core (NAcore) in mouse cue-induced relapse. Optical intracranial self-stimulation (oICSS) was established in DAT-Cre transgenic mice. We showed that optogenetic excitation of DA neurons in the VTA or their projection terminals in NAcore, NAshell or infralimbic prefrontal cortex (PFC-IL) was rewarding. Furthermore, activation of the VTA-NAcore pathway alone was sufficient and necessary to induce reinstatement of oICSS. In cocaine self-administration model, cocaine-associated cues activated VTA DA neurons as assessed by intracellular GCaMP signals. Cue-induced reinstatement of cocaine-seeking was triggered by optogenetic stimulation of the VTA-NAcore pathway, and inhibited by chemogenetic inhibition of this pathway. Together, these results demonstrate that cue-induced reinstatement of reward seeking is in part mediated by activation of the VTA-NAcore DA pathway.
Collapse
Affiliation(s)
- Man-Yi Jing
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Xiao-Yan Ding
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Xiao Han
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Tai-Yun Zhao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Min-Min Luo
- National Institute of Biological Sciences, Beijing, 102206, China
| | - Ning Wu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Jin Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Rui Song
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
| |
Collapse
|
87
|
How drug cravings affect metacognitive monitoring in methamphetamine abusers. Addict Behav 2022; 132:107341. [PMID: 35584984 DOI: 10.1016/j.addbeh.2022.107341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 04/24/2022] [Accepted: 04/24/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND OBJECTIVE Metacognitive monitoring refers to the process in which an individual analyzes their own mental state, then monitors and adjusts cognitive activities to achieve a predetermined goal. Recent research has suggested a strong link between metacognition and drug cravings. Conversely, few studies on the impact of metacognitive monitoring on methamphetamine (MA) cravings exist. Thus, this study investigated whether drug cravings would impair MA abusers' metacognitive monitoring and explored the prediction effects of drug cravings. METHOD Seventy MA abusers from the Zhejiang Compulsory Isolation Drug Rehabilitation Center and 65 non-users from the Wenzhou Medical University were recruited for this experimental study. The judgment of learning (JOL) paradigm was used to examine metacognitive monitoring, and cue-induced pictures were used to induce MA abusers' drug cravings. Analysis of covariance (ANCOVA), partial correlation, and regression analysis were performed. RESULTS Compared with non-users, MA abusers had significantly poorer metacognitive monitoring and tended to overestimate their performance. Furthermore, there was a significant correlation between the accuracy of JOLs and drug cravings, which indicated that metacognitive monitoring was weakened by drug cravings with higher cravings imposing more severe impacts. In addition, the regression analysis suggested that drug cravings can predict metacognitive monitoring.
Collapse
|
88
|
Brynildsen JK, Yang K, Lemchi C, Dani JA, De Biasi M, Blendy JA. A common SNP in Chrna5 enhances morphine reward in female mice. Neuropharmacology 2022; 218:109218. [PMID: 35973602 DOI: 10.1016/j.neuropharm.2022.109218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 08/01/2022] [Accepted: 08/08/2022] [Indexed: 11/25/2022]
Abstract
The single nucleotide polymorphism (SNP) D398N (rs16969968) in CHRNA5, the gene encoding the α5 subunit of the nicotinic acetylcholine receptors (nAChR), has been associated with both nicotine and opiate dependence in human populations. Expression of this SNP on presynaptic VTA dopaminergic (DA) neurons is known to cause a reduction in calcium signaling, leading to alterations in transmitter signaling and altered responses to drugs of abuse. To examine the impact of the Chrna5 SNP on opiate reward and underlying dopaminergic mechanisms, mice harboring two copies of the risk-associated allele (Chrna5 A/A) at a location equivalent to human rs16969968 were generated via CRISPR/cas9 genome editing. We sought to determine whether Chrna5 A/A mice show differences in sensitivity to rewarding properties of morphine using the conditioned place preference paradigm. When mice were tested two weeks after conditioning, female Chrna5 A/A mice showed significantly enhanced preference for the morphine-paired chamber relative to WT females, suggesting that this genotype may enhance opioid reward specifically in females. In contrast, Chrna5 genotype had no effect on locomotor sensitization in male or female mice. Relative to WT females, peak amplitude of ACh-gated currents recorded from VTA DA neurons in Chrna5 A/A females was potentiated 1 day after conditioning with morphine. Increased FOS expression was also observed in Chrna5 A/A mice relative to WT mice following exposure to the morphine CPP chamber. We propose that impaired α5 nAChR subunit function alters DA neuron response following repeated morphine exposures, and that this early cellular response could contribute to enhanced opiate reward two weeks after conditioning.
Collapse
Affiliation(s)
| | | | - Crystal Lemchi
- Department of Systems Pharmacology and Translational Therapeutics, USA
| | | | - Mariella De Biasi
- Department of Neuroscience, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Julie A Blendy
- Department of Systems Pharmacology and Translational Therapeutics, USA.
| |
Collapse
|
89
|
Kang T, Ding X, Zhao J, Li X, Xie R, Jiang H, He L, Hu Y, Liang J, Zhou G, Huo X. Influence of improved behavioral inhibition on decreased cue-induced craving in heroin use disorder: A preliminary intermittent theta burst stimulation study. J Psychiatr Res 2022; 152:375-383. [PMID: 35797913 DOI: 10.1016/j.jpsychires.2022.06.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Impaired behavioral inhibition is a critical factor in drug addiction and relapse. Repetitive transcranial magnetic stimulation (rTMS) reduces the craving of heroin-addicted individuals for drug-related cues. However, it is unclear whether this technique also improves impaired behavioral inhibition and how improved behavioral inhibition affects craving. OBJECTIVE The intermittent theta-burst stimulation (iTBS) has been recently shown to be non-inferior relative to rTMS for depression. Here, we aim to investigate the effect of iTBS on heroin-addicted individuals' behavioral inhibition and cue-induced craving and the relationship between the alteration of behavioral inhibition and craving. METHOD 42 of 56 initially recruited individuals with the heroin-use disorder in the abstinent-course treatment were randomized to undergo active or sham iTBS to the left dorsolateral prefrontal cortex and received three daily iTBS treatments for 10 consecutive days. We measured participants' performance during a two-choice oddball task (80% standard and 20% deviant trials) and heroin-related cue-induced craving before and immediately after treatment. RESULTS The group that received active iTBS showed significantly improved two-choice oddball task performance after 10 days of intervention compared to both pre-intervention and the group who received sham iTBS. Similarly, a significant reduction in cue-induced craving was observed after following the intervention in the active iTBS group but not the sham iTBS group. The moderation model indicated that iTBS categories play a significant moderating role in the relationship between accuracy cost changing and altered cue-induced craving. CONCLUSIONS The iTBS treatment protocol positively affects behavioral inhibition in patients with heroin addiction. Improvements in behavioral inhibition can substantially reduce craving.
Collapse
Affiliation(s)
- Tiejun Kang
- School of Psychology, Northwest Normal University, Lanzhou, Gansu, China; Key Laboratory of Behavioral and Mental Health of Gansu Province, Lanzhou, Gansu, China.
| | - Xiaobin Ding
- School of Psychology, Northwest Normal University, Lanzhou, Gansu, China; Key Laboratory of Behavioral and Mental Health of Gansu Province, Lanzhou, Gansu, China.
| | - Jing Zhao
- School of Psychology, Northwest Normal University, Lanzhou, Gansu, China
| | - Xiaoyan Li
- School of Psychology, Northwest Normal University, Lanzhou, Gansu, China
| | - Renqian Xie
- Lanzhou Hospital of Addiction Rehabilitation, Lanzhou, Gansu, China
| | - Heng Jiang
- School of Psychology, Northwest Normal University, Lanzhou, Gansu, China
| | - Liang He
- School of Psychology, Northwest Normal University, Lanzhou, Gansu, China
| | - Yajuan Hu
- School of Psychology, Northwest Normal University, Lanzhou, Gansu, China
| | - Jingjing Liang
- School of Psychology, Northwest Normal University, Lanzhou, Gansu, China
| | - Guifen Zhou
- Lanzhou Hospital of Addiction Rehabilitation, Lanzhou, Gansu, China
| | - Xiao Huo
- Lanzhou Hospital of Addiction Rehabilitation, Lanzhou, Gansu, China
| |
Collapse
|
90
|
Inbar K, Levi LA, Kupchik YM. Cocaine induces input and cell-type-specific synaptic plasticity in ventral pallidum-projecting nucleus accumbens medium spiny neurons. Neuropsychopharmacology 2022; 47:1461-1472. [PMID: 35121830 PMCID: PMC9205871 DOI: 10.1038/s41386-022-01285-6] [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: 09/23/2021] [Revised: 01/23/2022] [Accepted: 01/24/2022] [Indexed: 11/09/2022]
Abstract
Cocaine use and abstinence induce long-term synaptic alterations in the excitatory input to nucleus accumbens (NAc) medium spiny neurons (MSNs). The NAc regulates reward-related behaviors through two parallel projections to the ventral pallidum (VP)-originating in D1 or D2-expressing MSNs (D1-MSNs→VP; D2-MSNs→VP). The activity of these projections depends on their excitatory synaptic inputs, but it is not known whether and how abstinence from cocaine affects the excitatory transmission to D1-MSNs→VP and D2-MSNs→VP. Here we examined different forms of cocaine-induced synaptic plasticity in the inputs from the basolateral amygdala (BLA) and medial prefrontal cortex (mPFC) to NAc D1-MSNs→VP and putative D2-MSNs→VP (pD2-MSNs→VP) in the core and shell subcompartments of the NAc. We used the whole-cell patch-clamp technique to record excitatory postsynaptic currents from D1-tdTomato mice injected with ChR2 in either the BLA or the mPFC and retrograde tracer (RetroBeads) in the VP. We found that cocaine conditioned place preference (CPP) followed by abstinence potentiated the excitatory input from the BLA and mPFC to both D1-MSNs→VP and pD2-MSNs→VP. Interestingly, while the strengthening of the inputs to D1-MSNs→VP was of postsynaptic origin and manifested as increased AMPA to NMDA ratio, in pD2-MSNs→VP plasticity was predominantly presynaptic and was detected as changes in the paired-pulse ratio and coefficient of variation. Lastly, some of the changes were sex-specific. Overall our data show that abstinence from cocaine changes the excitatory inputs to both D1-MSNs→VP and pD2-MSNs→VP but with different mechanisms. This may help understand how circuits converging into the VP change after cocaine exposure.
Collapse
Affiliation(s)
- Kineret Inbar
- grid.9619.70000 0004 1937 0538Department of Medical Neurobiology, Faculty of Medicine, The Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Jerusalem, 9112102 Israel
| | - Liran A. Levi
- grid.9619.70000 0004 1937 0538Department of Medical Neurobiology, Faculty of Medicine, The Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Jerusalem, 9112102 Israel
| | - Yonatan M. Kupchik
- grid.9619.70000 0004 1937 0538Department of Medical Neurobiology, Faculty of Medicine, The Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Jerusalem, 9112102 Israel
| |
Collapse
|
91
|
Long-term exercise at different intensities can reduce the inflammatory response in the brains of methamphetamine-treated mice. Biochem Biophys Res Commun 2022; 613:201-206. [DOI: 10.1016/j.bbrc.2022.05.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 05/11/2022] [Indexed: 11/02/2022]
|
92
|
Chow JJ, Beacher NJ, Chabot JM, Oke M, Venniro M, Lin DT, Shaham Y. Characterization of operant social interaction in rats: effects of access duration, effort, peer familiarity, housing conditions, and choice between social interaction vs. food or remifentanil. Psychopharmacology (Berl) 2022; 239:2093-2108. [PMID: 35230469 PMCID: PMC10724845 DOI: 10.1007/s00213-022-06064-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/06/2022] [Indexed: 01/02/2023]
Abstract
RATIONALE AND OBJECTIVE Social factors play a critical role in drug addiction. We recently showed that rats will abstain from methamphetamine, cocaine, heroin, and remifentanil self-administration when given a choice between the addictive drug and operant social interaction. Here, we further characterized operant social interaction by determining the effects of access duration, effort, peer familiarity, and housing conditions. We also determined choice between social interaction vs. palatable food or remifentanil. METHODS We first trained single-housed male and female rats to lever-press for social interaction with a sex- and age-matched peer. Next, we determined effects of access duration (3.75 to 240 s), effort (increasing fixed-ratio schedule requirements or progressive ratio schedule), peer familiarity (familiar vs. unfamiliar), and housing conditions (single vs. paired housing) on social self-administration. We also determined choice between social interaction vs. palatable food pellets or intravenous remifentanil (0, 1, 10 µg/kg/infusion). RESULTS Increasing access duration to a peer decreased social self-administration under fixed ratio but not progressive ratio schedule; the rats showed similar preference for short vs. long access duration. Social self-administration under different fixed ratio requirements was higher in single-housed than in paired-housed rats and higher for a familiar vs. unfamiliar partner in single-housed but not paired-housed rats. Response rates of food-sated rats under increasing fixed-ratio requirements were higher for palatable food than for social interaction. The rats strongly preferred palatable food over social interaction and showed dose-dependent preference for social interaction vs. remifentanil. CONCLUSIONS We identified parameters influencing the reinforcing effects of operant social interaction and introduce a choice procedure sensitive to remifentanil self-administration dose.
Collapse
Affiliation(s)
| | | | - Jules M Chabot
- Intramural Research Program, NIDA, NIH, Baltimore, MD, USA
| | - Marvellous Oke
- Intramural Research Program, NIDA, NIH, Baltimore, MD, USA
| | - Marco Venniro
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Da-Ting Lin
- Intramural Research Program, NIDA, NIH, Baltimore, MD, USA
| | - Yavin Shaham
- Intramural Research Program, NIDA, NIH, Baltimore, MD, USA.
| |
Collapse
|
93
|
Ferranti AS, Johnson KA, Winder DG, Conn PJ, Joffe ME. Prefrontal cortex parvalbumin interneurons exhibit decreased excitability and potentiated synaptic strength after ethanol reward learning. Alcohol 2022; 101:17-26. [PMID: 35227826 PMCID: PMC9117490 DOI: 10.1016/j.alcohol.2022.02.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/21/2022] [Accepted: 02/21/2022] [Indexed: 12/13/2022]
Abstract
The prefrontal cortex (PFC) is intimately associated with behavioral characteristics of alcohol use disorders, including high motivation to drink and difficulty with moderation. Thus, continued mechanistic research investigating PFC cells and targets altered by ethanol experiences should inform translational efforts to craft new, efficacious treatments. Inhibitory interneurons expressing parvalbumin (PV-INs) comprise only a minor fraction of cells within the PFC, yet these cells are indispensable for coordinating PFC ensemble function, oscillatory activity, and subcortical output. Based on this, PV-INs represent an exciting target for the rational design of breakthrough treatments for alcohol use disorders. Here, we assessed experience-dependent physiological adaptations via ethanol place conditioning. By manipulating the timing of administration relative to conditioning sessions, equivalent ethanol exposure can form either rewarding or aversive memories in different individuals. Here, we found that female mice and male mice on a C57BL/6J background display conditioned place preference (CPP) or aversion (CPA) to an intoxicating dose of ethanol (2 g/kg, intraperitoneal [i.p.]) without overt differences between sexes. Ethanol reward learning was associated with decreased PV-IN excitability in deep layer prelimbic PFC, whereas PV-INs from CPA mice were not different from controls. Furthermore, PV-INs from mice in the CPP group, but not the CPA group, displayed potentiated excitatory synaptic strength that emerged during 1 week of abstinence. Taken together, these findings illustrate that synaptic and intrinsic adaptations associated with ethanol can depend on an individual's experience. These studies provide further context and support for PFC PV-INs as intriguing targets for modulating alcohol associations.
Collapse
Affiliation(s)
| | - Kari A. Johnson
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Danny G. Winder
- Department of Pharmacology, Vanderbilt University, Nashville, TN
- Vanderbilt Center for Addiction Research, Nashville, TN
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - P. Jeffrey Conn
- Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN
- Vanderbilt Center for Addiction Research, Nashville, TN
| | - Max E. Joffe
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
94
|
Feng Z, Wu Q, Wu L, Zeng T, Yuan J, Wang X, Kang C, Yang J. Effect of High-Frequency Repetitive Transcranial Magnetic Stimulation on Visual Selective Attention in Male Patients With Alcohol Use Disorder After the Acute Withdrawal. Front Psychiatry 2022; 13:869014. [PMID: 35573341 PMCID: PMC9098796 DOI: 10.3389/fpsyt.2022.869014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/25/2022] [Indexed: 11/20/2022] Open
Abstract
Objective To investigate the effect of high-frequency repetitive transcranial magnetic stimulation (rTMS) on attention cue reactivity in male patients with alcohol use disorder (AUD) after acute withdrawal. Methods A total of 90 male patients with AUD who were hospitalized were enrolled and divided into study and waiting groups by a random number table. During the study, 18 patients dropped out. After the alcohol withdrawal symptoms were eliminated, the study group received high-frequency rTMS at 10 Hz for 14 consecutive days, and the waiting group was administrated by sham rTMS. All subjects were evaluated for attention cue reactivity, impulsiveness, cognitive function by oddball paradigm, Barratt Impulsiveness Scale version II (BIS-II), and the Montreal Cognitive Assessment (MoCA) at baseline and after true or sham rTMS. Results 1. There was no significant difference between the study and the waiting groups regarding the drinking level, cognition level, and demographic data at baseline. 2. In the oddball paradigm, both for alcohol-related and non-alcohol-related cues, the response times were significantly shorter in the study group after rTMS treatment than in the waiting-for-treatment group, either between the two groups or within the study group. There was no significant difference in the accuracy rate for alcohol-related and non-alcohol-related cues between the two groups or within the study group after rTMS intervention. 3. The total score of MoCA was significantly increased, and the total score of BIS-II was significantly decreased in the study group after rTMS treatment, either between the two groups or within the study group. Conclusion The results suggested that high-frequency rTMS could improve the attention bias of alcohol-related cues and impulsivity for patients with AUD.
Collapse
Affiliation(s)
- Zuxing Feng
- Department of Psychiatry, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qiao Wu
- Department of Psychiatry, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Li Wu
- Department of Substance use Disorders, the Psychiatry Hospital of Yunnan, Kunming, China
| | - Tingting Zeng
- Department of Psychiatry, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jing Yuan
- Department of Psychiatry, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xin Wang
- Department of Psychiatry, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Chuanyuan Kang
- Department of Psychosomatic Medicine, Tongji University School of Medicine, Shanghai East Hospital, Shanghai, China
| | - Jianzhong Yang
- Department of Psychiatry, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
95
|
Darrag HM, Almuhanna HT, Hakami EH. Secondary Metabolites in Basil, Bio-Insecticide, Inhibition Effect, and In Silico Molecular Docking against Proteolytic Enzymes of the Red Palm Weevil (Rhynchophorus ferrugineus). PLANTS 2022; 11:plants11081087. [PMID: 35448815 PMCID: PMC9027599 DOI: 10.3390/plants11081087] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/13/2022] [Accepted: 04/15/2022] [Indexed: 11/18/2022]
Abstract
The purpose of this work was to determine the secondary metabolites generated by O. basilicum cell suspensions, as well as their insecticide and inhibitory activity against R. ferrugineus. The growth kinetics with inoculation Verticillium dahliae were determined and identified using LC-MS. Determination of total phenolic components (TFC), flavonoids (TF), and condensed tannins (TCT) were measured. Insecticidal activity of O. basilicum extract against R. ferrugineus (larva and adult) and proteolytic enzymes activity were assessed (in vitro and in vivo). The O.basilicum extract had an LC50 of 1238 µg/mL and an LD50 of 13.4 µg/larva. The LC50 of chicoric acid, ursolic acid, salvigenin, quercetin-3-O-rutinoside, rosmarinyl glucoside, and nepetoidin B demonstrated activity at an LC50 of 1132, 1167, 1189, 1214, 1275, and 1317 µg/mL, respectively. Chicoric acid, salvigenin, nepetoidin B, and rosmarinic acid demonstrated an LD50 activity of 10.23, 11.4, 11.9, and 12.4 µg/larva, respectively. The active extract of O. basilicum inhibited total protease, trypsin-like serine proteinases, elastase, cysteine, and metalloprotease activity with an IC50 (in vitro) of 119.4, 91, 102.4, 76.4, and 52.4 µg/mL, respectively. In silico studies of compounds were conducted, such as molecular docking and ADMET analysis. The study proposes using an efficient cell suspension technique to produce O. basilicum extract containing active secondary metabolites and accessible using as bio-insecticide.
Collapse
Affiliation(s)
- Hossam Moustafa Darrag
- Department of Research and Training, Research and Training Station, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- Pesticide Chemistry and Technology Department, Faculty of Agriculture, Alexandria University, Alexandria 21545, Egypt
- Correspondence: ; Tel.: +966-508299027
| | - Hani Taher Almuhanna
- Research and Training Station, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (H.T.A.); (E.H.H.)
| | - Emadaldeen Hamad Hakami
- Research and Training Station, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (H.T.A.); (E.H.H.)
| |
Collapse
|
96
|
Khajehpour H, Parvaz MA, Kouti M, Hosseini Rafsanjani T, Ekhtiari H, Bakht S, Noroozi A, Makkiabadi B, Mahmoodi M. Effects of Transcranial Direct Current Stimulation on Attentional Bias to Methamphetamine Cues and Its Association With EEG-Derived Functional Brain Network Topology. Int J Neuropsychopharmacol 2022; 25:631-644. [PMID: 35380672 PMCID: PMC9380716 DOI: 10.1093/ijnp/pyac018] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/04/2022] [Accepted: 03/30/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Although transcranial direct current stimulation (tDCS) has shown to potentially mitigate drug craving and attentional bias to drug-related stimuli, individual differences in such modulatory effects of tDCS are less understood. In this study, we aimed to investigate a source of the inter-subject variability in the tDCS effects that can be useful for tDCS-based treatments of individuals with methamphetamine (MA) use disorder (IMUD). METHODS Forty-two IMUD (all male) were randomly assigned to receive a single-session of either sham or real bilateral tDCS (anodal right/cathodal left) over the dorsolateral prefrontal cortex. The tDCS effect on MA craving and biased attention to drug stimuli were investigated by quantifying EEG-derived P3 (a measure of initial attentional bias) and late positive potential (LPP; a measure of sustained motivated attention) elicited by these stimuli. To assess the association of changes in P3 and LPP with brain connectivity network (BCN) topology, the correlation between topology metrics, specifically those related to the efficiency of information processing, and the tDCS effect was investigated. RESULTS The P3 amplitude significantly decreased following the tDCS session, whereas the amplitudes increased in the sham group. The changes in P3 amplitudes were significantly correlated with communication efficiency measured by BCN topology metrics (r = -0.47, P = .03; r = -0.49, P = .02). There was no significant change in LPP amplitude due to the tDCS application. CONCLUSIONS These findings validate that tDCS mitigates initial attentional bias, but not the sustained motivated attention, to MA stimuli. Importantly, however, results also show that the individual differences in the effects of tDCS may be underpinned by communication efficiency of the BCN topology, and therefore, these BCN topology metrics may have the potential to robustly predict the effectiveness of tDCS-based interventions on MA craving and attentional bias to MA stimuli among IMUD.
Collapse
Affiliation(s)
- Hassan Khajehpour
- Correspondence: Hassan Khajehpour, PhD, Department of Physics, Concordia University, Richard J. Renaud Science Complex, Loyola Campus, 7141 Sherbrooke St. W., Montreal, H4B 1R6, Quebec, Canada ()
| | - Muhammad A Parvaz
- Department of Psychiatry, Icahn School of Medicine, Mount Sinai, New York, USA,Department of Neuroscience, Icahn School of Medicine, Mount Sinai, New York, USA
| | - Mayadeh Kouti
- Department of Electrical Engineering, Shohadaye Hoveizeh Campus of Technology, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | | | - Hamed Ekhtiari
- Laureate Institute for Brain Research, Tulsa, Oklahoma, USA,Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Bakht
- Department of Cognitive Psychology, Institute for Cognitive Sciences Studies, Tehran, Iran
| | - Alireza Noroozi
- Neuroscience and Addiction Studies Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran (Dr Noroozi)
| | - Bahador Makkiabadi
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran,Iran,Research Center for Biomedical Technology and Robotics, Institute of Advanced Medical Technologies, Tehran University of Medical Sciences, Tehran,Iran
| | - Maryam Mahmoodi
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran,Iran,Research Center for Biomedical Technology and Robotics, Institute of Advanced Medical Technologies, Tehran University of Medical Sciences, Tehran,Iran
| |
Collapse
|
97
|
Singh A, Xie Y, Davis A, Wang ZJ. Early social isolation stress increases addiction vulnerability to heroin and alters c-Fos expression in the mesocorticolimbic system. Psychopharmacology (Berl) 2022; 239:1081-1095. [PMID: 34997861 DOI: 10.1007/s00213-021-06024-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 11/04/2021] [Indexed: 01/02/2023]
Abstract
RATIONALE Adverse psychosocial factors during early childhood or adolescence compromise neural structure and brain function, inducing susceptibility for many psychiatric disorders such as substance use disorder. Nevertheless, the mechanisms underlying early life stress-induced addiction vulnerability is still unclear, especially for opioids. OBJECTIVES To address this, we used a mouse heroin self-administration model to examine how chronic early social isolation (ESI) stress (5 weeks, beginning at weaning) affects the behavioral and neural responses to heroin during adulthood. RESULTS We found that ESI stress did not alter the acquisition for sucrose or heroin self-administration, nor change the motivation for sucrose on a progressive ratio schedule. However, ESI stress induced an upward shift of heroin dose-response curve in female mice and increased motivation and seeking for heroin in both sexes. Furthermore, we examined the neuronal activity (measured by c-Fos expression) within the key brain regions of the mesocorticolimbic system, including the prelimbic cortex (PrL), infralimbic cortex (IL), nucleus accumbens (NAc) core and shell, caudate putamen, and ventral tegmental area (VTA). We found that ESI stress dampened c-Fos expression in the PrL, IL, and VTA after 14-day forced abstinence, while augmented the neuronal responses to heroin-predictive context and cue in the IL and NAc core. Moreover, ESI stress disrupted the association between c-Fos expression and attempted infusions during heroin-seeking test in the PrL. CONCLUSIONS These data indicate that ESI stress leads to increased seeking and motivation for heroin, and this may be associated with distinct changes in neuronal activities in different subregions of the mesocorticolimbic system.
Collapse
Affiliation(s)
- Archana Singh
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, 1251 Wescoe Hall Drive, Lawrence, KS, 66045, USA
| | - Yang Xie
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, 1251 Wescoe Hall Drive, Lawrence, KS, 66045, USA
| | - Ashton Davis
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, 1251 Wescoe Hall Drive, Lawrence, KS, 66045, USA
| | - Zi-Jun Wang
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, 1251 Wescoe Hall Drive, Lawrence, KS, 66045, USA.
| |
Collapse
|
98
|
Gillespie A, Mayberry HL, Wimmer ME, Sillivan SE. microRNA expression levels in the nucleus accumbens correlate with morphine-taking but not morphine-seeking behaviour in male rats. Eur J Neurosci 2022; 55:1742-1755. [PMID: 35320877 PMCID: PMC9314918 DOI: 10.1111/ejn.15650] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/04/2022] [Accepted: 03/15/2022] [Indexed: 12/01/2022]
Abstract
A powerful motivation to seek opioids remains after drug cessation and intensifies during extended periods of abstinence. Unfortunately, biomarkers associated with continued drug seeking have not been described. Moreover, previous studies have focused on the effects of early abstinence with little exploration into the long-term drug-induced mechanisms that occur after extended abstinence. Here we demonstrated that 30 days (D) of forced abstinence results in a time-dependent increase in morphine seeking in a rat model of morphine self-administration (SA). We measured expression of known drug-responsive microRNAs (miRNAs) in the nucleus accumbens, an area critical for reward-related plasticity, during early or late abstinence in animals that underwent either a cue-induced relapse test or no relapse test. miRNAs are small noncoding RNAs that represent suitable biomarker candidates due to their long-lasting nature. mir-32-5p levels during early abstinence negatively correlated with active lever pressing in both cue-exposed and cue-naïve animals. mir-1298-5p positively correlated with drug SA history after a relapse test during late abstinence. When animals underwent acute abstinence with no relapse test, mir-1298-5p correlated with drug infusions and active lever pressing during SA. In late abstinence with no relapse test, mir-137-3p negatively correlated with drug infusions. Regulation of mir-32-5p target genes and significant correlation of target gene mRNA with mir-32-5p was observed after abstinence. These results indicate that lasting regulation of miRNA expression is associated with drug intake following morphine SA. In addition, we conclude that the miRNA profile undergoes regulation from early to late abstinence and miRNA expression may indicate past drug history.
Collapse
Affiliation(s)
- Aria Gillespie
- Center for Substance Abuse ResearchTemple UniversityPhiladelphiaPennsylvaniaUSA,Department of Neural SciencesTemple UniversityPhiladelphiaPAUSA
| | - Hannah L. Mayberry
- Department of Psychology and Neuroscience Program, College of Liberal ArtsTemple UniversityPhiladelphiaPennsylvaniaUSA
| | - Mathieu E. Wimmer
- Department of Psychology and Neuroscience Program, College of Liberal ArtsTemple UniversityPhiladelphiaPennsylvaniaUSA
| | - Stephanie E. Sillivan
- Center for Substance Abuse ResearchTemple UniversityPhiladelphiaPennsylvaniaUSA,Department of Neural SciencesTemple UniversityPhiladelphiaPAUSA
| |
Collapse
|
99
|
Alegre-Zurano L, Berbegal-Sáez P, Luján MÁ, Cantacorps L, Martín-Sánchez A, García-Baos A, Valverde O. Cannabidiol decreases motivation for cocaine in a behavioral economics paradigm but does not prevent incubation of craving in mice. Biomed Pharmacother 2022; 148:112708. [DOI: 10.1016/j.biopha.2022.112708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 02/02/2022] [Accepted: 02/07/2022] [Indexed: 11/26/2022] Open
|
100
|
Opioid and Sucrose Craving Are Accompanied by Unique Behavioral and Affective Profiles after Extended Abstinence in Male and Female Rats. eNeuro 2022; 9:ENEURO.0515-21.2022. [PMID: 35241453 PMCID: PMC9007407 DOI: 10.1523/eneuro.0515-21.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/17/2022] [Accepted: 02/24/2022] [Indexed: 11/21/2022] Open
Abstract
Incubation of craving refers to the intensification of drug-seeking behavior in response to reward-paired cues over the course of abstinence. In rodents, craving and drug-seeking behaviors have been measured by an increase in lever pressing in the absence of reinforcer availability in response to cue presentations. However, craving in rodents is difficult to define and little is known about the behavioral signatures that accompany increased drug-seeking behavior measured by lever pressing. The affective components of relapse are also important, but understudied in rodents. Hormonal fluctuations influence craving for psychostimulants, but little is known about the impact of the estrous cycle on opioid-seeking behavior. This study sought to delineate the behavioral and affective signatures associated with craving, and to examine the influence of the female estrous cycle on craving. Male and female rats underwent 10 d of intravenous opioid self-administration. Separate cohorts of control rats self-administered oral sucrose, a natural nondrug reward. Cue-induced seeking tests were conducted after 1 or 30d of forced abstinence. These sessions were recorded and scored for overall locomotion, instances of sniffing, grooming, or hyperactivity. Ultrasonic vocalizations (USVs) were also recorded to determine affective profiles that accompany opioid seeking. Although active lever presses and overall locomotion increased unanimously over extended abstinence from heroin and sucrose, a sex- and reinforcer-specific behavioral and affective signature of craving emerged. Furthermore, although the female estrous cycle did not affect taking or seeking, it appears to influence more granular behaviors.
Collapse
|