1
|
Xu Y, Lin Y, Yu M, Zhou K. The nucleus accumbens in reward and aversion processing: insights and implications. Front Behav Neurosci 2024; 18:1420028. [PMID: 39184934 PMCID: PMC11341389 DOI: 10.3389/fnbeh.2024.1420028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/26/2024] [Indexed: 08/27/2024] Open
Abstract
The nucleus accumbens (NAc), a central component of the brain's reward circuitry, has been implicated in a wide range of behaviors and emotional states. Emerging evidence, primarily drawing from recent rodent studies, suggests that the function of the NAc in reward and aversion processing is multifaceted. Prolonged stress or drug use induces maladaptive neuronal function in the NAc circuitry, which results in pathological conditions. This review aims to provide comprehensive and up-to-date insights on the role of the NAc in motivated behavior regulation and highlights areas that demand further in-depth analysis. It synthesizes the latest findings on how distinct NAc neuronal populations and pathways contribute to the processing of opposite valences. The review examines how a range of neuromodulators, especially monoamines, influence the NAc's control over various motivational states. Furthermore, it delves into the complex underlying mechanisms of psychiatric disorders such as addiction and depression and evaluates prospective interventions to restore NAc functionality.
Collapse
Affiliation(s)
| | | | | | - Kuikui Zhou
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| |
Collapse
|
2
|
Andersen FD, Joca S, Hvingelby V, Arjmand S, Pinilla E, Steffensen SC, Simonsen U, Andersen CU. Combined effects of quetiapine and opioids: A study of autopsy cases, drug users and sedation in rats. Addict Biol 2022; 27:e13214. [PMID: 36001431 PMCID: PMC9541371 DOI: 10.1111/adb.13214] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/17/2022] [Accepted: 07/12/2022] [Indexed: 11/28/2022]
Abstract
Fatal opioid poisonings often involve methadone or morphine. This study aimed to elucidate if quetiapine, a widely used sedative antipsychotic medication, may increase the risk of fatal opioid poisoning by additive inhibitory effects on the central nervous system. We used data from 323 cases of fatal methadone or/and morphine poisonings autopsied from 2013 to 2020, a survey of 34 drug users, and performed blinded placebo‐controlled studies in 75 Flinders Resistant Line rats receiving three cumulative intraperitoneal doses of vehicle, methadone (2.5, 10 and 15 mg/kg), morphine (3.75, 15 and 22.5 mg/kg), quetiapine (3, 10 and 30 mg/kg) or quetiapine combined with methadone or morphine. Quetiapine was detected in 20.4% of fatal opioid poisonings with a significantly increased frequency over time, primarily in low or therapeutic concentrations, and was not associated with methadone or morphine concentrations. Use of quetiapine, most commonly in low‐to‐moderate doses to obtain a sleep‐inducing or tranquillizing effect, was reported by 67.6% of survey respondents. In the animal studies, a significant impairment of sedation score, performance on the rotarod and open field mobility was observed in all treatment groups compared with vehicle. However, the effect of quetiapine plus the opioid was not significantly different from that of the opioid alone. Thus, no additive sedative effects were observed in rats. Our results suggest that quetiapine is more often an innocent bystander than a contributor to fatal opioid poisoning. However, the combined effects on other parameters, including blood pressure, cardiac rhythm and respiratory rate, need investigation.
Collapse
Affiliation(s)
| | - Sâmia Joca
- Department of Biomedicine Aarhus University Aarhus Denmark
| | - Victor Hvingelby
- Department of Clinical Medicine – Nuclear Medicine and PET Aarhus University Aarhus Denmark
| | - Shokouh Arjmand
- Translational Neuropsychiatry Unit, Department of Clinical Medicine Aarhus University Aarhus Denmark
| | | | - Simon Comerma Steffensen
- Department of Biomedicine Aarhus University Aarhus Denmark
- Department of Biomedical Sciences/Animal Physiology, Faculty of Veterinary Central University of Venezuela
| | - Ulf Simonsen
- Department of Biomedicine Aarhus University Aarhus Denmark
| | - Charlotte Uggerhøj Andersen
- Department of Forensic Medicine Aarhus University Hospital Aarhus Denmark
- Department of Biomedicine Aarhus University Aarhus Denmark
- Department of Clinical Pharmacology Aarhus University Hospital Aarhus Denmark
| |
Collapse
|
3
|
Yevoo PE, Maffei A. Women in Neuroscience: Four Women’s Contributions to Science and Society. Front Integr Neurosci 2022; 15:810331. [PMID: 35153689 PMCID: PMC8825414 DOI: 10.3389/fnint.2021.810331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 12/27/2021] [Indexed: 11/24/2022] Open
Abstract
There has been increased cognizance of gender inequity and the importance of an inclusive and diverse approach to scientific research in recent years. However, the innovative work of women scientists is still undervalued based on reports of fewer women in leadership positions, limited citations of research spearheaded by women, reduced federal grant awards, and lack of recognition. Women have been involved in trailblazing work that paved the way for contemporary scientific inquiry. The strides made in current neuroscience include contributions from women who deserve more recognition. In this review, we discuss the work of four women whose groundbreaking scientific work has made ineffaceable marks in the neuroscience field. These women are pioneers of research and innovators and, in addition, contribute to positive change that bolsters the academic community and society. This article celebrates these women scientists, their substantial impacts in neuroscience, and the positive influence of their work on advancing society and culture.
Collapse
Affiliation(s)
- Priscilla E. Yevoo
- Department of Neurobiology and Behavior, SUNY – Stony Brook, Stony Brook, NY, United States
- Graduate Program in Neuroscience, SUNY – Stony Brook, Stony Brook, NY, United States
- *Correspondence: Priscilla E. Yevoo,
| | - Arianna Maffei
- Department of Neurobiology and Behavior, SUNY – Stony Brook, Stony Brook, NY, United States
- Graduate Program in Neuroscience, SUNY – Stony Brook, Stony Brook, NY, United States
| |
Collapse
|
4
|
The role of the nucleus accumbens and ventral pallidum in feeding and obesity. Prog Neuropsychopharmacol Biol Psychiatry 2021; 111:110394. [PMID: 34242717 DOI: 10.1016/j.pnpbp.2021.110394] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 05/31/2021] [Accepted: 06/29/2021] [Indexed: 02/04/2023]
Abstract
Obesity is a growing global epidemic that stems from the increasing availability of highly-palatable foods and the consequent enhanced calorie consumption. Extensive research has shown that brain regions that are central to reward seeking modulate feeding and evidence linking obesity to pathology in such regions have recently started to accumulate. In this review we focus on the contribution of two major interconnected structures central to reward processing, the nucleus accumbens and the ventral pallidum, to obesity. We first review the known literature linking these structures to feeding behavior, then discuss recent advances connecting pathology in the nucleus accumbens and ventral pallidum to obesity, and finally examine the similarities and differences between drug addiction and obesity in the context of these two structures. The understanding of how pathology in brain regions involved in reward seeking and consumption may drive obesity and how mechanistically similar obesity and addiction are, is only now starting to be revealed. We hope that future research will advance knowledge in the field and open new avenues to studying and treating obesity.
Collapse
|
5
|
Maguire DR, Minervini V. Interactions between opioids and stimulants: Behavioral pharmacology of abuse-related effects. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 93:1-33. [PMID: 35341563 DOI: 10.1016/bs.apha.2021.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Opioid abuse continues to be a significant public health challenge, with rates of opioid-related overdose deaths increasing continuously over the last two decades. There also has been a sharp increase in overdose deaths involving stimulant drugs, primarily cocaine and methamphetamine. Recent estimates indicate a high prevalence of co-use of opioids and stimulants, which is a particularly complex problem. Behavioral pharmacology research over the last few decades has characterized interactions between opioids and stimulants as well as evaluated potential treatments. This chapter describes interactions between opioids and stimulants, with a focus on pre-clinical studies of abuse-related behavioral effects using self-administration, reinstatement, drug discrimination, place conditioning, and intracranial self-stimulation paradigms in laboratory animals. In general, the literature provides substantial evidence of mutual enhancement between opioids and stimulants for abuse-related effects, although such results are not ubiquitous. Enhanced abuse-related effects could manifest in many ways including engaging in drug seeking and taking behaviors with greater persistence, effort, and motivation and/or increased likelihood of relapse. Moreover, studies on opioid/stimulant combinations set the stage for evaluating potential treatments for polysubstance use. Behavioral pharmacology research has proven invaluable for elucidating these relationships using rigorous experimental designs and quantitative analyses of pharmacological and behavioral data.
Collapse
Affiliation(s)
- David R Maguire
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States; Addiction Research Treatment and Training Center of Excellence, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States.
| | - Vanessa Minervini
- Department of Psychological Science, Creighton University, Omaha, NE, United States
| |
Collapse
|
6
|
Seaman RW, Collins GT. Impact of Morphine Dependence and Withdrawal on the Reinforcing Effectiveness of Fentanyl, Cocaine, and Methamphetamine in Rats. Front Pharmacol 2021; 12:691700. [PMID: 34093214 PMCID: PMC8175987 DOI: 10.3389/fphar.2021.691700] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/10/2021] [Indexed: 11/23/2022] Open
Abstract
Recent estimates suggest increased popularity of the concurrent use of opioids and stimulants, with over 50% of treatment-seeking opioid users reporting regular stimulant use. The goal of the current study was to determine how opioid dependence and withdrawal affect the reinforcing effects of fentanyl, cocaine, and methamphetamine. Male Sprague-Dawley rats were allowed to self-administer fentanyl under a progressive ratio (PR) schedule of reinforcement. Baseline evaluations of reinforcing effectiveness of fentanyl, cocaine, and methamphetamine were determined. Opioid dependence was then established by administering escalating doses of morphine (10–40 mg/kg) twice-daily for four days and subsequently maintained by once-daily injections of 40 mg/kg morphine. To evaluate the impact of opioid dependence and withdrawal on the self-administration of fentanyl, cocaine, and methamphetamine, sessions occurred either 12 or 20 h after the morphine, respectively. During opioid withdrawal, the fentanyl dose-response curve was shifted rightward with an increase in maximal effectiveness, whereas it was shifted rightward with a reduction in maximal effectiveness when evaluated in rats currently dependent on opioids, relative to baseline. The reinforcing effects of cocaine and methamphetamine were unchanged by either condition. The current studies provide direct evidence that the reinforcing effects of fentanyl are increased in opioid-withdrawn rats and reduced in opioid-dependent rats, relative to rats that are not physically dependent on opioids. These findings suggest that motivations to use opioids are dependent on the state of the individual whereas stimulants retain their reinforcing effects regardless of whether the individual is in an opioid-dependent or withdrawn state.
Collapse
Affiliation(s)
- Robert W Seaman
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States.,South Texas Veterans Health Care System, San Antonio, TX, United States
| | - Gregory T Collins
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States.,South Texas Veterans Health Care System, San Antonio, TX, United States
| |
Collapse
|
7
|
De Sa Nogueira D, Bourdy R, Filliol D, Romieu P, Befort K. Hippocampal mu opioid receptors are modulated following cocaine self-administration in rat. Eur J Neurosci 2021; 53:3341-3349. [PMID: 33811699 DOI: 10.1111/ejn.15217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 03/18/2021] [Indexed: 11/30/2022]
Abstract
Cocaine addiction is a complex pathology induced by long-term brain changes. Understanding the neurochemical changes underlying the reinforcing effects of this drug of abuse is critical for reducing the societal burden of drug addiction. The mu opioid receptor plays a major role in drug reward. This receptor is modulated by chronic cocaine treatment in specific brain structures, but few studies investigated neurochemical adaptations induced by voluntary cocaine intake. In this study, we investigated whether intravenous cocaine-self administration (0.33 mg/kg/injection, fixed-ratio 1 [FR1], 10 days) in rats induces transcriptional and functional changes of the mu opioid receptor in reward-related brain regions. Epigenetic processes with histone modifications were examined for two activating marks, H3K4Me3, and H3K27Ac. We found an increase of mu opioid receptor gene expression along with a potentiation of its functionality in hippocampus of cocaine self-administering animals compared to saline controls. Chromatin immunoprecipitation followed by qPCR revealed no modifications of the histone mark H3K4Me3 and H3K27Ac levels at mu opioid receptor promoter. Our study highlights the hippocampus as an important target to further investigate neuroadaptive processes leading to cocaine addiction.
Collapse
Affiliation(s)
- David De Sa Nogueira
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA UMR7364), Centre de la Recherche Nationale Scientifique, Université de Strasbourg, Strasbourg, France
| | - Romain Bourdy
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA UMR7364), Centre de la Recherche Nationale Scientifique, Université de Strasbourg, Strasbourg, France
| | - Dominique Filliol
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA UMR7364), Centre de la Recherche Nationale Scientifique, Université de Strasbourg, Strasbourg, France
| | - Pascal Romieu
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA UMR7364), Centre de la Recherche Nationale Scientifique, Université de Strasbourg, Strasbourg, France
| | - Katia Befort
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA UMR7364), Centre de la Recherche Nationale Scientifique, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
8
|
Baidoo N, Wolter M, Holahan MR, Teale T, Winters B, Leri F. The effects of morphine withdrawal and conditioned withdrawal on memory consolidation and c-Fos expression in the central amygdala. Addict Biol 2021; 26:e12909. [PMID: 32418296 DOI: 10.1111/adb.12909] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/31/2020] [Accepted: 04/05/2020] [Indexed: 12/24/2022]
Abstract
The current study tested the hypothesis that drug withdrawal contributes to the addiction cycle in part because of an action on memory consolidation. Hence, four experiments in male Sprague-Dawley rats compared the effects of precipitated morphine withdrawal and conditioned morphine withdrawal on the consolidation of object memory and on activation of c-Fos in the central nucleus of the amygdala (CeA). It was found that immediate, but not 6 h delayed, post sample administration of 3 mg/kg of naltrexone significantly enhanced object memory in rats maintained, or previously maintained, on 10 mg/kg/day of morphine via osmotic minipumps. To establish whether conditioned withdrawal could also alter object memory, a contextual conditioning procedure was employed whereby morphine-maintained (10 mg/kg/day) animals received naltrexone (3 mg/kg) in a distinctive context (CS+) and vehicle in a separate context (CS-) for 10 days. During conditioning in the CS+, naltrexone suppressed locomotor activity, caused a rapid body weight loss and increased frequency of wet dog shakes. Interestingly, confinement to this CS+ immediately, but not 6 h, after the sample phase, also enhanced object memory. Finally, posttraining naltrexone and exposure to the CS+ both induced significant expression of c-Fos in the CeA. Therefore, this study reports for the first time that both acute precipitated withdrawal and conditioned withdrawal can facilitate memory consolidation, possibly through a common neural pathway that involves the central amygdala.
Collapse
Affiliation(s)
- Nana Baidoo
- Department of Psychology & Neuroscience University of Guelph Guelph Canada
| | - Michael Wolter
- Department of Psychology & Neuroscience University of Guelph Guelph Canada
| | | | - Tanisse Teale
- Department of Neuroscience Carleton University Ottawa Canada
| | - Boyer Winters
- Department of Psychology & Neuroscience University of Guelph Guelph Canada
| | - Francesco Leri
- Department of Psychology & Neuroscience University of Guelph Guelph Canada
| |
Collapse
|
9
|
Gu Y, Zhu D. nNOS-mediated protein-protein interactions: promising targets for treating neurological and neuropsychiatric disorders. J Biomed Res 2020; 35:1-10. [PMID: 33402546 PMCID: PMC7874267 DOI: 10.7555/jbr.34.20200108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neurological and neuropsychiatric disorders are one of the leading causes of disability worldwide and affect the health of billions of people. Nitric oxide (NO), a free gas with multitudinous bioactivities, is mainly produced from the oxidation of L-arginine by neuronal nitric oxide synthase (nNOS) in the brain. Inhibiting nNOS benefits a variety of neurological and neuropsychiatric disorders, including stroke, depression and anxiety disorders, post-traumatic stress disorder, Parkinson's disease, Alzheimer's disease, chronic pain, and drug addiction. Due to critical roles of nNOS in learning and memory and synaptic plasticity, direct inhibition of nNOS may cause severe side effects. Importantly, interactions of several proteins, including post-synaptic density 95 (PSD-95), carboxy-terminal PDZ ligand of nNOS (CAPON) and serotonin transporter (SERT), with the PSD/Disc-large/ZO-1 homologous (PDZ) domain of nNOS have been demonstrated to influence the subcellular distribution and activity of the enzyme in the brain. Therefore, it will be a preferable means to interfere with nNOS-mediated protein-protein interactions (PPIs), which do not lead to undesirable effects. Herein, we summarize the current literatures on nNOS-mediated PPIs involved in neurological and neuropsychiatric disorders, and the discovery of drugs targeting the PPIs, which is expected to provide potential targets for developing novel drugs and new strategy for the treatment of neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Yuanyuan Gu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Dongya Zhu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Institution of Stem Cell and Neuroregeneration, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
10
|
Cocaine Self-administration Regulates Transcription of Opioid Peptide Precursors and Opioid Receptors in Rat Caudate Putamen and Prefrontal Cortex. Neuroscience 2020; 443:131-139. [PMID: 32730947 DOI: 10.1016/j.neuroscience.2020.07.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 07/18/2020] [Accepted: 07/20/2020] [Indexed: 11/23/2022]
Abstract
The brain opioid system plays an important role in cocaine reward. Altered signaling in the opioid system by chronic cocaine exposure contributes to cocaine-seeking and taking behavior. The current study investigated concurrent changes in the gene expression of multiple components in rat brain opioid system following cocaine self-administration. Animals were limited to 40 infusions (1.5 mg/kg/infusion) within 6 h per day for five consecutive days. We then examined the mRNA levels of opioid receptors including mu (Oprm), delta (Oprd), and kappa (Oprk), and their endogenous opioid peptide precursors including proopiomelanocortin (Pomc), proenkephalin (Penk), prodynorphin (Pdyn) in the dorsal striatum (CPu) and the prefrontal cortex (PFC) 18 h after the last cocaine infusion. We found that cocaine self-administration significantly increased the mRNA levels of Oprm and Oprd in both the CPu and PFC, but had no effect on Oprk mRNA levels in either brain region. Moreover, cocaine had a greater influence on the mRNA levels of opioid peptide precursors in rat CPu than in the PFC. In the CPu, cocaine self-administration significantly increased the mRNA levels of Penk and Pdyn and abolished the mRNA levels of Pomc. In the PFC, cocaine self-administration only increased Pdyn mRNA levels without changing the mRNA levels of Pomc and Penk. These data suggest that cocaine self-administration influences the expression of multiple genes in the brain opioid system, and the concurrent changes in these targets may underlie cocaine-induced reward and habitual drug-seeking behavior.
Collapse
|
11
|
Baidoo N, Wolter M, Leri F. Opioid withdrawal and memory consolidation. Neurosci Biobehav Rev 2020; 114:16-24. [PMID: 32294487 DOI: 10.1016/j.neubiorev.2020.03.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 03/20/2020] [Accepted: 03/29/2020] [Indexed: 12/14/2022]
Abstract
It is well established that learning and memory are central to substance dependence. This paper specifically reviews the effect of opioid withdrawal on memory consolidation. Although there is evidence that opioid withdrawal can interfere with initial acquisition and retrieval of older memories, there are several reasons to postulate a facilitatory action on the consolidation of newly acquired memories. In fact, there is substantial evidence that memory consolidation is facilitated by the release of stress hormones, that it requires the activation of the amygdala, of central noradrenergic and cholinergic pathways, and that it involves long-term potentiation. This review highlights evidence that very similar neurobiological processes are involved in opioid withdrawal, and summarizes recent results indicating that naltrexone-precipitated withdrawal enhanced consolidation in rats. From this neurocognitive perspective, therefore, opioid use may escalate during the addiction cycle in part because memories of stimuli and actions experienced during withdrawal are strengthened.
Collapse
Affiliation(s)
- Nana Baidoo
- Department of Psychology & Neuroscience, Guelph, Ontario, Canada
| | - Michael Wolter
- Department of Psychology & Neuroscience, Guelph, Ontario, Canada
| | - Francesco Leri
- Department of Psychology & Neuroscience, Guelph, Ontario, Canada.
| |
Collapse
|
12
|
Crummy EA, Donckels EA, Baskin BM, Bentzley BS, Ferguson SM. The impact of cocaine and heroin drug history on motivation and cue sensitivity in a rat model of polydrug abuse. Psychopharmacology (Berl) 2020; 237:55-68. [PMID: 31463541 PMCID: PMC7458349 DOI: 10.1007/s00213-019-05349-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 08/09/2019] [Indexed: 11/30/2022]
Abstract
RATIONALE Comorbid use of heroin and cocaine is highly prevalent among drug users and can greatly increase addiction risk. Nonetheless, little is known regarding how a multi-drug history impacts motivation and cue responsivity to individual drugs. OBJECTIVE We used behavioral-economic procedures to examine motivation to maintain drug consumption and tests of drug-seeking to drug-associated cues to assess sensitivity to heroin and cocaine-associated cues in rats that had a self-administration history of heroin, cocaine, or both drugs. RESULTS Unexpectedly, we found that groups with a polydrug history of heroin and cocaine did not have higher levels of motivation or cue-induced reinstatement of drug-seeking for either cocaine or heroin compared to single drug groups. Nonetheless, we did find drug-specific differences in both economic price and cue sensitivity. Specifically, demand elasticity was lower for cocaine compared to heroin in animals with a single drug history, but not with polydrug groups. In addition, cocaine demand was predictive of the degree of cue-induced reinstatement of drug-seeking for cocaine following extinction, whereas heroin demand was predictive of the degree of reactivity to a heroin-associated cue. Furthermore, although cue reactivity following the initial self-administration phase did not differ across cues and drug history, reactivity to both heroin and cocaine cues was greater during subsequent heroin use compared to cocaine use, and this enhanced reactivity to heroin cues persisted during forced abstinence. CONCLUSIONS These results indicate that there is a greater motivation to maintain cocaine consumption, but higher sensitivity to drug-associated cues with a history of heroin use, suggesting that cocaine and heroin may drive continued drug use through different behavioral processes.
Collapse
Affiliation(s)
- Elizabeth A. Crummy
- Neuroscience Graduate Program, University of Washington, Seattle, WA, USA,Center for Integrative Brain Research, Seattle Children’s Research Institute, 1900 Ninth Ave., Seattle, WA 98101, USA
| | - Elizabeth A. Donckels
- Center for Integrative Brain Research, Seattle Children’s Research Institute, 1900 Ninth Ave., Seattle, WA 98101, USA
| | - Britahny M. Baskin
- Neuroscience Graduate Program, University of Washington, Seattle, WA, USA,Center for Integrative Brain Research, Seattle Children’s Research Institute, 1900 Ninth Ave., Seattle, WA 98101, USA
| | - Brandon S. Bentzley
- Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, CA, USA
| | - Susan M. Ferguson
- Neuroscience Graduate Program, University of Washington, Seattle, WA, USA,Center for Integrative Brain Research, Seattle Children’s Research Institute, 1900 Ninth Ave., Seattle, WA 98101, USA,Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
| |
Collapse
|
13
|
Storace A, Daniels S, Zhou Y, Kalisch B, Parker L, Rock E, Limebeer C, Lapointe T, Leri F. A study of limbic brain derived neurotrophic factor gene expression in male Sprague-Dawley rats trained on a learned helplessness task. Behav Brain Res 2019; 376:112174. [PMID: 31449911 DOI: 10.1016/j.bbr.2019.112174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/08/2019] [Accepted: 08/22/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Brain derived neurotrophic factor (BDNF) has been linked to the etiology and pathology of Major Depressive Disorder (MDD). Here, the relationship between learned helplessness (LH), a cognitive/motivational state relevant to MDD, and BDNF mRNA in various limbic regions, was investigated. METHODS In Sprague-Dawley male rats, LH was induced by escape training, using a triadic design of stressor controllability involving exposure to no shocks (NS), escapable shocks (ES) or yoked inescapable shocks (IES). LH was subsequently assessed in an active avoidance task, and levels of BDNF mRNA in limbic brain regions were compared across the triad following testing. RESULTS Although the IES group displayed greater LH, BDNF mRNA levels were lower in the hippocampus and higher in the nucleus accumbens of both IES and ES groups. In contrast, BDNF mRNA in the basolateral amygdala was elevated only in rats exposed to IES. CONCLUSION These results suggest that the inability to control an aversive stimulus can lead to a LH behavioural phenotype that is associated with region-specific alterations of BDNF gene expression in limbic nuclei.
Collapse
Affiliation(s)
- Alexandra Storace
- Department of Psychology, University of Guelph, Guelph, Ontario, Canada
| | - Stephen Daniels
- Department of Psychology, University of Guelph, Guelph, Ontario, Canada
| | - Yan Zhou
- Laboratory of the Biology of Addictive Disease, Rockefeller University, New York, NY, United States.
| | - Bettina Kalisch
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada.
| | - Linda Parker
- Department of Psychology, University of Guelph, Guelph, Ontario, Canada
| | - Erin Rock
- Department of Psychology, University of Guelph, Guelph, Ontario, Canada
| | - Cheryl Limebeer
- Department of Psychology, University of Guelph, Guelph, Ontario, Canada
| | - Thomas Lapointe
- Department of Psychology, University of Guelph, Guelph, Ontario, Canada
| | - Francesco Leri
- Department of Psychology, University of Guelph, Guelph, Ontario, Canada.
| |
Collapse
|
14
|
Wolter M, Huff E, Speigel T, Winters BD, Leri F. Cocaine, nicotine, and their conditioned contexts enhance consolidation of object memory in rats. ACTA ACUST UNITED AC 2019; 26:46-55. [PMID: 30651377 PMCID: PMC6340119 DOI: 10.1101/lm.048579.118] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/18/2018] [Indexed: 12/28/2022]
Abstract
To test the hypothesis that drugs of abuse and their conditioned stimuli (CSs) enhance memory consolidation, the effects of post-training exposure to cocaine and nicotine were compared to the effects of post-training exposure to contextual stimuli that were paired with the effects of these drugs. Using the object recognition (OR) task, it was first demonstrated that both 10 and 20 mg/kg cocaine, and 0.2 and 0.4 mg/kg nicotine, enhanced recognition memory when administered immediately after, but not 6 h after the sample phase. To establish the drug CSs, rats were confined for 2 h in a chamber (the CS+) after injections of 20 mg/kg cocaine, or 0.4 mg/kg nicotine, and in another chamber (the CS−) after injections of vehicle. This was repeated over 10 d (5 drug/CS+ and 5 vehicle/CS− pairings in total). At the end of this conditioning period, when tested in a drug-free state, rats displayed conditioned hyperactivity in the CS+ relative to the CS−. More important, immediate, but not delayed, post-sample exposure to the cocaine CS+, or nicotine CS+, enhanced OR memory. Therefore, this study reports for the first time that contextual stimuli paired with cocaine and nicotine, like the drugs themselves, have the ability to enhance memory consolidation.
Collapse
Affiliation(s)
- Michael Wolter
- Department of Psychology and Collaborative Program in Neuroscience, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Ethan Huff
- Department of Psychology and Collaborative Program in Neuroscience, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Talia Speigel
- Department of Psychology and Collaborative Program in Neuroscience, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Boyer D Winters
- Department of Psychology and Collaborative Program in Neuroscience, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Francesco Leri
- Department of Psychology and Collaborative Program in Neuroscience, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| |
Collapse
|
15
|
The gut microbiota mediates reward and sensory responses associated with regimen-selective morphine dependence. Neuropsychopharmacology 2018; 43:2606-2614. [PMID: 30258112 PMCID: PMC6224506 DOI: 10.1038/s41386-018-0211-9] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 08/17/2018] [Accepted: 08/20/2018] [Indexed: 02/06/2023]
Abstract
Opioid use for long-term pain management is limited by adverse side effects, such as hyperalgesia and negative affect. Neuroinflammation in the brain and spinal cord is a contributing factor to the development of symptoms associated with chronic opioid use. Recent studies have described a link between neuroinflammation and behavior that is mediated by a gut-brain signaling axis, where alterations in indigenous gut bacteria contribute to several inflammation-related psychopathologies. As opioid receptors are highly expressed within the digestive tract and opioids influence gut motility, we hypothesized that systemic opioid treatment will impact the composition of the gut microbiota. Here, we explored how opioid treatments, and cessation, impacts the mouse gut microbiome and whether opioid-induced changes in the gut microbiota influences inflammation-driven hyperalgesia and impaired reward behavior. Male C57Bl6/J mice were treated with either intermittent or sustained morphine. Using 16S rDNA sequencing, we describe changes in gut microbiota composition following different morphine regimens. Manipulation of the gut microbiome was used to assess the causal relationship between the gut microbiome and opioid-dependent behaviors. Intermittent, but not sustained, morphine treatment was associated with microglial activation, hyperalgesia, and impaired reward response. Depletion of the gut microbiota via antibiotic treatment surprisingly recapitulated neuroinflammation and sequelae, including reduced opioid analgesic potency and impaired cocaine reward following intermittent morphine treatment. Colonization of antibiotic-treated mice with a control microbiota restored microglial activation state and behaviors. Our findings suggest that differing opioid regimens uniquely influence the gut microbiome that is causally related to behaviors associated with opioid dependence.
Collapse
|
16
|
Thériault RK, Leri F, Kalisch B. The role of neuronal nitric oxide synthase in cocaine place preference and mu opioid receptor expression in the nucleus accumbens. Psychopharmacology (Berl) 2018; 235:2675-2685. [PMID: 29992335 DOI: 10.1007/s00213-018-4961-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 06/27/2018] [Indexed: 10/28/2022]
Abstract
RATIONALE There is evidence that central mu opioid receptors (MORs) are implicated in several aspects of cocaine addiction, and that MOR expression is elevated by cocaine in vitro and in the nucleus accumbens (NAc) when administered in vivo. OBJECTIVE To understand the cellular mechanisms involved in regulating MOR expression, this study explored whether neuronal nitric oxide synthase (nNOS) modulates the neurochemical and behavioral effects of acute and repeated cocaine administration. METHODS Male Sprague-Dawley rats received a single cocaine injection (20 mg/kg, i.p.) in combination with the selective nNOS inhibitor 7-nitroindazole (7-NI) (0, 25, or 50 mg/kg, i.p.), and the expression of MOR and nNOS messenger RNA (mRNA) and protein levels in the NAc were measured. In a separate conditioned place preference (CPP) experiment, 7-NI (0, 25, or 50 mg/kg, i.p.) was administered prior to cocaine (0 or 20 mg/kg, i.p.) conditioning sessions, and levels of MOR and nNOS mRNA and protein in the NAc were measured following CPP test. RESULTS Acute cocaine administration significantly enhanced nNOS and MOR mRNA and protein expression in the NAc, and this increase in MOR expression was blocked by 7-NI. Furthermore, in 7-NI pre-treated rats, cocaine-induced CPP was not statistically significant and the increase in MOR mRNA expression in the NAc in these animals was attenuated. CONCLUSIONS These findings suggest that nNOS modulates MOR expression following acute cocaine administration, and that cocaine CPP and associated upregulation of MOR expression involve both nNOS-dependent and independent mechanisms. Elucidation of these molecular events may identify useful therapeutic target for cocaine addiction.
Collapse
Affiliation(s)
- Rachel-Karson Thériault
- Department of Biomedical Sciences, University of Guelph (ON), Guelph, Ontario, N1G 2W1, Canada.,Collaborative Neuroscience Program, University of Guelph (ON), Guelph, Ontario, N1G 2W1, Canada
| | - Francesco Leri
- Collaborative Neuroscience Program, University of Guelph (ON), Guelph, Ontario, N1G 2W1, Canada.,Department of Psychology, University of Guelph (ON), Guelph, Ontario, N1G 2W1, Canada
| | - Bettina Kalisch
- Department of Biomedical Sciences, University of Guelph (ON), Guelph, Ontario, N1G 2W1, Canada. .,Collaborative Neuroscience Program, University of Guelph (ON), Guelph, Ontario, N1G 2W1, Canada.
| |
Collapse
|
17
|
Levy A, Daniels S, Hudson R, Horman T, Flynn A, Zhou Y, Leri F. Bupropion and naltrexone combination alters high fructose corn syrup self-administration and gene expression in rats. Neuropharmacology 2018; 135:547-554. [DOI: 10.1016/j.neuropharm.2018.01.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 01/18/2018] [Accepted: 01/24/2018] [Indexed: 12/22/2022]
|
18
|
Daniels S, Pratt M, Zhou Y, Leri F. Effect of steady-state methadone on high fructose corn syrup consumption in rats. J Psychopharmacol 2018; 32:215-222. [PMID: 29207922 DOI: 10.1177/0269881117742116] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Patients undergoing methadone maintenance treatment self-report enhanced preferences for, and excessive consumption of, foods rich in sugar. However, it is unclear whether these are direct pharmacological effects of methadone or the consequences of metabolic dysfunctions induced by addiction to illicit opiates. Hence, the current study in drug-naïve male Sprague-Dawley rats explored the effects of steady-state methadone delivered by osmotic mini-pumps (13 days; 0, 10, 30 mg/kg/day) on consumption of rat chow and a palatable, sweet, liquid high fructose corn syrup solution. Six days after the removal of the pumps, mRNA expression of genes involved in responses to stress and rewards were quantified: pro-opiomelanocortin in the hypothalamus, mu-opioid receptor in the nucleus accumbens, and dopamine D2 receptor in the dorsal striatum. Taste reactivity and locomotion tests were also performed throughout the study. It was found that methadone increased caloric intake from high fructose corn syrup and reduced caloric intake from chow, effects that could not be directly ascribed to changes in high fructose corn syrup taste reactivity or motor functions. However, the changes in caloric intake displayed significant tolerance, and mRNA expression analysis suggested that methadone attenuated the effect of high fructose corn syrup on pro-opiomelanocortin mRNA, and possibly on dopamine D2 receptor mRNA. These findings in rats suggest that the pharmacological effect of methadone, administered to achieve steady-state maintenance, may not be the primary cause of dietary alterations reported by patients maintained on methadone.
Collapse
Affiliation(s)
| | | | - Yan Zhou
- 2 The Rockefeller University, New York, USA
| | | |
Collapse
|
19
|
Taylor AMW, Castonguay A, Ghogha A, Vayssiere P, Pradhan AAA, Xue L, Mehrabani S, Wu J, Levitt P, Olmstead MC, De Koninck Y, Evans CJ, Cahill CM. Neuroimmune Regulation of GABAergic Neurons Within the Ventral Tegmental Area During Withdrawal from Chronic Morphine. Neuropsychopharmacology 2016; 41:949-59. [PMID: 26202104 PMCID: PMC4748420 DOI: 10.1038/npp.2015.221] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 07/10/2015] [Accepted: 07/11/2015] [Indexed: 12/13/2022]
Abstract
Opioid dependence is accompanied by neuroplastic changes in reward circuitry leading to a negative affective state contributing to addictive behaviors and risk of relapse. The current study presents a neuroimmune mechanism through which chronic opioids disrupt the ventral tegmental area (VTA) dopaminergic circuitry that contributes to impaired reward behavior. Opioid dependence was induced in rodents by treatment with escalating doses of morphine. Microglial activation was observed in the VTA following spontaneous withdrawal from chronic morphine treatment. Opioid-induced microglial activation resulted in an increase in brain-derived neurotrophic factor (BDNF) expression and a reduction in the expression and function of the K(+)Cl(-) co-transporter KCC2 within VTA GABAergic neurons. Inhibition of microglial activation or interfering with BDNF signaling prevented the loss of Cl(-) extrusion capacity and restored the rewarding effects of cocaine in opioid-dependent animals. Consistent with a microglial-derived BDNF-induced disruption of reward, intra-VTA injection of BDNF or a KCC2 inhibitor resulted in a loss of cocaine-induced place preference in opioid-naïve animals. The loss of the extracellular Cl(-) gradient undermines GABAA-mediated inhibition, and represents a mechanism by which chronic opioid treatments can result in blunted reward circuitry. This study directly implicates microglial-derived BDNF as a negative regulator of reward in opioid-dependent states, identifying new therapeutic targets for opiate addictive behaviors.
Collapse
Affiliation(s)
- Anna M W Taylor
- Department of Anesthesiology and Perioperative Care, University of California, Irvine, CA, USA
- Hatos Center for Neuropharmacology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Annie Castonguay
- Institut Universitaire en Santé Mentale de Québec, Québec, QC, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec, QC, Canada
| | - Atefeh Ghogha
- Hatos Center for Neuropharmacology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Pia Vayssiere
- Hatos Center for Neuropharmacology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Amynah A A Pradhan
- Hatos Center for Neuropharmacology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Lihua Xue
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Sadaf Mehrabani
- Hatos Center for Neuropharmacology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Juli Wu
- Children's Hospital Los Angeles and the Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Pat Levitt
- Children's Hospital Los Angeles and the Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Mary C Olmstead
- Department of Psychology, Queen's University, Kingston, ON, Canada
| | - Yves De Koninck
- Institut Universitaire en Santé Mentale de Québec, Québec, QC, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec, QC, Canada
| | - Christopher J Evans
- Hatos Center for Neuropharmacology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Catherine M Cahill
- Department of Anesthesiology and Perioperative Care, University of California, Irvine, CA, USA
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| |
Collapse
|
20
|
Fructose:glucose ratios--a study of sugar self-administration and associated neural and physiological responses in the rat. Nutrients 2015; 7:3869-90. [PMID: 26007337 PMCID: PMC4446784 DOI: 10.3390/nu7053869] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 04/21/2015] [Accepted: 05/11/2015] [Indexed: 11/16/2022] Open
Abstract
This study explored whether different ratios of fructose (F) and glucose (G) in sugar can engender significant differences in self-administration and associated neurobiological and physiological responses in male Sprague-Dawley rats. In Experiment 1, animals self-administered pellets containing 55% F + 45% G or 30% F + 70% G, and Fos immunoreactivity was assessed in hypothalamic regions regulating food intake and reward. In Experiment 2, rats self-administered solutions of 55% F + 42% G (high fructose corn syrup (HFCS)), 50% F + 50% G (sucrose) or saccharin, and mRNA of the dopamine 2 (D2R) and mu-opioid (MOR) receptor genes were assessed in striatal regions involved in addictive behaviors. Finally, in Experiment 3, rats self-administered HFCS and sucrose in their home cages, and hepatic fatty acids were quantified. It was found that higher fructose ratios engendered lower self-administration, lower Fos expression in the lateral hypothalamus/arcuate nucleus, reduced D2R and increased MOR mRNA in the dorsal striatum and nucleus accumbens core, respectively, as well as elevated omega-6 polyunsaturated fatty acids in the liver. These data indicate that a higher ratio of fructose may enhance the reinforcing effects of sugar and possibly lead to neurobiological and physiological alterations associated with addictive and metabolic disorders.
Collapse
|
21
|
Wang Y, Wang H, Li W, Zhu J, Gold MS, Zhang D, Wang L, Li Y, Yan X, Cheng J, Li Q, Wang W. Reduced responses to heroin-cue-induced craving in the dorsal striatum: effects of long-term methadone maintenance treatment. Neurosci Lett 2014; 581:120-4. [PMID: 25157798 DOI: 10.1016/j.neulet.2014.08.026] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 07/29/2014] [Accepted: 08/14/2014] [Indexed: 10/24/2022]
Abstract
Methadone maintenance treatment (MMT) is safe and effective for heroin addiction, but the neural basis of the length effects of long-term MMT on brain activity during craving in former heroin addicts is unclear. This study explored it by comparing the brain activations of heroin addicts with different length of MMT during pictorial presentation of heroin-related cue. Fifteen male former heroin addicts successfully treated by MMT less than 1 year (Group A), 15 matched patients with 2-3 year MMT (Group B) and 17 healthy controls underwent functional magnetic resonance imaging while heroin-related and neutral stimuli were present to them. Subjective cue-elicited craving was measured with visual analog scale before and after imaging. Then, partial correlation analysis to reveal the relationship between drug-related blood oxygen level dependent (BOLD) signal intensity and heroin or methadone use history. Finally, self-reported craving was not different between Group A and B before and after scanning. Compared with Group A, Group B had a significant reduced brain activity to heroin-related minus neural cues in the bilateral caudate. After controlling for the variable heroin use history, the drug-related BOLD signal intensity in the bilateral caudate was negatively correlated with MMT duration and total methadone consumption. When MMT history was controlled, the drug-related activity intensity in right caudate had a positive correlation with heroin daily dosage. Long-term MMT may improve heroin-craving response by modulating the impaired function in the bilateral dorsal striatum caused by former heroin use.
Collapse
Affiliation(s)
- Yarong Wang
- Department of Radiology, Tangdu Hospital, the Fourth Military Medical University, 569 Xinsi Road, Baqiao District, Xi'an 710038, China; Department of Psychiatry and Addiction Medicine, University of Florida College of Medicine and McKnight Brain Institute, Gainesville, Florida 32610, USA
| | - Hanyue Wang
- Department of Radiology, Tangdu Hospital, the Fourth Military Medical University, 569 Xinsi Road, Baqiao District, Xi'an 710038, China
| | - Wei Li
- Department of Radiology, Tangdu Hospital, the Fourth Military Medical University, 569 Xinsi Road, Baqiao District, Xi'an 710038, China
| | - Jia Zhu
- Department of Radiology, Tangdu Hospital, the Fourth Military Medical University, 569 Xinsi Road, Baqiao District, Xi'an 710038, China
| | - Mark S Gold
- Department of Psychiatry and Addiction Medicine, University of Florida College of Medicine and McKnight Brain Institute, Gainesville, Florida 32610, USA
| | - Dongsheng Zhang
- Department of Radiology, Tangdu Hospital, the Fourth Military Medical University, 569 Xinsi Road, Baqiao District, Xi'an 710038, China
| | - Lina Wang
- Department of Radiology, Tangdu Hospital, the Fourth Military Medical University, 569 Xinsi Road, Baqiao District, Xi'an 710038, China
| | - Yongbin Li
- Department of Radiology, Tangdu Hospital, the Fourth Military Medical University, 569 Xinsi Road, Baqiao District, Xi'an 710038, China
| | - Xuejiao Yan
- Department of Radiology, Tangdu Hospital, the Fourth Military Medical University, 569 Xinsi Road, Baqiao District, Xi'an 710038, China
| | - Jiajie Cheng
- Department of Radiology, Tangdu Hospital, the Fourth Military Medical University, 569 Xinsi Road, Baqiao District, Xi'an 710038, China
| | - Qiang Li
- Department of Radiology, Tangdu Hospital, the Fourth Military Medical University, 569 Xinsi Road, Baqiao District, Xi'an 710038, China.
| | - Wei Wang
- Department of Radiology, Tangdu Hospital, the Fourth Military Medical University, 569 Xinsi Road, Baqiao District, Xi'an 710038, China.
| |
Collapse
|
22
|
Drugs of abuse as memory modulators: a study of cocaine in rats. Psychopharmacology (Berl) 2014; 231:2339-48. [PMID: 24337026 DOI: 10.1007/s00213-013-3390-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 11/29/2013] [Indexed: 01/22/2023]
Abstract
RATIONALE It has been proposed that drugs of abuse reinforce behavior partly, or wholly, because they facilitate learning by enhancing memory consolidation. Cocaine can clearly serve as a reinforcer, but its effect on learning has not been fully characterized. OBJECTIVES To explore the effects of different regimens of pre- and post-training cocaine administration on win-stay and object learning. METHODS Cocaine naïve and cocaine pre-exposed (30 mg/kg/day, × 5 days followed by 7 days drug-free) male Sprague-Dawley rats received cocaine (0, 1, 2.5, 7.5, or 20 mg/kg, i.p.) immediately following training on a win-stay task in a radial maze or following the sample phase of an object learning task. Win-stay performance was also assessed in tests of extinction and after a set shift. RESULTS Post-training cocaine did not improve accuracy on the win-stay task and produced performance deficits at 20 mg/kg. These deficits were attenuated by prior cocaine exposure. There was indirect evidence of facilitated learning in extinction and set shift tests, but the effective dosage was different (2.5 and 7.5 mg/kg, respectively). Post-training cocaine produced dose-dependent improvements in object learning. CONCLUSION Post-training cocaine administration can facilitate learning, but this effect is highly dependent on the dose and the type of task employed.
Collapse
|
23
|
Bidlack JM. Mixed κ/μ partial opioid agonists as potential treatments for cocaine dependence. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 69:387-418. [PMID: 24484983 DOI: 10.1016/b978-0-12-420118-7.00010-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Cocaine use activates the dopamine reward pathway, leading to the reinforcing effects of dopamine. There is no FDA-approved medication for treating cocaine dependence. Opioid agonists and antagonists have been approved for treating opioid and alcohol dependence. Agonists that activate the μ opioid receptor increase dopamine levels in the nucleus accumbens, while μ receptor antagonists decrease dopamine levels by blocking the effects of endogenous opioid peptides. Activation of the κ opioid receptor decreases dopamine levels and leads to dysphoria. In contrast, inhibition of the κ opioid receptor decreases dopamine levels in the nucleus accumbens. Antagonists acting at the κ receptor reduce stress-mediated behaviors and anxiety. Mixed partial μ/κ agonists have the potential of striking a balance between dopamine levels and attenuating relapse to cocaine. The pharmacological properties of mixed μ/κ opioid receptor agonists will be discussed and results from clinical and preclinical studies will be presented. Results from studies with some of the classical benzomorphans and morphinans will be presented as they lay the foundation for structure-activity relationships. Recent results with other partial opioid agonists, including buprenorphine derivatives and the mixed μ/κ peptide CJ-15,208, will be discussed. The behavioral effects of the mixed μ/κ MCL-741, an aminothiazolomorphinan, in attenuating cocaine-induced locomotor activity will be presented. While not a mixed μ/κ opioid, results obtained with GSK1521498, a μ receptor inverse agonist, will be discussed. Preclinical strategies and successes will lay the groundwork for the further development of mixed μ/κ opioid receptor agonists to treat cocaine dependence.
Collapse
Affiliation(s)
- Jean M Bidlack
- School of Medicine and Dentistry, University of Rochester, Rochester, New York, USA.
| |
Collapse
|
24
|
Chang SL, Connaghan KP, Wei Y, Li MD. NeuroHIV and use of addictive substances. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2014; 118:403-40. [PMID: 25175871 DOI: 10.1016/b978-0-12-801284-0.00013-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the past three decades, substance abuse has been identified as a key comorbidity of human immunodeficiency virus-1 (HIV-1) infection. Many studies have found that the use and abuse of addictive substances hastens the progression of HIV-1 infection and HIV-associated neurocognitive disorders. Advances in highly active antiretroviral therapy (HAART) in the mid-1990s have been successful in limiting the HIV-1 viral load and maintaining a relatively healthy immune response, allowing the life expectancy of patients infected with HIV to approach that of the general population. However, even with HAART, HIV-1 viral proteins are still expressed and eradication of the virus, particularly in the brain, the key reservoir organ, does not occur. In the post-HAART era, the clinical challenge in the treatment of HIV infection is inflammation of the central nervous system (CNS) and its subsequent neurological disorders. To date, various explicit and implicit connections have been identified between the neuronal circuitry involved in immune responses and brain regions affected by and implicated in substance abuse. This chapter discusses past and current medical uses of prototypical substances of abuse, including morphine, alcohol, cocaine, methamphetamine, marijuana, and nicotine, and the evidence that systemic infections, particularly HIV-1 infection, cause neurological dysfunction as a result of inflammation in the CNS, which can increase the risk of substance abuse.
Collapse
Affiliation(s)
- Sulie L Chang
- Institute of NeuroImmune Pharmacology, Seton Hall University, South Orange, New Jersey, USA; Department of Biological Sciences, Seton Hall University, South Orange, New Jersey, USA.
| | - Kaitlyn P Connaghan
- Institute of NeuroImmune Pharmacology, Seton Hall University, South Orange, New Jersey, USA
| | - Yufeng Wei
- Institute of NeuroImmune Pharmacology, Seton Hall University, South Orange, New Jersey, USA
| | - Ming D Li
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
25
|
Effect of post-training administration of cocaine, diazepam and their combination on a win-stay task. Pharmacol Biochem Behav 2013; 116:69-74. [PMID: 24246909 DOI: 10.1016/j.pbb.2013.11.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 10/31/2013] [Accepted: 11/07/2013] [Indexed: 12/18/2022]
Abstract
According to the memory-enhancing hypothesis of addictive drugs, post-training administration of cocaine should enhance consolidation and thus facilitate learning. This hypothesis has not been tested in appetitive tasks reinforced by sucrose. The current study assessed the effect of post-training cocaine administration on the acquisition of a win-stay task, and modulation of this effect by co-administration of diazepam. Male Sprague-Dawley rats (n=63) were trained for 5 days on a win-stay task performed on an 8-arm radial maze, and were administered cocaine (0, 2.5, 7.5 or 20mg/kg), diazepam (1mg/kg), or cocaine (7.5mg/kg)+diazepam (1mg/kg) immediately following each training session. Post-training cocaine caused dose-dependent impairments that appeared linked to the development of cocaine-induced sucrose taste avoidance and/or cocaine-induced anxiety. When it was attempted to modify these learned side effects of cocaine by co-administration of diazepam, it was observed that the drug combination slowed task completion and reduced overall number of nose pokes. These findings suggest that post-training cocaine can alter behavior on appetitive tasks through learned motivational deficits rather than through a selective action on memory consolidation. The implications for the memory-enhancing hypothesis of addictive drugs are discussed.
Collapse
|
26
|
Cummins E, Leri F. Animal studies trigger new insights on the use of methadone maintenance. Expert Opin Drug Discov 2013; 4:577-86. [PMID: 23485087 DOI: 10.1517/17460440902915533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Although steady-state methadone (SSM) treatment is mainly used for opioid addiction, some clinical studies indicate that it also reduces cocaine abuse in opioid-dependent individuals. OBJECTIVE/METHODS To present evidence suggesting that SSM may be useful in the treatment of cocaine addiction without pre-existing opioid dependence. We review studies in animals investigating the effects of SSM on behaviors motivated by cocaine and on cocaine-induced alterations of genes expression in the rat brain. CONCLUSION SSM reduces cocaine intake, blocks cocaine seeking and normalizes expression of genes known to regulate cocaine seeking. These findings suggest that SSM could be an effective pharmacological agent to assist cocaine detoxification and prevention of relapse to cocaine abuse in individuals not co-dependent on opioid.
Collapse
Affiliation(s)
- Erin Cummins
- PhD Student University of Guelph, Department of Psychology, Guelph (ON), N1G 2W1, Canada
| | | |
Collapse
|
27
|
Yoo JH, Kitchen I, Bailey A. The endogenous opioid system in cocaine addiction: what lessons have opioid peptide and receptor knockout mice taught us? Br J Pharmacol 2012; 166:1993-2014. [PMID: 22428846 DOI: 10.1111/j.1476-5381.2012.01952.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Cocaine addiction has become a major concern in the UK as Britain tops the European 'league table' for cocaine abuse. Despite its devastating health and socio-economic consequences, no effective pharmacotherapy for treating cocaine addiction is available. Identifying neurochemical changes induced by repeated drug exposure is critical not only for understanding the transition from recreational drug use towards compulsive drug abuse but also for the development of novel targets for the treatment of the disease and especially for relapse prevention. This article focuses on the effects of chronic cocaine exposure and withdrawal on each of the endogenous opioid peptides and receptors in rodent models. In addition, we review the studies that utilized opioid peptide or receptor knockout mice in order to identify and/or clarify the role of different components of the opioid system in cocaine-addictive behaviours and in cocaine-induced alterations of brain neurochemistry. The review of these studies indicates a region-specific activation of the µ-opioid receptor system following chronic cocaine exposure, which may contribute towards the rewarding effect of the drug and possibly towards cocaine craving during withdrawal followed by relapse. Cocaine also causes a region-specific activation of the κ-opioid receptor/dynorphin system, which may antagonize the rewarding effect of the drug, and at the same time, contribute to the stress-inducing properties of the drug and the triggering of relapse. These conclusions have important implications for the development of effective pharmacotherapy for the treatment of cocaine addiction and the prevention of relapse.
Collapse
Affiliation(s)
- Ji Hoon Yoo
- Division of Biochemistry, Faculty of Health & Medical Sciences, University of Surrey, Guildford, Surrey, UK
| | | | | |
Collapse
|
28
|
Winick-Ng W, Leri F, Kalisch BE. Nitric oxide and histone deacetylases modulate cocaine-induced mu-opioid receptor levels in PC12 cells. BMC Pharmacol Toxicol 2012; 13:11. [PMID: 23079001 PMCID: PMC3520874 DOI: 10.1186/2050-6511-13-11] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 10/11/2012] [Indexed: 11/24/2022] Open
Abstract
Background Cocaine exposure has been reported to alter central μ-opioid receptor (MOR) expression in vivo. The present study employed an in vitro cellular model to explore possible mechanisms that may be involved in this action of cocaine. Methods To assess the effects of cocaine on MOR levels, two treatment regimens were tested in PC12 cells: single continuous or multiple intermittent. MOR protein levels were assessed by western blot analysis and quantitative PCR was used to determine relative MOR mRNA expression levels. To evaluate the role of nitric oxide (NO) and histone acetylation in cocaine-induced MOR expression, cells were pre-treated with the NO synthase inhibitor Nω-nitro-L-arginine methylester (L-NAME) or the non-selective histone acetyltransferase inhibitor curcumin. Results Both cocaine treatment regimens significantly increased MOR protein levels and protein stability, but only multiple intermittent treatments increased MOR mRNA levels as well as c-fos mRNA levels and activator protein 1 binding activity. Both regimens increased NO production, and pre-treatment with L-NAME prevented cocaine-induced increases in MOR protein and mRNA levels. Single and multiple cocaine treatment regimens inhibited histone deacetylase activity, and pre-treatment with curcumin prevented cocaine-induced up-regulation of MOR protein expression. Conclusions In the PC12 cell model, both NO and histone deacetylase activity regulate cocaine-induced MOR expression at both the transcriptional and post-transcriptional levels. Based on these novel findings, it is hypothesized that epigenetic mechanisms are implicated in cocaine’s action on MOR expression in neurons.
Collapse
Affiliation(s)
- Warren Winick-Ng
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, N1G 2W1, Canada
| | | | | |
Collapse
|
29
|
Kreek MJ, Levran O, Reed B, Schlussman SD, Zhou Y, Butelman ER. Opiate addiction and cocaine addiction: underlying molecular neurobiology and genetics. J Clin Invest 2012; 122:3387-93. [PMID: 23023708 DOI: 10.1172/jci60390] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Addictive diseases, including addiction to heroin, prescription opioids, or cocaine, pose massive personal and public health costs. Addictions are chronic relapsing diseases of the brain caused by drug-induced direct effects and persisting neuroadaptations at the epigenetic, mRNA, neuropeptide, neurotransmitter, or protein levels. These neuroadaptations, which can be specific to drug type, and their resultant behaviors are modified by various internal and external environmental factors, including stress responsivity, addict mindset, and social setting. Specific gene variants, including variants encoding pharmacological target proteins or genes mediating neuroadaptations, also modify vulnerability at particular stages of addiction. Greater understanding of these interacting factors through laboratory-based and translational studies have the potential to optimize early interventions for the therapy of chronic addictive diseases and to reduce the burden of relapse. Here, we review the molecular neurobiology and genetics of opiate addiction, including heroin and prescription opioids, and cocaine addiction.
Collapse
Affiliation(s)
- Mary Jeanne Kreek
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, New York 10065, USA.
| | | | | | | | | | | |
Collapse
|
30
|
The effects of acute and chronic steady state methadone on memory retrieval in rats. Psychopharmacology (Berl) 2012; 222:225-35. [PMID: 22258154 DOI: 10.1007/s00213-012-2638-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2011] [Accepted: 12/30/2011] [Indexed: 10/14/2022]
Abstract
RATIONALE Although widely prescribed to treat opioid addiction, little is known about the possible side effects of methadone on memory functions. OBJECTIVES The aim of this study is to compare the effects of acute and chronic methadone on memory retrieval in rats and to explore the selectivity of possible deficits. METHODS Administration of acute (0, 1.25, 2.5, and 5 mg/kg SC) and chronic steady state methadone (0, 10, 30, and 55 mg/kg/day SC by osmotic mini-pump) was tested on recall of three different types of information: stimulus-reward (10-arm parallel maze), stimulus-response (8-arm radial maze), and stimulus-stimulus (Barnes maze). Acute and steady state methadone doses were also compared on tests of locomotor activity and reactivity to aversive stimuli (i.e., swimming and acoustic startle). RESULTS In the stimulus-reward task, acute methadone impaired performance as a result of severe depression of locomotion. This motor deficit, however, was modulated by the motivational valence of environmental stimulation. In fact, acute methadone did not eliminate forced swimming behavior. In the stimulus-response and stimulus-stimulus tasks, accuracy was impaired independently of direct motor deficits, but rats were hyper-reactive to aversive stimulation and, in fact, 5 mg/kg enhanced acoustic startle. Importantly, chronic steady state methadone did not affect accuracy of memory retrieval, did not depress motor or swimming activity, and did not change startle reactivity. CONCLUSION Only acute methadone impaired accuracy and/or performance on three tests of memory retrieval. These findings in rats suggest that memory deficits reported in methadone-maintained individuals may not be directly attributable to methadone.
Collapse
|
31
|
Leri F, Zhou Y, Carmichael B, Cummins E, Kreek MJ. Treatment-like steady-state methadone in rats interferes with incubation of cocaine sensitization and associated alterations in gene expression. Eur Neuropsychopharmacol 2012; 22:143-52. [PMID: 21745729 PMCID: PMC3810140 DOI: 10.1016/j.euroneuro.2011.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 05/12/2011] [Accepted: 06/14/2011] [Indexed: 01/11/2023]
Abstract
In a previous study, steady-state methadone treatment was found to prevent associative cocaine learning, as well as related decreases in mRNA expression of preprohypocretin/preproorexin (ppHcrt) in the lateral hypothalamus (LH) and dopamine D2 receptor (DR2) in the caudate-putamen (CP), and increases in mu-opioid receptor in the ventral striatum of rats. To investigate whether the same regimen of methadone exposure could prevent the incubation of cocaine sensitization and related alterations in gene expression, male Sprague-Dawley rats received 45 mg/kg/day steady-dose "binge" cocaine administration (IP) for 14 days followed by mini-pumps releasing 30 mg/kg/day methadone (SC). After 14 days of methadone, and a subsequent 10-day drug-free period, all rats were tested for sensitization (cocaine test dose: 15 mg/kg) and brain tissue was collected to quantify mRNA expression. Rats exposed to cocaine displayed cocaine-induced stereotypy at test, as well as enhanced ppHcrt mRNA in the LH and reduced DR2 mRNA in the CP. Importantly, these alterations were significantly reduced in rats treated with methadone following cocaine. These results suggest that steady-state methadone can interfere with the incubation of neuroadaptations underlying changes in behavioral responses to cocaine and cocaine-associated stimuli, and that these effects can be observed even after withdrawal from methadone.
Collapse
|
32
|
Abstract
Major well-defined medical problems that are, in part, the unfortunate outcome of a negative social environment may include specific addictive diseases and other mental health disorders, in particular the affective disorders of anxiety, depression, social phobia, and posttraumatic stress syndrome. This overview touches on the topic of extreme marginalization associated with addiction and other mental health disorders, along with arrest, imprisonment, and parole. All of these are characterized by a lasting stigma that hauntingly continues to affect each person suffering from any of these problems.
Collapse
Affiliation(s)
- Mary Jeanne Kreek
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, USA.
| |
Collapse
|
33
|
Vucetic Z, Kimmel J, Reyes TM. Chronic high-fat diet drives postnatal epigenetic regulation of μ-opioid receptor in the brain. Neuropsychopharmacology 2011; 36:1199-206. [PMID: 21326195 PMCID: PMC3077442 DOI: 10.1038/npp.2011.4] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Revised: 01/04/2011] [Accepted: 01/05/2011] [Indexed: 12/25/2022]
Abstract
Opioid system dysregulation has been observed in both genetic and high-fat diet (HFD)-induced models of obesity. An understanding of the molecular mechanisms of MOR transcriptional regulation, particularly within an in vivo context, is lacking. Using a diet-induced model of obesity (DIO), mice were fed a high-fat diet (60% calories from fat) from weaning to >18 weeks of age. Compared with mice fed the control diet, DIO mice had a decreased preference for sucrose. MOR mRNA expression was decreased in reward-related circuitry (ventral tegmental area (VTA), nucleus accumbens (NAc), and prefrontal cortex (PFC)) but not the hypothalamus, important in the homeostatic regulation of feeding. DNA methylation is an epigenetic modification that links environmental exposures to altered gene expression. We found a significant increase in DNA methylation in the MOR promoter region within the reward-related brain regions. Methyl CpG-binding protein 2 (MeCP2) can bind methylated DNA and repress transcription, and DIO mice showed increased binding of MeCP2 to the MOR promoter in reward-related regions of the brain. Finally, using ChIP assays we examined H3K9 methylation (inactive chromatin) and H3 acetylation (active chromatin) within the MOR promoter region and found increased H3K9 methylation and decreased H3 acetylation. These data are the first to identify DNA methylation, MeCP2 recruitment, and chromatin remodeling as mechanisms leading to transcriptional repression of MOR in the brains of mice fed a high-fat diet.
Collapse
Affiliation(s)
- Zivjena Vucetic
- Institute for Translational Medicine and Therapeutics, Department of Pharmacology University of Pennsylvania, School of Medicine, Philadelphia, PA, USA
| | - Jessica Kimmel
- Institute for Translational Medicine and Therapeutics, Department of Pharmacology University of Pennsylvania, School of Medicine, Philadelphia, PA, USA
| | - Teresa M Reyes
- Institute for Translational Medicine and Therapeutics, Department of Pharmacology University of Pennsylvania, School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
34
|
Brown TE, Lee BR, Ryu V, Herzog T, Czaja K, Dong Y. Reducing hippocampal cell proliferation in the adult rat does not prevent the acquisition of cocaine-induced conditioned place preference. Neurosci Lett 2010; 481:41-6. [PMID: 20600607 DOI: 10.1016/j.neulet.2010.06.048] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Revised: 06/14/2010] [Accepted: 06/16/2010] [Indexed: 02/07/2023]
Abstract
Neurogenesis is important for developing certain forms of memory. Recently, hippocampal cell proliferation has been implicated in the development of drug addiction, an extreme form of emotional/motivational pathological memory. Aiming to explore the role of hippocampal neural cell proliferation in cocaine-induced conditioned place preference (CPP), we treated rats with whole brain X-irradiation, which substantially decreases the number of progenitor cells in the subventricular zone of the lateral ventricles and subgranular zone of the dentate gyrus. Surprisingly, there was no difference in the expression of cocaine-induced CPP. These results suggest that the existing neural network, rather than potential new neural circuits mediated by adult neurogenesis, is sufficient for the acquisition of cocaine-induced CPP.
Collapse
Affiliation(s)
- Travis E Brown
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, RI 02912, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Le Merrer J, Becker JAJ, Befort K, Kieffer BL. Reward processing by the opioid system in the brain. Physiol Rev 2009; 89:1379-412. [PMID: 19789384 DOI: 10.1152/physrev.00005.2009] [Citation(s) in RCA: 716] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The opioid system consists of three receptors, mu, delta, and kappa, which are activated by endogenous opioid peptides processed from three protein precursors, proopiomelanocortin, proenkephalin, and prodynorphin. Opioid receptors are recruited in response to natural rewarding stimuli and drugs of abuse, and both endogenous opioids and their receptors are modified as addiction develops. Mechanisms whereby aberrant activation and modifications of the opioid system contribute to drug craving and relapse remain to be clarified. This review summarizes our present knowledge on brain sites where the endogenous opioid system controls hedonic responses and is modified in response to drugs of abuse in the rodent brain. We review 1) the latest data on the anatomy of the opioid system, 2) the consequences of local intracerebral pharmacological manipulation of the opioid system on reinforced behaviors, 3) the consequences of gene knockout on reinforced behaviors and drug dependence, and 4) the consequences of chronic exposure to drugs of abuse on expression levels of opioid system genes. Future studies will establish key molecular actors of the system and neural sites where opioid peptides and receptors contribute to the onset of addictive disorders. Combined with data from human and nonhuman primate (not reviewed here), research in this extremely active field has implications both for our understanding of the biology of addiction and for therapeutic interventions to treat the disorder.
Collapse
Affiliation(s)
- Julie Le Merrer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Département Neurobiologie et Génétique, Illkirch, France
| | | | | | | |
Collapse
|
36
|
Drug-induced and genetic alterations in stress-responsive systems: Implications for specific addictive diseases. Brain Res 2009; 1314:235-52. [PMID: 19914222 DOI: 10.1016/j.brainres.2009.11.015] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2009] [Revised: 11/03/2009] [Accepted: 11/06/2009] [Indexed: 11/22/2022]
Abstract
From the earliest work in our laboratory, we hypothesized, and with studies conducted in both clinical research and animal models, we have shown that drugs of abuse, administered or self-administered, on a chronic basis, profoundly alter stress-responsive systems. Alterations of expression of specific genes involved in stress responsivity, with increases or decreases in mRNA levels, receptor, and neuropeptide levels, and resultant changes in hormone levels, have been documented to occur after chronic intermittent exposure to heroin, morphine, other opiates, cocaine, other stimulants, and alcohol in animal models and in human molecular genetics. The best studied of the stress-responsive systems in humans and mammalian species in general is undoubtedly the HPA axis. In addition, there are stress-responsive systems in other parts in the brain itself, and some of these include components of the HPA axis, such as CRF and CRF receptors, along with POMC gene and gene products. Several other stress-responsive systems are known to influence the HPA axis, such as the vasopressin-vasopressin receptor system. Orexin-hypocretin, acting at its receptors, may effect changes which suggest that it should be properly categorized as a stress-responsive system. However, less is known about the interactions and connectivity of some of these different neuropeptide and receptor systems, and in particular, about the possible connectivity of fast-acting (e.g., glutamate and GABA) and slow-acting (including dopamine, serotonin, and norepinephrine) neurotransmitters with each of these stress-responsive components and the resultant impact, especially in the setting of chronic exposure to drugs of abuse. Several of these stress-responsive systems and components, primarily based on our laboratory-based and human molecular genetics research of addictive diseases, will be briefly discussed in this review.
Collapse
|
37
|
Sticht M, Mitsubata J, Tucci M, Leri F. Reacquisition of heroin and cocaine place preference involves a memory consolidation process sensitive to systemic and intra-ventral tegmental area naloxone. Neurobiol Learn Mem 2009; 93:248-60. [PMID: 19857583 DOI: 10.1016/j.nlm.2009.10.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2009] [Revised: 09/29/2009] [Accepted: 10/20/2009] [Indexed: 02/08/2023]
Abstract
To investigate the effect of naloxone on a putative memory consolidation process underlying reacquisition of heroin and cocaine conditioned place preference, four studies were conducted in male Sprague-Dawley rats using a common procedure involving: place conditioning (0.3 or 1mg/kg heroin or 20mg/kg cocaine; x4 sessions), extinction (vehiclex4 sessions), and reconditioning (0 or 1mg/kg heroin or 20mg/kg cocaine; x1 session). Systemic naloxone injections (0, 1 and 3mg/kg) or bilateral intra-ventral tegmental area (VTA) naloxone methiodide infusions (2 nmol in 0.5 microl x side) were administered at different times following reconditioning. Post-reconditioning administration of naloxone dose-dependently blocked, attenuated and had no effect on reacquisition of heroin CPP when administered immediately, 1h and 6h after reconditioning, respectively. The highest dose of naloxone also blocked reacquisition of cocaine CPP, and did not produce a conditioned place aversion in heroin-naïve and heroin pre-treated animals. Post-reconditioning infusions in the VTA, but not in adjacent structures, blocked reacquisition of heroin CPP when administered immediately, but not 6h, after reconditioning. These data suggest that reacquisition of drug-cues associations involves a memory consolidation process sensitive to manipulations of the endogenous opioid system, and indicate that opioid receptors in the VTA may be critically involved in the re-emergence of drug seeking behavior.
Collapse
Affiliation(s)
- Martin Sticht
- Department of Psychology, University of Guelph, Guelph, Canada N1G 2W1
| | | | | | | |
Collapse
|
38
|
Leri F, Zhou Y, Goddard B, Levy A, Jacklin D, Kreek MJ. Steady-state methadone blocks cocaine seeking and cocaine-induced gene expression alterations in the rat brain. Eur Neuropsychopharmacol 2009; 19:238-49. [PMID: 18990547 PMCID: PMC2900837 DOI: 10.1016/j.euroneuro.2008.09.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2008] [Revised: 08/22/2008] [Accepted: 09/30/2008] [Indexed: 11/26/2022]
Abstract
To elucidate the effects of steady-state methadone exposure on responding to cocaine conditioned stimuli and on cocaine-induced alterations in central opioid, hypocretin/orexin, and D2 receptor systems, male Sprague-Dawley rats received intravenous infusions of 1 mg/kg/inf cocaine paired with an audiovisual stimulus over three days of conditioning. Then, mini pumps releasing vehicle or 30 mg/kg/day methadone were implanted (SC), and lever pressing for the stimulus was assessed in the absence of cocaine and after a cocaine prime (20 mg/kg, IP). It was found that rats treated with vehicle, but not methadone, responded for the cocaine conditioned stimulus and displayed elevated mu-opioid receptor mRNA expression in the nucleus accumbens core and basolateral amygdala, reduced hypocretin/orexin mRNA in the lateral hypothalamus, and reduced D2 receptor mRNA in the caudate-putamen. This is the first demonstration that steady-state methadone administered after cocaine exposure blocks cocaine-induced behavioral and neural adaptations.
Collapse
MESH Headings
- Acoustic Stimulation/methods
- Analgesics, Opioid/administration & dosage
- Analysis of Variance
- Animals
- Behavior, Animal/drug effects
- Brain/drug effects
- Cocaine/pharmacology
- Cocaine-Related Disorders/drug therapy
- Cocaine-Related Disorders/metabolism
- Conditioning, Classical/drug effects
- Conditioning, Operant/drug effects
- Dopamine Uptake Inhibitors/pharmacology
- Drug Administration Schedule
- Drug Delivery Systems/methods
- Dynorphins/genetics
- Dynorphins/metabolism
- Gene Expression Regulation/drug effects
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Locomotion/drug effects
- Male
- Methadone/administration & dosage
- Neuropeptides/genetics
- Neuropeptides/metabolism
- Orexins
- Photic Stimulation/methods
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Dopamine D2/genetics
- Receptors, Dopamine D2/metabolism
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
Collapse
Affiliation(s)
- Francesco Leri
- Department of Psychology, University of Guelph, Guelph, Canada.
| | | | | | | | | | | |
Collapse
|
39
|
Nielsen DA, Yuferov V, Hamon S, Jackson C, Ho A, Ott J, Kreek MJ. Increased OPRM1 DNA methylation in lymphocytes of methadone-maintained former heroin addicts. Neuropsychopharmacology 2009; 34:867-73. [PMID: 18650805 PMCID: PMC2778040 DOI: 10.1038/npp.2008.108] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2008] [Revised: 05/23/2008] [Accepted: 06/16/2008] [Indexed: 01/19/2023]
Abstract
The mu-opioid receptor is the site of action of opiates and opioids. We examined whether there are differences in cytosine:guanine (CpG) dinucleotide methylation in the OPRM1 promoter between former heroin addicts and controls. We analyzed methylation at 16 CpG dinucleotides in DNA obtained from lymphocytes of 194 Caucasian former severe heroin addicts stabilized in methadone maintenance treatment and 135 Caucasian control subjects. Direct sequencing of bisulfite-treated DNA showed that the percent methylation at two CpG sites was significantly associated with heroin addiction. The level of methylation at the -18 CpG site was 25.4% in the stabilized methadone-maintained former heroin addicts and 21.4% in controls (p=0.0035, generalized estimating equations (GEE); p=0.0077, t-test; false discovery rate (FDR)=0.048), and the level of methylation at the +84 CpG dinucleotide site was 7.4% in cases and 5.6% in controls (p=0.0095, GEE; p=0.0067, t-test; FDR=0.080). Both the -18 and the +84 CpG sites are located in potential Sp1 transcription factor-binding sites. Methylation of these CpG sites may lead to reduced OPRM1 expression in the lymphocytes of these former heroin addicts.
Collapse
Affiliation(s)
- David A Nielsen
- Laboratory of the Biology of the Addictive Diseases, The Rockefeller University, New York, NY 10065, USA.
| | | | | | | | | | | | | |
Collapse
|
40
|
Kreek MJ, Zhou Y, Butelman ER, Levran O. Opiate and cocaine addiction: from bench to clinic and back to the bench. Curr Opin Pharmacol 2009; 9:74-80. [PMID: 19155191 PMCID: PMC2741727 DOI: 10.1016/j.coph.2008.12.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2008] [Revised: 12/23/2008] [Accepted: 12/23/2008] [Indexed: 11/28/2022]
Abstract
This review primarily focuses on our recent findings in bidirectional translational research on opiate and cocaine addictions. First, we present neurobiological and molecular studies on endogenous opioid systems (e.g. proopiomelanocortin, mu opioid receptor, dynorphin, and kappa opioid receptor), brain stress-responsive systems (e.g. orexin, arginine vasopressin, V1b receptor, and corticotropin-releasing factor), hypothalamic-pituitary-adrenal axis, and neurotransmitters (especially dopamine), in response to both chronic cocaine or opiate exposure and to drug withdrawal, using several newly developed animal models and molecular approaches. The second aspect is human molecular genetic association investigations including hypothesis-driven studies and genome-wide array studies, to define particular systems involved in vulnerability to develop specific addictions, and response to pharmacotherapy.
Collapse
Affiliation(s)
- Mary Jeanne Kreek
- The Rockefeller University, Laboratory of the Biology of Addictive Diseases, 1230 York Avenue, Box 171, New York, NY 10065
| | - Yan Zhou
- The Rockefeller University, Laboratory of the Biology of Addictive Diseases, 1230 York Avenue, Box 171, New York, NY 10065
| | - Eduardo R. Butelman
- The Rockefeller University, Laboratory of the Biology of Addictive Diseases, 1230 York Avenue, Box 171, New York, NY 10065
| | - Orna Levran
- The Rockefeller University, Laboratory of the Biology of Addictive Diseases, 1230 York Avenue, Box 171, New York, NY 10065
| |
Collapse
|
41
|
Brown TE, Lee BR, Sorg BA. The NMDA antagonist MK-801 disrupts reconsolidation of a cocaine-associated memory for conditioned place preference but not for self-administration in rats. Learn Mem 2008; 15:857-65. [PMID: 19050157 DOI: 10.1101/lm.1152808] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Recent research suggests that drug-related memories are reactivated after exposure to environmental cues and may undergo reconsolidation, a process that can strengthen memories. Conversely, reconsolidation may be disrupted by certain pharmacological agents such that the drug-associated memory is weakened. Several studies have demonstrated disruption of memory reconsolidation using a drug-induced conditioned place preference (CPP) task, but no studies have explored whether cocaine-associated memories can be similarly disrupted in cocaine self-administering animals after a cocaine priming injection, which powerfully reinstates drug-seeking behavior. Here we used cocaine-induced CPP and cocaine self-administration to investigate whether the N-methyl-D-aspartate receptor antagonist (+)-5methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine maleate (MK-801) given just prior to reactivation sessions would suppress subsequent cocaine-primed reinstatement (disruption of reconsolidation). Systemic injection of MK-801 (0.05 or 0.20 mg/kg administered intraperitoneally) in rats just prior to reactivation of the cocaine-associated memory in the CPP context attenuated subsequent cocaine-primed reinstatement, while no disruption occurred in rats that did not receive reactivation in the CPP context. However, in rats trained to self-administer cocaine, systemic administration of MK-801 just prior to either of two different types of reactivation sessions had no effect on subsequent cocaine-primed reinstatement of lever-pressing behavior. Thus, systemic administration of MK-801 disrupted the reconsolidation of a cocaine-associated memory for CPP but not for self-administration. These findings suggest that cocaine-CPP and self-administration do not use similar neurochemical processes to disrupt reconsolidation or that cocaine-associated memories in self-administering rats do not undergo reconsolidation, as assessed by lever-pressing behavior under cocaine reinstatement conditions.
Collapse
Affiliation(s)
- Travis E Brown
- Alcohol and Drug Abuse Research Program and Program in Neuroscience, Department of Veterinary and Comparative Anatomy, Pharmacology and Physiology, Washington State University, Pullman, Washington 99164-6520, USA
| | | | | |
Collapse
|
42
|
Devidze N, Zhou Y, Ho A, Zhang Q, Pfaff D, Kreek M. Steady-state methadone effect on generalized arousal in male and female mice. Behav Neurosci 2008; 122:1248-56. [PMID: 19045944 PMCID: PMC2672421 DOI: 10.1037/a0013276] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Methadone is widely used in treatment of short-acting opiate addiction. The on-off effects of opioids have been documented to have profound differences from steady-state opioids. The authors hypothesize that opioids play important roles in either generalized arousal (GA) or aversive state of arousal during opioid withdrawal. Both male and female C57BL6 mice received steady-state methadone (SSM) through osmotic pumps at 10 or 20 mg/kg/day, and GA was measured in voluntary motor activity, sensory responsivity, and contextual fear conditioning. SSM did not have any effect on those GA behaviors in either sex. Females had higher activity and less fear conditioning than males. The effects of SSM on stress-responsive orexin gene expression in the lateral hypothalamus (LH) and medial hypothalamus (MH, including perifornical and dorsomedial areas) were measured after the behavioral tests. Females showed significantly lower basal LH (but not MH) orexin mRNA levels than males. A panel of GA stressors increased LH orexin mRNA levels in females only; these increases were blunted by SSM at 20 mg/kg. In summary, SSM had no effect on GA behaviors. In females, SSM blunted the GA stress-induced LH orexin gene expression.
Collapse
Affiliation(s)
- N. Devidze
- Laboratory of Neurobiology and Behavior; The Rockefeller University, New York, NY
| | - Y. Zhou
- Laboratory of the Biology of Addictive Diseases; The Rockefeller University, New York, NY
| | - A. Ho
- Laboratory of the Biology of Addictive Diseases; The Rockefeller University, New York, NY
| | - Q. Zhang
- Laboratory of Neurobiology and Behavior; The Rockefeller University, New York, NY
| | - D.W. Pfaff
- Laboratory of Neurobiology and Behavior; The Rockefeller University, New York, NY
| | - M.J. Kreek
- Laboratory of the Biology of Addictive Diseases; The Rockefeller University, New York, NY
| |
Collapse
|
43
|
Kreek MJ, Schlussman SD, Reed B, Zhang Y, Nielsen DA, Levran O, Zhou Y, Butelman ER. Bidirectional translational research: Progress in understanding addictive diseases. Neuropharmacology 2008; 56 Suppl 1:32-43. [PMID: 18725235 PMCID: PMC2741728 DOI: 10.1016/j.neuropharm.2008.07.042] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2008] [Revised: 07/29/2008] [Accepted: 07/30/2008] [Indexed: 11/17/2022]
Abstract
The focus of this review is primarily on recent developments in bidirectional translational research on the addictions, within the Laboratory of the Biology of Addictive Diseases at The Rockefeller University. This review is subdivided into major interacting aspects, including (a) Investigation of neurobiological and molecular adaptations (e.g., in genes for the opioid receptors or endogenous neuropeptides) in response to cocaine or opiates, administered under laboratory conditions modeling chronic patterns of human self-exposure (e.g., chronic escalating "binge"). (b) The impact of such drug exposure on the hypothalamic-pituitary-adrenal (HPA) axis and interacting neuropeptidergic systems (e.g., opioid, orexin and vasopressin). (c) Molecular genetic association studies using candidate gene and whole genome approaches, to define particular systems involved in vulnerability to develop specific addictions, and response to pharmacotherapy. (d) Neuroendocrine challenge studies in normal volunteers and current addictive disease patients along with former addicts in treatment, to investigate differential pharmacodynamics and responsiveness of molecular targets, in particular those also investigated in the experimental and molecular genetic approaches as described above.
Collapse
Affiliation(s)
- M J Kreek
- Laboratory of the Biology of Addictive Diseases, Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Zhou Y, Cui CL, Schlussman SD, Choi JC, Ho A, Han JS, Kreek MJ. Effects of cocaine place conditioning, chronic escalating-dose "binge" pattern cocaine administration and acute withdrawal on orexin/hypocretin and preprodynorphin gene expressions in lateral hypothalamus of Fischer and Sprague-Dawley rats. Neuroscience 2008; 153:1225-34. [PMID: 18436386 PMCID: PMC2494861 DOI: 10.1016/j.neuroscience.2008.03.023] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2007] [Revised: 02/02/2008] [Accepted: 03/12/2008] [Indexed: 10/22/2022]
Abstract
Recent evidence suggests an important role for hypothalamic orexins/hypocretins in modulation of drug reward and addiction-like behaviors in rodents. Our recent study has shown that the aversive state of arousal during acute morphine withdrawal is associated with increased orexin gene expression in lateral hypothalamus (LH) of Fischer 344 (F344) inbred rats, with no change in the expression of preprodynorphin (ppDyn), a gene co-expressed with LH orexin. Therefore, we determined whether orexin and ppDyn mRNA levels in LH or medial hypothalamus (including perifornical and dorsomedial areas) of F344 or Sprague-Dawley (SD) outbred rats, are altered following: 1) cocaine (10 mg/kg, i.p.) conditioned place preference (CPP); 2) chronic (14 days) cocaine exposure using both "binge" pattern administration in steady-dose (45 mg/kg/day) and escalating-dose (45-90 mg/kg/day) regimens; and 3) acute (1 day) and chronic (14 days) withdrawal from cocaine with opioid receptor antagonist naloxone treatment (1 mg/kg). We found that orexin mRNA levels were decreased after cocaine place conditioning in the LH of SD rats. A decreased LH orexin mRNA level was also observed after chronic escalating-dose cocaine (but not CPP pattern regimen without conditioning, or steady-dose regimen) in both strains. In F344 rats only, acute withdrawal from chronic escalating-dose cocaine administration resulted in increases in both LH orexin and ppDyn mRNA levels, which were unaltered by naloxone or after chronic withdrawal. Our results suggest that (1) alteration of LH orexin gene expression is region-specific after cocaine place conditioning in SD rats and dose-dependent after chronic exposure in both strains; and (2) increased LH orexin and ppDyn gene expressions in F344 rats may contribute to negative affective states in cocaine withdrawal.
Collapse
Affiliation(s)
- Y Zhou
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
The articulated goals of Dialogues in Clinical Neuroscience are to serve as "an interface between clinical neuropsychiatry and the neurosciences by providing state-of-the-art information and original insights into relevant clinical, biological, and therapeutic aspects." My laboratory the Laboratory of the Biology of Addictive Diseases at The Rockefeller University, has for years been focused on "bidirectional translational research," that is, learning by careful observations and study in patient populations with the disorders under study, in this case primarily specific addictive diseases, and then using that knowledge to create improved animal models or other laboratory-based research paradigms, while, at the same time, taking research findings made at the bench into the clinic as promptly as that is appropriate and feasible. In this invited review, therefore, the focus will be on perspectives of our Laboratory of the Biology of Addictive Diseases and related National Institutes of Health/National Institute on Drug Abuse research Center, including laboratory-based molecular neurobiological research, research using several animal models designed to mimic human patterns of drug abuse and addiction, as well as basic clinical research, intertwined with treatment-related research.
Collapse
Affiliation(s)
- Mary Jeanne Kreek
- Laboratory of Biology of Addictive Diseases, Rockefeller University, New York, NY 10021, USA.
| |
Collapse
|
46
|
Zhou Y, Leri F, Cummins E, Hoeschele M, Kreek MJ. Involvement of arginine vasopressin and V1b receptor in heroin withdrawal and heroin seeking precipitated by stress and by heroin. Neuropsychopharmacology 2008; 33:226-36. [PMID: 17443128 DOI: 10.1038/sj.npp.1301419] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
A previous study has shown that the stress responsive neurohormone arginine vasopressin (AVP) is activated in the amygdala during early withdrawal from cocaine. The present studies were undertaken to determine whether (1) AVP mRNA levels in the amygdala or hypothalamus, as well as hypothalamic-pituitary-adrenal (HPA) activity, would be altered during chronic intermittent escalating heroin administration (10 days; 7.5-60 mg/kg/day) or during early (12 h) and late (10 days) spontaneous withdrawal; (2) foot shock stress would alter AVP mRNA levels in the amygdala or hypothalamus in rats withdrawn from heroin self-administration (7 days, 3 h/day, 0.05 mg/kg/infusion); and (3) the selective V1b receptor antagonist SSR149415 (1 and 30 mg/kg, intraperitoneal) would alter heroin seeking during tests of reinstatement induced by foot shock stress and by heroin primes (0.25 mg/kg), as well as HPA hormonal responses to foot shock. We found that AVP mRNA levels were increased during early spontaneous withdrawal in the amygdala only. This amygdalar AVP mRNA increase was no longer observed at the later stage of heroin withdrawal. Foot shock stress increased AVP mRNA levels in the amygdala of rats withdrawn from heroin self-administration, but not in heroin naïve rats. Behaviorally, SSR149415 dose-dependently attenuated foot shock-induced reinstatement and blocked heroin-induced reinstatement. Finally, SSR149415 blunted the HPA activation by foot shock. Together, these data in rats suggest that stress responsive AVP/V1b receptor systems (including the amygdala) may be critical components of the neural circuitry underlying the aversive emotional consequences of drug withdrawal, as well as the effect of negative emotional states on drug-seeking behavior.
Collapse
Affiliation(s)
- Yan Zhou
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10021, USA.
| | | | | | | | | |
Collapse
|
47
|
Leri F, Sorge RE, Cummins E, Woehrling D, Pfaus JG, Stewart J. High-dose methadone maintenance in rats: effects on cocaine self-administration and behavioral side effects. Neuropsychopharmacology 2007; 32:2290-300. [PMID: 17314916 DOI: 10.1038/sj.npp.1301357] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
It has been demonstrated that high-dose methadone maintenance is efficacious in reducing cocaine abuse in opioid-dependent individuals, but it is not clear whether this is caused by an action of methadone on the direct reinforcing properties of cocaine or on cocaine seeking. Also, it is not clear whether high-dose methadone maintenance may induce behavioral side effects, which could limit its clinical use. Here, we report that high-dose methadone maintenance (20-40 mg/kg/day) does not reduce, and even enhances cocaine (10-30 mg/kg, i.p.)-induced elevation in dopamine concentration in the ventral striatum measured by in vivo microdialysis. In parallel, however, rats maintained on high-dose methadone (30 mg/kg/day) seek and consume significantly less cocaine than controls when tested for intravenous cocaine (0.5 mg/kg/infusion) self-administration on a progressive ratio schedule of reinforcement. This reduction in cocaine self-administration does not result from impaired sensory-motor functioning as rats maintained on high-dose methadone show normal locomotor activity. Furthermore, the reduction in responding for cocaine does not seem to result from general behavioral deficits as male rats maintained on high methadone doses respond normally to palatable food and thermal pain, although their sexual responses to receptive females are greatly suppressed. Taken together, these results from studies in rats support the usefulness of larger doses of methadone to reduce severe cocaine abuse in opioid-dependent individuals and possibly in the management of pure-cocaine addiction.
Collapse
Affiliation(s)
- Francesco Leri
- Department of Psychology, University of Guelph, Guelph, ON, Canada NIG 2WI.
| | | | | | | | | | | |
Collapse
|
48
|
Tzschentke TM. Measuring reward with the conditioned place preference (CPP) paradigm: update of the last decade. Addict Biol 2007; 12:227-462. [PMID: 17678505 DOI: 10.1111/j.1369-1600.2007.00070.x] [Citation(s) in RCA: 1032] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Conditioned place preference (CPP) continues to be one of the most popular models to study the motivational effects of drugs and non-drug treatments in experimental animals. This is obvious from a steady year-to-year increase in the number of publications reporting the use this model. Since the compilation of the preceding review in 1998, more than 1000 new studies using place conditioning have been published, and the aim of the present review is to provide an overview of these recent publications. There are a number of trends and developments that are obvious in the literature of the last decade. First, as more and more knockout and transgenic animals become available, place conditioning is increasingly used to assess the motivational effects of drugs or non-drug rewards in genetically modified animals. Second, there is a still small but growing literature on the use of place conditioning to study the motivational aspects of pain, a field of pre-clinical research that has so far received little attention, because of the lack of appropriate animal models. Third, place conditioning continues to be widely used to study tolerance and sensitization to the rewarding effects of drugs induced by pre-treatment regimens. Fourth, extinction/reinstatement procedures in place conditioning are becoming increasingly popular. This interesting approach is thought to model certain aspects of relapse to addictive behavior and has previously almost exclusively been studied in drug self-administration paradigms. It has now also become established in the place conditioning literature and provides an additional and technically easy approach to this important phenomenon. The enormous number of studies to be covered in this review prevented in-depth discussion of many methodological, pharmacological or neurobiological aspects; to a large extent, the presentation of data had to be limited to a short and condensed summary of the most relevant findings.
Collapse
Affiliation(s)
- Thomas M Tzschentke
- Grünenthal GmbH, Preclinical Research and Development, Department of Pharmacology, Aachen, Germany.
| |
Collapse
|
49
|
Zhou Y, Adomako-Mensah J, Yuferov V, Ho A, Zhang J, Xu M, Kreek MJ. Effects of acute “binge” cocaine on mRNA levels of μ opioid receptor and neuropeptides in dopamine D1 or D3 receptor knockout mice. Synapse 2006; 61:50-9. [PMID: 17068774 DOI: 10.1002/syn.20340] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
In humans, elevations of mu opioid receptor (MOP-r) binding potential (BP) in the frontal cortex (FC) are associated with cocaine craving during early abstinence. In rats, decreases in dopaminergic (DAergic) transmission in the medial FC are associated with increased cocaine-seeking behavior. DA D1 or D3 receptor homozygous knockout (D1-/- or D3-/-) mice offer the opportunity to test the roles of these specific receptors in regulating MOP-r gene expression in response to cocaine. In the present studies, we found an increase in basal MOP-r mRNA levels in the FC of both D1-/- and D3-/- mice compared to wild type controls, with no change in the nucleus accumbens (NAc) core or caudate-putamen (CPu). Acute "binge" cocaine (3 x 15 mg/kg for 2.5 h) returned FC MOP-r mRNA levels in D1-/- or D3-/- mice to those in wild type controls. In the NAc core, the MOP-r mRNA levels after acute "binge" cocaine were decreased in D1-/- mice while increased in D3-/- mice. In the CPu, however, the MOP-r mRNA levels after acute "binge" cocaine were increased in D1-/- mice while decreased in D3-/- mice. We also found a decrease in basal orexin mRNA levels in the lateral hypothalamus of the D3-/- mice, which was unaltered by acute "binge" cocaine. Together, our findings suggest that: (1) both D1 and D3 receptors are involved in FC MOP-r gene regulation; and (2) D1 and D3 receptors play opposite roles in the effects of cocaine on MOP-r gene regulation differentially in the NAc core or CPu.
Collapse
Affiliation(s)
- Yan Zhou
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, New York 10021, USA.
| | | | | | | | | | | | | |
Collapse
|