1
|
Wu H, Song Z, Chen Q, Yan R, Zhao H, Li H. Disrupting reconsolidation by systemic inhibition of Thioredoxin-1 attenuates cocaine and morphine relapse. Biomed Pharmacother 2025; 186:118037. [PMID: 40199134 DOI: 10.1016/j.biopha.2025.118037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/30/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025] Open
Abstract
The enduring nature of drug-associated memories is an essential factor contributing to the relapse. Drug-related cues can activate drug memories, making them enter reconsolidation, during which interventions can effectively disrupt these memories. Interventions targeting memory reconsolidation present a promising therapeutic strategy for addressing substance use disorders (SUDs). Oxidative stress can disrupt neural function and impair memory. Thioredoxin-1 (Trx-1) effectively alleviates oxidative stress and reduces inflammation levels. However, few studies have connected Trx-1 to drug memory or explored its specific role in reconsolidation. This research employed the conditioned place preference (CPP) model to investigate the effects of Trx-1 inhibitors on the reconsolidation of morphine- and cocaine-related memories. Results show that immediate administration of PX-12, a Trx-1 inhibitor, after retrieval significantly attenuated the reinstatement of cocaine and morphine CPP induced by both cues and the drug itself, with the effect lasting for at least 14 days. In contrast, the inhibition of Trx-1, either 6 hours following retrieval or in the absence of retrieval, does not influence drug-seeking behaviors associated with cocaine or morphine. Furthermore, Trx-1 inhibitor itself did not produce any preferences. In summary, our results indicate that Trx-1 activity is crucial for cocaine- and morphine-related memories, and that the Trx-1 inhibitor may serve as a potential treatment for drug abuse.
Collapse
Affiliation(s)
- Hao Wu
- Department of Anesthesiology, Peking Union Medical College Hospital, Peking Union Medical College, and Chinese Academy of Medical Science, Beijing, China
| | - Zhihao Song
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center for Skull Base Surgery and Neurooncology in Hunan Province, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qijun Chen
- Department of Clinical Laboratory, The Affiliated Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou 510620, China
| | - Ruyu Yan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center for Skull Base Surgery and Neurooncology in Hunan Province, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Haiting Zhao
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| | - Haoyu Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center for Skull Base Surgery and Neurooncology in Hunan Province, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
2
|
Xie B, Wang Y, Lu Y, Wang M, Hui R, Yu H, Li W, Zhang L, Yu F, Ni Z, Cong B, Ma C, Wen D. A novel intervention of molecular hydrogen on the unbalance of the gut microbiome in opioid addiction: Experimental and human studies. Biomed Pharmacother 2024; 178:117273. [PMID: 39116782 DOI: 10.1016/j.biopha.2024.117273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/04/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024] Open
Abstract
The gut-brain axis mediates the interaction pathway between microbiota and opioid addiction. In recent years, many studies have shown that molecular hydrogen has therapeutic and preventive effects on various diseases. This study aimed to investigate whether molecular hydrogen could serve as pharmacological intervention agent to reduce risks of reinstatement of opioid seeking and explore the mechanism of gut microbiota base on animal experiments and human studies. Morphine-induced conditioned place preference (CPP) was constructed to establish acquisition, extinction, and reinstatement stage, and the potential impact of H2 on the behaviors related to morphine-induced drug extinction was determined using both free accessible and confined CPP extinction paradigms. The effects of morphine on microbial diversity and composition of microbiota, as well as the subsequent changes after H2 intervention, were assessed using 16 S rRNA gene sequencing. Short-Chain Fatty Acids (SCFAs) in mice serum were detected by gas chromatography-mass spectrometry (GC-MS). Meanwhile, we also conducted molecular hydrogen intervention and gut microbiota testing in opioid-addicted individuals. Our results revealed that molecular hydrogen could enhance the extinction of morphine-related behavior, reducing morphine reinstatement. Gut microbes may be a potential mechanism behind the therapeutic effects of molecular hydrogen on morphine addiction. Additionally, molecular hydrogen improved symptoms of depression and anxiety, as well as gut microbial features, in individuals with opioid addiction. This study supports molecular hydrogen as a novel and effective intervention for morphine-induced addiction and reveals the mechanism of gut microbiota.
Collapse
Affiliation(s)
- Bing Xie
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Yong Wang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Yun Lu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Mengmeng Wang
- Affiliated Hospital of Hebei University, College of Clinical Medicine, Hebei University, Collaborative Innovation Center of Tumor Microecological Metabolism Regulation, Baoding, Hebei Province 071000, PR China
| | - Rongji Hui
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Hailei Yu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Wenbo Li
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Ludi Zhang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, Hebei Province 050017, PR China
| | - Feng Yu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Zhiyu Ni
- Affiliated Hospital of Hebei University, College of Clinical Medicine, Hebei University, Collaborative Innovation Center of Tumor Microecological Metabolism Regulation, Baoding, Hebei Province 071000, PR China; Clinical Medical College, Hebei University of Engineering, Handan, Hebei Province 056038, PR China
| | - Bin Cong
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Chunling Ma
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China.
| | - Di Wen
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China.
| |
Collapse
|
3
|
Alvarado-Torres JK, Morales-Silva R, Sanabria Ponce de Leon A, Rodriguez-Torres G, Perez-Torres J, Perez-Perez Y, Mueller D, Sepulveda-Orengo MT. Estradiol reduction through aromatase inhibition impairs cocaine seeking in male rats. Front Behav Neurosci 2024; 17:1307606. [PMID: 38292056 PMCID: PMC10824998 DOI: 10.3389/fnbeh.2023.1307606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/26/2023] [Indexed: 02/01/2024] Open
Abstract
Introduction Clinical and preclinical research on cocaine use disorder (CUD) has shown that sex differences in drug seeking are influenced by hormonal fluctuations. Estradiol (E2), a sex steroid hormone, has been linked to female drug effects, vulnerability to use/abuse, and psychosocial factors. Preclinical studies show that estradiol in females facilitates the extinction of cocaine-seeking behavior indicating a possible role in regulating extinction learning. Similar to females, males' brains contain the aromatase enzyme which converts testosterone to estradiol. However, it is unclear whether estradiol plays a role in male extinction learning as it does in females. Furthermore, how endogenously aromatized estradiol affects drug addiction in males is unknown. Therefore, this study investigated whether endogenous estradiol regulates cocaine seeking in male rats. We hypothesized that decreased aromatase enzyme activity, resulting in decreased estradiol synthesis in male brains, will impair extinction learning leading to increased cocaine-seeking behavior. Methods This hypothesis was tested using cocaine-conditioned place preference (CPP), and short access self-administration (SA), followed by extinction and reinstatement. Before each extinction session for CPP or SA, male rats received an injection of either 1 (low dose) or 2.5 mg/kg (high dose) of the aromatase inhibitor Fadrozole (FAD), or vehicle. Results FAD groups showed dose-dependent effects on cocaine-seeking behavior compared to the vehicle group during CPP extinction. Specifically, low dose FAD facilitated extinction of cocaine CPP, whereas high dose FAD impaired it. In contrast, neither dose of FAD had any effects on the extinction of cocaine SA. Interestingly, only the low dose FAD group had decreased active lever pressing during cue- and cocaine-primed reinstatement compared to the vehicle group. Neither dose of FAD had an effect on sucrose extinction or reinstatement of sucrose seeking. Discussion These results from CPP experiments suggest that estradiol may impact extinction learning, as a low dose of FAD may strengthen the formation of cocaine extinction memory. Additionally, in male rats undergoing cocaine SA, the same low dose of aromatase inhibitor effectively reduced reinstatement of cocaine-seeking behavior. Thus, estradiol impacts cocaine seeking and extinction in both males and females, and it may also influence the development of sex-specific treatment strategies for CUD.
Collapse
Affiliation(s)
- John K. Alvarado-Torres
- Basic Sciences Department, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Roberto Morales-Silva
- Basic Sciences Department, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | | | - Genesis Rodriguez-Torres
- Basic Sciences Department, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Joshua Perez-Torres
- Basic Sciences Department, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Yobet Perez-Perez
- Basic Sciences Department, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Devin Mueller
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Marian T. Sepulveda-Orengo
- Basic Sciences Department, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| |
Collapse
|
4
|
Domi E, Barchiesi R, Barbier E. Epigenetic Dysregulation in Alcohol-Associated Behaviors: Preclinical and Clinical Evidence. Curr Top Behav Neurosci 2023. [PMID: 36717533 DOI: 10.1007/7854_2022_410] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Alcohol use disorder (AUD) is characterized by loss of control over intake and drinking despite harmful consequences. At a molecular level, AUD is associated with long-term neuroadaptations in key brain regions that are involved in reward processing and decision-making. Over the last decades, a great effort has been made to understand the neurobiological basis underlying AUD. Epigenetic mechanisms have emerged as an important mechanism in the regulation of long-term alcohol-induced gene expression changes. Here, we review the literature supporting a role for epigenetic processes in AUD. We particularly focused on the three most studied epigenetic mechanisms: DNA methylation, Histone modification and non-coding RNAs. Clinical studies indicate an association between AUD and DNA methylation both at the gene and global levels. Using behavioral paradigms that mimic some of the characteristics of AUD, preclinical studies demonstrate that changes in epigenetic mechanisms can functionally impact alcohol-associated behaviors. While many studies support a therapeutic potential for targeting epigenetic enzymes, more research is needed to fully understand their role in AUD. Identification of brain circuits underlying alcohol-associated behaviors has made major advances in recent years. However, there are very few studies that investigate how epigenetic mechanisms can affect these circuits or impact the neuronal ensembles that promote alcohol-associated behaviors. Studies that focus on the role of circuit-specific and cell-specific epigenetic changes for clinically relevant alcohol behaviors may provide new insights on the functional role of epigenetic processes in AUD.
Collapse
Affiliation(s)
- Esi Domi
- Department of Biomedical and Clinical Sciences, Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
- School of Pharmacy, Pharmacology Unit, Center for Neuroscience, University of Camerino, Camerino, Italy
| | - Riccardo Barchiesi
- Department of Neuroscience, Waggoner Center for Alcohol and Alcohol Addiction Research, University of Texas at Austin, Austin, TX, USA
| | - Estelle Barbier
- Department of Biomedical and Clinical Sciences, Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden.
| |
Collapse
|
5
|
High Morphine Use Disorder Susceptibility Is Predicted by Impaired Learning Ability in Mice. Brain Sci 2022; 12:brainsci12121650. [PMID: 36552110 PMCID: PMC9776386 DOI: 10.3390/brainsci12121650] [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: 09/24/2022] [Revised: 10/24/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
An obvious reason for substance uses disorders (SUDs) is drug craving and seeking behavior induced by conditioned context, which is an abnormal solid context memory. The relationship between susceptibility to SUD and learning ability remains unclear in humans and animal models. In this study, we found that susceptibility to morphine use disorder (MUD) was negatively correlated with learning ability in conditioned place preference (CPP) in C57 mice. By using behavioral tests, we identified the FVB mouse as learning impaired. In addition, we discovered that learning-relevant proteins, such as the glutamate receptor subunits GluA1, NR1, and NR2A, were decreased in FVB mice. Finally, we assessed the context learning ability of FVB mice using the CPP test and priming. We found that FVB mice had lower learning performance with respect to normal memory but higher performance of morphine-reinstatement memory. Compared to C57 mice, FVB mice are highly sensitive to MUDs. Our results suggest that SUD susceptibility is predicted by impaired learning ability in mice; therefore, learning ability can play a simple and practical role in identifying high-risk SUD groups.
Collapse
|
6
|
Atehortua Martinez LA, Curis E, Mekdad N, Larrieu C, Courtin C, Jourdren L, Blugeon C, Laplanche JL, Megarbane B, Marie-Claire C, Benturquia N. Individual differences in cocaine-induced conditioned place preference in male rats: Behavioral and transcriptomic evidence. J Psychopharmacol 2022; 36:1161-1175. [PMID: 36121009 PMCID: PMC9548661 DOI: 10.1177/02698811221123047] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Substance use disorder emerges in a small proportion of drug users and has the characteristics of a chronic relapsing pathology. AIMS Our study aimed to demonstrate and characterize the variability in the expression of the rewarding effects of cocaine in the conditioned place preference (CPP) paradigm. METHODS A cocaine-CPP paradigm in male Sprague-Dawley rats with an extinction period of 12 days and reinstatement was conducted. A statistical model was developed to distinguish rats expressing or not a cocaine-induced place preference. RESULTS Two groups of rats were identified: rats that did express rewarding effects (CPP expression (CPPE), score >102 s) and rats that did not (no CPP expression (nCPPE), score between -85 and 59 s). These two groups did not show significant differences in a battery of behavioral tests. To identify differentially expressed genes in the CPPE and nCPPE groups, a whole-transcriptome ribonucleic acid-sequencing analysis was performed in the nucleus accumbens (NAc) 24 h after the CPP test. Four immediate early genes (Fos, Egr2, Nr4a1, and Zbtb37) were differentially expressed in the NAc of CPPE rats after expression of CPP. Variability in cocaine-induced place preference persisted in the CPPE and nCPPE groups after the extinction and reinstatement phases. Transcriptomic differences observed after reinstatement were distinct from those observed immediately after expression of CPP. CONCLUSION These new findings provide insights into the identification of mechanisms underlying interindividual variability in the response to cocaine's rewarding effects.
Collapse
Affiliation(s)
- Luisa Alessandra Atehortua Martinez
- Institut National de la Santé et de la Recherche Médicale UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Paris, France
| | - Emmanuel Curis
- UR 7537 BioSTM, Université Paris Cité, Paris, France
- Laboratoire d’Hématologie, Hôpital Lariboisière, APHP, Paris, France
| | - Nawel Mekdad
- Institut National de la Santé et de la Recherche Médicale UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Paris, France
| | - Claire Larrieu
- Institut National de la Santé et de la Recherche Médicale UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Paris, France
| | - Cindie Courtin
- Institut National de la Santé et de la Recherche Médicale UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Paris, France
| | - Laurent Jourdren
- Genomic Facility, Institut de Biologie de l’École Normale Supérieure, École Normale Supérieure, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, PSL Research University, Paris, France
| | - Corinne Blugeon
- Genomic Facility, Institut de Biologie de l’École Normale Supérieure, École Normale Supérieure, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, PSL Research University, Paris, France
| | - Jean-Louis Laplanche
- Institut National de la Santé et de la Recherche Médicale UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Paris, France
| | - Bruno Megarbane
- Institut National de la Santé et de la Recherche Médicale UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Paris, France
| | - Cynthia Marie-Claire
- Institut National de la Santé et de la Recherche Médicale UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Paris, France
| | - Nadia Benturquia
- Institut National de la Santé et de la Recherche Médicale UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Paris, France
| |
Collapse
|
7
|
Marrero-Cristobal G, Gelpi-Dominguez U, Morales-Silva R, Alvarado-Torres J, Perez-Torres J, Perez-Perez Y, Sepulveda-Orengo M. Aerobic exercise as a promising nonpharmacological therapy for the treatment of substance use disorders. J Neurosci Res 2022; 100:1602-1642. [PMID: 34850988 PMCID: PMC9156662 DOI: 10.1002/jnr.24990] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 10/06/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022]
Abstract
Despite the prevalence and public health impact of substance use disorders (SUDs), effective long-term treatments remain elusive. Aerobic exercise is a promising, nonpharmacological treatment currently under investigation as a strategy for preventing drug relapse. Aerobic exercise could be incorporated into the comprehensive treatment regimens for people with substance abuse disorders. Preclinical studies of SUD with animal models have shown that aerobic exercise diminishes drug-seeking behavior, which leads to relapse, in both male and female rats. Nevertheless, little is known regarding the effects of substance abuse-induced cellular and physiological adaptations believed to be responsible for drug-seeking behavior. Accordingly, the overall goal of this review is to provide a summary and an assessment of findings to date, highlighting evidence of the molecular and neurological effects of exercise on adaptations associated with SUD.
Collapse
Affiliation(s)
| | - Ursula Gelpi-Dominguez
- School of Behavioral and Brain Sciences, Ponce Health Sciences University, Ponce, PR, USA
| | - Roberto Morales-Silva
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR, USA
| | - John Alvarado-Torres
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR, USA
| | - Joshua Perez-Torres
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR, USA
| | - Yobet Perez-Perez
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR, USA
| | - Marian Sepulveda-Orengo
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR, USA
| |
Collapse
|
8
|
Singh PK, Lutfy K. The Role of Beta-Endorphin in Cocaine-Induced Conditioned Place Preference, Its Extinction, and Reinstatement in Male and Female Mice. Front Behav Neurosci 2021; 15:763336. [PMID: 34955777 PMCID: PMC8702804 DOI: 10.3389/fnbeh.2021.763336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/16/2021] [Indexed: 11/21/2022] Open
Abstract
Endogenous opioids have been implicated in cocaine reward. However, the role of each opioid peptide in this regard is unknown. Notably, the role of each peptide in extinction and reinstatement is not fully characterized. Thus, we assessed whether cocaine-induced conditioned place preference (CPP) and its extinction and reinstatement would be altered in the absence of beta-endorphin. We also examined if sex-related differences would exist in these processes. Male and female mice lacking beta-endorphin and their respective controls were tested for baseline place preference on day 1. On day 2, mice were treated with saline/cocaine (15 mg/kg) and confined to the vehicle- or drug-paired chamber for 30 min, respectively. In the afternoon, mice were treated with the alternate treatment and confined to the opposite chamber. Mice were then tested for CPP on day 3. Mice then received additional conditioning on this day as well as on day 4. Mice were then tested for CPP on day 5. Mice then received extinction training on day 9. On day 10, mice were tested for extinction and then reinstatement of CPP following a priming dose of cocaine (7.5 mg/kg). Male and female mice lacking beta-endorphin did not exhibit CPP following single conditioning with cocaine. On the other hand, only male mice lacking beta-endorphin failed to show CPP after repeated conditioning. Nonetheless, reinstatement of CPP was blunted in both male and female mice lacking beta-endorphin compared to controls. The present results suggest that beta-endorphin plays a functional role in cocaine-induced CPP and its reinstatement, and sex-related differences exist in the regulatory action of beta-endorphin on the acquisition but not reinstatement of cocaine CPP.
Collapse
Affiliation(s)
- Prableen K Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| | - Kabirullah Lutfy
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| |
Collapse
|
9
|
Hersey M, Bacon AK, Bailey LG, Coggiano MA, Newman AH, Leggio L, Tanda G. Psychostimulant Use Disorder, an Unmet Therapeutic Goal: Can Modafinil Narrow the Gap? Front Neurosci 2021; 15:656475. [PMID: 34121988 PMCID: PMC8187604 DOI: 10.3389/fnins.2021.656475] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/20/2021] [Indexed: 12/11/2022] Open
Abstract
The number of individuals affected by psychostimulant use disorder (PSUD) has increased rapidly over the last few decades resulting in economic, emotional, and physical burdens on our society. Further compounding this issue is the current lack of clinically approved medications to treat this disorder. The dopamine transporter (DAT) is a common target of psychostimulant actions related to their use and dependence, and the recent availability of atypical DAT inhibitors as a potential therapeutic option has garnered popularity in this research field. Modafinil (MOD), which is approved for clinical use for the treatment of narcolepsy and sleep disorders, blocks DAT just like commonly abused psychostimulants. However, preclinical and clinical studies have shown that it lacks the addictive properties (in both behavioral and neurochemical studies) associated with other abused DAT inhibitors. Clinical availability of MOD has facilitated its off-label use for several psychiatric disorders related to alteration of brain dopamine (DA) systems, including PSUD. In this review, we highlight clinical and preclinical research on MOD and its R-enantiomer, R-MOD, as potential medications for PSUD. Given the complexity of PSUD, we have also reported the effects of MOD on psychostimulant-induced appearance of several symptoms that could intensify the severity of the disease (i.e., sleep disorders and impairment of cognitive functions), besides the potential therapeutic effects of MOD on PSUD.
Collapse
Affiliation(s)
- Melinda Hersey
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Amanda K. Bacon
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Lydia G. Bailey
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Mark A. Coggiano
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Amy H. Newman
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Lorenzo Leggio
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
- Clinical Psychoneuroendo- crinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
- National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research, National Institutes of Health, Bethesda, MD, United States
| | - Gianluigi Tanda
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
10
|
Domi E, Domi A, Adermark L, Heilig M, Augier E. Neurobiology of alcohol seeking behavior. J Neurochem 2021; 157:1585-1614. [PMID: 33704789 DOI: 10.1111/jnc.15343] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 03/02/2021] [Accepted: 03/02/2021] [Indexed: 12/29/2022]
Abstract
Alcohol addiction is a chronic relapsing brain disease characterized by an impaired ability to stop or control alcohol use despite adverse consequences. A main challenge of addiction treatment is to prevent relapse, which occurs in more than >50% of newly abstinent patients with alcohol disorder within 3 months. In people suffering from alcohol addiction, stressful events, drug-associated cues and contexts, or re-exposure to a small amount of alcohol trigger a chain of behaviors that frequently culminates in relapse. In this review, we first present the preclinical models that were developed for the study of alcohol seeking behavior, namely the reinstatement model of alcohol relapse and compulsive alcohol seeking under a chained schedule of reinforcement. We then provide an overview of the neurobiological findings obtained using these animal models, focusing on the role of opioids systems, corticotropin-release hormone and neurokinins, followed by dopaminergic, glutamatergic, and GABAergic neurotransmissions in alcohol seeking behavior.
Collapse
Affiliation(s)
- Esi Domi
- Center for Social and Affective Neuroscience, BKV, Linköping University, Linköping, Sweden
| | - Ana Domi
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Louise Adermark
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Markus Heilig
- Center for Social and Affective Neuroscience, BKV, Linköping University, Linköping, Sweden
| | - Eric Augier
- Center for Social and Affective Neuroscience, BKV, Linköping University, Linköping, Sweden
| |
Collapse
|
11
|
Hammad AM, Alasmari F, Sari Y. Effect of Modulation of the Astrocytic Glutamate Transporters' Expression on Cocaine-Induced Reinstatement in Male P Rats Exposed to Ethanol. Alcohol Alcohol 2021; 56:210-219. [PMID: 33063090 PMCID: PMC11004936 DOI: 10.1093/alcalc/agaa104] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/18/2020] [Accepted: 09/19/2020] [Indexed: 11/14/2022] Open
Abstract
AIM Reinforcing properties of ethanol and cocaine are mediated in part through the glutamatergic system. Extracellular glutamate concentration is strictly maintained through several glutamate transporters, such as glutamate transporter 1 (GLT-1), cystine/glutamate transporter (xCT) and glutamate aspartate transporter (GLAST). Previous findings revealed that cocaine and ethanol exposure downregulated GLT-1 and xCT, and that β-lactam antibiotics restored their expression. METHODS In this study, we investigated the effect of ampicillin/sulbactam (AMP/SUL) (200 mg/kg, i.p.), a β-lactam antibiotic, on cocaine-induced reinstatement and locomotor activity in male alcohol preferring (P) rats using free choice ethanol (15 and 30%, v/v) and water. We also investigated the effect of co-exposure to ethanol and cocaine (20 mg/kg, i.p.) on GLT-1, xCT and GLAST expression in the nucleus accumbens (NAc) core, NAc shell and dorsomedial prefrontal cortex (dmPFC). RESULTS Cocaine exposure decreased ethanol intake and preference. Cocaine and ethanol co-exposure acquired place preference and increased locomotor activity compared to ethanol-exposed rats. GLT-1 and xCT expression were downregulated after cocaine and ethanol co-exposure in the NAc core and shell, but not in dmPFC. AMP/SUL attenuated reinstatement to cocaine as well attenuated the decrease in locomotor activity and ethanol intake and preference. These effects were associated with upregulation of GLT-1 and xCT expression in the NAc core/shell and dmPFC. GLAST expression was not affected after ethanol and cocaine co-exposure or AMP/SUL treatment. CONCLUSION Our findings demonstrate that astrocytic glutamate transporters within the mesocorticolimbic area are critical targets in modulating cocaine-seeking behavior while being consuming ethanol.
Collapse
Affiliation(s)
- Alaa M Hammad
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | - Fawaz Alasmari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| |
Collapse
|
12
|
Abstract
A growing body of evidence from the past 15 years implicates epigenetic mechanisms in the behavioral effects of addictive drugs. The main focus of these studies has been epigenetic mechanisms of psychomotor sensitization and drug reinforcement, as assessed by the conditioned place preference and drug self-administration procedures. Some of these studies have documented long-lasting changes in the expression of epigenetic enzymes and molecules that persist for weeks after the last drug exposure. These observations have inspired more recent investigations on the epigenetic mechanisms of relapse to drug seeking after prolonged abstinence. Here, we review studies that have examined epigenetic mechanisms (e.g., histone modifications, chromatin remodeler-associated modifications, and DNA methylation) that contribute to relapse to cocaine, amphetamine, methamphetamine, morphine, heroin, nicotine, or alcohol seeking, as assessed in rodent models. We first provide a brief overview of studies that have examined persistent epigenetic changes in the brain after prolonged abstinence from noncontingent drug exposure or drug self-administration. Next, we review studies on the effect of either systemic or brain site-specific epigenetic manipulations on the reinstatement of drug-conditioned place preference after extinction of the learned preference, the reinstatement of drug seeking after operant drug self-administration and extinction of the drug-reinforced responding, and the incubation of drug craving (the time-dependent increase in drug seeking after cessation of drug self-administration). We conclude by discussing the implications of these studies for understanding mechanisms contributing to persistent relapse vulnerability after prolonged abstinence. We also discuss the implications of these results for translational research on the potential use of systemically administered epigenetic enzyme inhibitors for relapse prevention in human drug users.
Collapse
|
13
|
Reis HS, Rodrigues IRS, Anjos-Santos A, Libarino-Santos M, Serra YA, Cata-Preta EG, Oliveira-Campos D, Kisaki ND, Barros-Santos T, Yokoyama TS, Cruz FC, Oliveira-Lima AJ, Barbosa PCR, Berro LF, Marinho EAV. Ayahuasca blocks the reinstatement of methylphenidate-induced conditioned place preference in mice: behavioral and brain Fos expression evaluations. Psychopharmacology (Berl) 2020; 237:3269-3281. [PMID: 32676773 DOI: 10.1007/s00213-020-05609-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 07/09/2020] [Indexed: 10/23/2022]
Abstract
RATIONALE Accumulating evidence suggests that ayahuasca, a hallucinogenic beverage used in traditional Amazonian communities for ritualistic and curative purposes, has been associated with reduced rates of substance use disorders. However, the brain mechanisms underlying the therapeutic effects of ayahuasca have not yet been fully elucidated. OBJECTIVES The aim of the present study was to investigate the effects of treatment with ayahuasca on the rewarding properties of the psychostimulant methylphenidate. METHODS The rewarding properties of ayahuasca (100 mg/kg, orally) and methylphenidate (10 mg/kg, i.p.) were investigated using the conditioned place preference (CPP) model. Furthermore, we evaluated the effects of repeated treatment with ayahuasca on the reinstatement of methylphenidate-induced CPP. Fos expression was evaluated in different limbic structures (cingulate cortex-area 1, prelimbic cortex, infralimbic cortex, orbitofrontal cortex-lateral orbital area, nucleus accumbens core and shell, ventral tegmental area, dorsal striatum, and basolateral amygdala) upon each experimental phase. RESULTS Both ayahuasca and methylphenidate induced CPP in mice. However, ayahuasca had limited effects on Fos expression, while methylphenidate altered Fos expression in several brain regions associated with the behavioral effects of drugs of abuse. Treatment with ayahuasca after conditioning with methylphenidate blocked the reinstatement of methylphenidate-induced CPP. Those behavioral effects were accompanied by changes in Fos expression patterns, with ayahuasca generally blocking the changes in Fos expression induced by conditioning with methylphenidate and/or reexposure to methylphenidate. CONCLUSIONS Our findings suggest that ayahuasca restored normal brain function in areas associated with the long-term expression of drug wanting/seeking in animals conditioned to methylphenidate.
Collapse
Affiliation(s)
- Henrique S Reis
- Department of Health Sciences, Universidade Estadual de Santa Cruz, Rod. Ilhéus/Itabuna, Km 16, Ilhéus, BA, 45662-0, Brazil
| | - Isa R S Rodrigues
- Department of Health Sciences, Universidade Estadual de Santa Cruz, Rod. Ilhéus/Itabuna, Km 16, Ilhéus, BA, 45662-0, Brazil
| | - Alexia Anjos-Santos
- Department of Health Sciences, Universidade Estadual de Santa Cruz, Rod. Ilhéus/Itabuna, Km 16, Ilhéus, BA, 45662-0, Brazil
| | - Matheus Libarino-Santos
- Department of Health Sciences, Universidade Estadual de Santa Cruz, Rod. Ilhéus/Itabuna, Km 16, Ilhéus, BA, 45662-0, Brazil
| | - Yasmim A Serra
- Department of Health Sciences, Universidade Estadual de Santa Cruz, Rod. Ilhéus/Itabuna, Km 16, Ilhéus, BA, 45662-0, Brazil
| | - Elisângela G Cata-Preta
- Department of Health Sciences, Universidade Estadual de Santa Cruz, Rod. Ilhéus/Itabuna, Km 16, Ilhéus, BA, 45662-0, Brazil
| | - Daniella Oliveira-Campos
- Department of Health Sciences, Universidade Estadual de Santa Cruz, Rod. Ilhéus/Itabuna, Km 16, Ilhéus, BA, 45662-0, Brazil
| | - Natali D Kisaki
- Department of Health Sciences, Universidade Estadual de Santa Cruz, Rod. Ilhéus/Itabuna, Km 16, Ilhéus, BA, 45662-0, Brazil
| | - Thaísa Barros-Santos
- Department of Health Sciences, Universidade Estadual de Santa Cruz, Rod. Ilhéus/Itabuna, Km 16, Ilhéus, BA, 45662-0, Brazil
| | - Thais S Yokoyama
- Department of Pharmacology, Universidade Federal de São Paulo, Leal Prado Building, Botucatu 862 Street, 04024-002, Vila Clementino, São Paulo, SP, Brazil
| | - Fabio C Cruz
- Department of Pharmacology, Universidade Federal de São Paulo, Leal Prado Building, Botucatu 862 Street, 04024-002, Vila Clementino, São Paulo, SP, Brazil
| | - Alexandre J Oliveira-Lima
- Department of Health Sciences, Universidade Estadual de Santa Cruz, Rod. Ilhéus/Itabuna, Km 16, Ilhéus, BA, 45662-0, Brazil
| | - Paulo C R Barbosa
- Department of Health Sciences, Universidade Estadual de Santa Cruz, Rod. Ilhéus/Itabuna, Km 16, Ilhéus, BA, 45662-0, Brazil
| | - Lais F Berro
- Department of Health Sciences, Universidade Estadual de Santa Cruz, Rod. Ilhéus/Itabuna, Km 16, Ilhéus, BA, 45662-0, Brazil.
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, 2500 N State St, Jackson, MS, 39216, USA.
| | - Eduardo A V Marinho
- Department of Health Sciences, Universidade Estadual de Santa Cruz, Rod. Ilhéus/Itabuna, Km 16, Ilhéus, BA, 45662-0, Brazil.
| |
Collapse
|
14
|
Larrimore KE, Kannan L, Kendle RP, Jamal T, Barcus M, Stefanko K, Kilbourne J, Brimijoin S, Zhan CG, Neisewander J, Mor TS. A plant-derived cocaine hydrolase prevents cocaine overdose lethality and attenuates cocaine-induced drug seeking behavior. Prog Neuropsychopharmacol Biol Psychiatry 2020; 102:109961. [PMID: 32387315 PMCID: PMC7398606 DOI: 10.1016/j.pnpbp.2020.109961] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 05/01/2020] [Accepted: 05/02/2020] [Indexed: 12/21/2022]
Abstract
Cocaine use disorders include short-term and acute pathologies (e.g. overdose) and long-term and chronic disorders (e.g. intractable addiction and post-abstinence relapse). There is currently no available treatment that can effectively reduce morbidity and mortality associated with cocaine overdose or that can effectively prevent relapse in recovering addicts. One recently developed approach to treat these problems is the use of enzymes that rapidly break down the active cocaine molecule into inactive metabolites. In particular, rational design and site-directed mutagenesis transformed human serum recombinant butyrylcholinesterase (BChE) into a highly efficient cocaine hydrolase with drastically improved catalytic efficiency toward (-)-cocaine. A current drawback preventing the clinical application of this promising enzyme-based therapy is the lack of a cost-effective production strategy that is also flexible enough to rapidly scale-up in response to continuous improvements in enzyme design. Plant-based expression systems provide a unique solution as this platform is designed for fast scalability, low cost and the advantage of performing eukaryotic protein modifications such as glycosylation. A Plant-derived form of the Cocaine Super Hydrolase (A199S/F227A/S287G/A328W/Y332G) we designate PCocSH protects mice from cocaine overdose, counters the lethal effects of acute cocaine overdose, and prevents reinstatement of extinguished drug-seeking behavior in mice that underwent place conditioning with cocaine. These results demonstrate that the novel PCocSH enzyme may well serve as an effective therapeutic for cocaine use disorders in a clinical setting.
Collapse
Affiliation(s)
| | - Latha Kannan
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA; Center of Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85287-4501, USA.
| | - R Player Kendle
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA.
| | - Tameem Jamal
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA.
| | - Matthew Barcus
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA.
| | - Kathryn Stefanko
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA
| | - Jacquelyn Kilbourne
- Center of Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85287-4501, USA.
| | - Stephen Brimijoin
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA.
| | - Chang-Guo Zhan
- Molecular Modeling and Biopharmaceutical Center and Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA.
| | - Janet Neisewander
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA.
| | - Tsafrir S Mor
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA; Center of Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85287-4501, USA.
| |
Collapse
|
15
|
Cann C, Venniro M, Hope BT, Ramsey LA. Parametric investigation of social place preference in adolescent mice. Behav Neurosci 2020; 134:435-443. [PMID: 32672990 DOI: 10.1037/bne0000406] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Social interaction promotes survival by helping animals to form stable and supportive groups. Additionally, maladaptive social behavior is a hallmark of disorders such as autism and schizophrenia. In many different animal species, including humans, social interaction can be inherently rewarding. Lately there has been growing interest in studying the neurobiological underpinnings of social interaction and learned social behavior in rodent behavioral models. One common procedure is conditioned place preference (CPP) to measure the rewarding effects of social interaction and social reward learning. Social CPP was originally used in rats but has been adapted recently for use in mice, enabling use of the vast array of genetic tools available in mice. Here we studied the role of age, sex, bedding, and prior social isolation on the expression of social CPP in male and female mice. We found that without social deprivation male mice display moderate and temporary social CPP during early adolescence but not adulthood. Early life social isolation increased social CPP in female but not male mice. In contrast, cocaine CPP was robust and long-lasting in male and female mice. Our results demonstrate that social CPP in mice is variable, occurring under only specific conditions, and that social isolation promotes social reward in female but not male mice. We discuss potential methodological and interpretive issues of the mouse social CPP model. (PsycInfo Database Record (c) 2020 APA, all rights reserved).
Collapse
Affiliation(s)
- Courtney Cann
- Behavioral Neuroscience Research Branch, National Institute on Drug Abuse Intramural Research Program
| | - Marco Venniro
- Behavioral Neuroscience Research Branch, National Institute on Drug Abuse Intramural Research Program
| | - Bruce T Hope
- Behavioral Neuroscience Research Branch, National Institute on Drug Abuse Intramural Research Program
| | - Leslie A Ramsey
- Behavioral Neuroscience Research Branch, National Institute on Drug Abuse Intramural Research Program
| |
Collapse
|
16
|
Madison CA, Wellman PJ, Eitan S. Pre-exposure of adolescent mice to morphine results in stronger sensitization and reinstatement of conditioned place preference than pre-exposure to hydrocodone. J Psychopharmacol 2020; 34:771-777. [PMID: 32489137 DOI: 10.1177/0269881120926675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Opioids are commonly prescribed to treat moderate-to-severe pain. However, their use can trigger the development of opioid use disorder. A major problem in treating opioid use disorder remains the high rate of relapse. AIM The purpose of this study was to determine whether there are differences among opioids in their ability to trigger relapse after pre-exposure during adolescence. METHODS On postnatal day 33, mice were examined for the acute locomotor response to saline, morphine, or hydrocodone (5 mg/kg). They were administered with the corresponding opioid or saline during postnatal days 34-38 (20 mg/kg) and 40-44 (40 mg/kg). On postnatal day 45, they were recorded for the development of locomotor sensitization (5 mg/kg). Starting on postnatal day 55, mice were examined for the acquisition (1, 5, 10, 20, and 40 mg/kg), extinction, and drug-induced reinstatement (1, 2.5, and 5 mg/kg) of conditioned place preference. RESULTS There were no significant differences in the acute locomotor response to morphine and hydrocodone. Morphine induced significantly stronger locomotor sensitization as compared to hydrocodone. Pre-exposure to morphine, but not hydrocodone, sensitized the acquisition of conditioned place preference. There were no significant differences in extinction rates. Mice pre-exposed to morphine reinstate conditioned place preference after priming with a 1 mg/kg dose. In contrast, higher priming doses were required for reinstatement in all other experimental groups. CONCLUSIONS Adolescent mice administered with morphine develop greater sensitization to its effects and subsequently reinstate conditioned place preference more readily than mice administered with hydrocodone. This suggests higher risk for relapse after pre-exposure to morphine during adolescence as compared to hydrocodone.
Collapse
Affiliation(s)
- Caitlin A Madison
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, USA
| | - Paul J Wellman
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, USA
| | - Shoshana Eitan
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, USA
| |
Collapse
|
17
|
Shen X, Hui R, Luo Y, Yu H, Feng S, Xie B, Bi H, Galaj E, Cong B, Ma C, Wen D. Berberine Facilitates Extinction of Drug-Associated Behavior and Inhibits Reinstatement of Drug Seeking. Front Pharmacol 2020; 11:476. [PMID: 32390837 PMCID: PMC7194034 DOI: 10.3389/fphar.2020.00476] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 03/26/2020] [Indexed: 01/19/2023] Open
Abstract
A high rate of relapse is a major clinical problem among drug-addicted individuals. Persistent traces of drug-associated reward memories contribute to intense craving and often trigger relapse. A number of interventions on drug-associated memories have shown significant benefits in relapse prevention. Among them are pre- or post-extinction pharmacological manipulations that facilitate the extinction of drug-associated behavior. Berberine, a bioactive isoquinoline alkaloid, has been recently reported to provide therapeutic benefits for a number of central nervous system (CNS) disorders, including morphine addiction. The present study aimed to investigate whether berberine could serve as a post-extinction pharmacological intervention agent to reduce risks of reinstatement of drug seeking. We found that an intragastric administration of berberine at doses of 25 and 50 mg/kg during the critical time window significantly facilitated the extinction of morphine-reward related behavior in free access and confined conditioned place preference (CPP) extinction paradigms, and subsequently, it prevented reinstatement and spontaneous recovery of morphine-induced CPP in mice. Intriguingly, the berberine treatment with or without extinction training altered expression of plasticity-related proteins such as brain-derived neurotrophic factor (BDNF), AMPA receptors (GluA1 and GluA2) in the nucleus accumbens (NAc). Moreover, the post-extinction berberine treatment significantly reduced reinstatement of cocaine-induced CPP and operant intravenous self-administration (IVSA) memories in rats. Altogether, our findings suggest that extinction training combined with the post-extinction berberine treatment can facilitate extinction of drug-associated behavior making it an attractive therapeutic candidate in relapse prevention.
Collapse
Affiliation(s)
- Xi Shen
- College of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Rongji Hui
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang, China
| | - Yixiao Luo
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Hailei Yu
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang, China
| | - Suiyuan Feng
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang, China
| | - Bing Xie
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang, China
| | - Haitao Bi
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang, China
| | - Ewa Galaj
- Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Baltimore, MD, United States
| | - Bin Cong
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang, China
| | - Chunling Ma
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang, China
| | - Di Wen
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
18
|
Khaksari M, Nakhaei P, Khastar H, Bakhtazad A, Rahimi K, Garmabi B. Circadian fluctuation in curiosity is a risk factor for morphine preference. BIOL RHYTHM RES 2020. [DOI: 10.1080/09291016.2020.1719682] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Mehdi Khaksari
- Addiction Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Parham Nakhaei
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Hosein Khastar
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Atefeh Bakhtazad
- Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kasra Rahimi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Behzad Garmabi
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
- Study and Treatment of Circadian Rhythms Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
19
|
Sampedro-Piquero P, Ávila-Gámiz F, Moreno Fernández RD, Castilla-Ortega E, Santín LJ. The presence of a social stimulus reduces cocaine-seeking in a place preference conditioning paradigm. J Psychopharmacol 2019; 33:1501-1511. [PMID: 31542987 DOI: 10.1177/0269881119874414] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND One challenge in the treatment of substance use disorders is to re-engage the interest toward non-drug-related activities. Among these activities, social interaction has had a prominent role due to its positive influence on treatment outcome. AIMS AND METHODS Our aim was to study whether the presence of a social stimulus during the cocaine-induced conditioned place preference test was able to reduce the time spent in the drug-paired compartment. For that purpose, mice were trained for four days on a conditioned place preference task with one compartment paired with cocaine and the opposite with saline. On the test day, we introduced an unfamiliar juvenile male mouse into the saline-conditioned compartment (inside a pencil cup) to analyse the animal preference towards the two rewarding stimuli (cocaine vs mouse). Additionally, to discard the possible effect of novelty, as well as the housing condition (social isolation) on social preference, we decided to include a novel object during the test session, as well as perform the same conditioned place preference protocol with a group of animals in social housing conditions. RESULTS The social stimulus was able to reduce the preference for cocaine and enhance the active interaction with the juvenile mouse (sniffing) compared to the empty pencil cup paired with the drug. The introduction of a novel object during the test session did not reduce the preference for the cocaine-paired compartment, and interestingly, the preference for the social stimulus was independent of the housing condition. c-Fos immunohistochemistry revealed a different pattern of activation based on cocaine-paired conditioning or the presence of social stimulus. CONCLUSIONS These results suggest that social interaction could constitute a valuable component in the treatment of substance use disorders by reducing the salience of the drug.
Collapse
Affiliation(s)
- Patricia Sampedro-Piquero
- Instituto de Investigación Biomédica de Málaga (IBIMA), Malaga, Spain.,Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, Malaga, Spain
| | - Fabiola Ávila-Gámiz
- Instituto de Investigación Biomédica de Málaga (IBIMA), Malaga, Spain.,Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, Malaga, Spain
| | - Román D Moreno Fernández
- Instituto de Investigación Biomédica de Málaga (IBIMA), Malaga, Spain.,Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, Malaga, Spain
| | - Estela Castilla-Ortega
- Instituto de Investigación Biomédica de Málaga (IBIMA), Malaga, Spain.,Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Malaga, Spain
| | - Luis J Santín
- Instituto de Investigación Biomédica de Málaga (IBIMA), Malaga, Spain.,Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, Malaga, Spain
| |
Collapse
|
20
|
Kuhn BN, Kalivas PW, Bobadilla AC. Understanding Addiction Using Animal Models. Front Behav Neurosci 2019; 13:262. [PMID: 31849622 PMCID: PMC6895146 DOI: 10.3389/fnbeh.2019.00262] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/15/2019] [Indexed: 12/13/2022] Open
Abstract
Drug addiction is a neuropsychiatric disorder with grave personal consequences that has an extraordinary global economic impact. Despite decades of research, the options available to treat addiction are often ineffective because our rudimentary understanding of drug-induced pathology in brain circuits and synaptic physiology inhibits the rational design of successful therapies. This understanding will arise first from animal models of addiction where experimentation at the level of circuits and molecular biology is possible. We will review the most common preclinical models of addictive behavior and discuss the advantages and disadvantages of each. This includes non-contingent models in which animals are passively exposed to rewarding substances, as well as widely used contingent models such as drug self-administration and relapse. For the latter, we elaborate on the different ways of mimicking craving and relapse, which include using acute stress, drug administration or exposure to cues and contexts previously paired with drug self-administration. We further describe paradigms where drug-taking is challenged by alternative rewards, such as appetitive foods or social interaction. In an attempt to better model the individual vulnerability to drug abuse that characterizes human addiction, the field has also established preclinical paradigms in which drug-induced behaviors are ranked by various criteria of drug use in the presence of negative consequences. Separation of more vulnerable animals according to these criteria, along with other innate predispositions including goal- or sign-tracking, sensation-seeking behavior or impulsivity, has established individual genetic susceptibilities to developing drug addiction and relapse vulnerability. We further examine current models of behavioral addictions such as gambling, a disorder included in the DSM-5, and exercise, mentioned in the DSM-5 but not included yet due to insufficient peer-reviewed evidence. Finally, after reviewing the face validity of the aforementioned models, we consider the most common standardized tests used by pharmaceutical companies to assess the addictive potential of a drug during clinical trials.
Collapse
Affiliation(s)
| | - Peter W. Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Ana-Clara Bobadilla
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
21
|
Vulnerability to diet-induced obesity is associated with greater food priming-induced reinstatement of palatable food seeking. Physiol Behav 2019; 213:112730. [PMID: 31678197 DOI: 10.1016/j.physbeh.2019.112730] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/29/2019] [Accepted: 10/29/2019] [Indexed: 01/24/2023]
Abstract
We examined whether individual differences in weight gain during exposure to a "junk-food" diet were related to differences in later relapse-like behavior in a rat model. Following free access to a junk-food diet for 7 weeks, rats were trained to press a lever for palatable food pellets. Following extinction training, rats were tested for cue- and pellet priming-induced reinstatement. Results showed that rats prone to obesity while on the junk-food diet displayed greater pellet priming-, but not cue-, induced reinstatement relative to obesity-resistant rats, suggesting that obesity vulnerability is a factor determining one's chances for some types of relapse.
Collapse
|
22
|
Aldhafiri A, Dodu JC, Alalawi A, Emadzadeh N, Soderstrom K. Delta-9-THC exposure during zebra finch sensorimotor vocal learning increases cocaine reinforcement in adulthood. Pharmacol Biochem Behav 2019; 185:172764. [DOI: 10.1016/j.pbb.2019.172764] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/22/2019] [Accepted: 08/22/2019] [Indexed: 10/26/2022]
|
23
|
Niedzielska-Andres E, Mizera J, Sadakierska-Chudy A, Pomierny-Chamioło L, Filip M. Changes in the glutamate biomarker expression in rats vulnerable or resistant to the rewarding effects of cocaine and their reversal by ceftriaxone. Behav Brain Res 2019; 370:111945. [DOI: 10.1016/j.bbr.2019.111945] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/11/2019] [Accepted: 05/13/2019] [Indexed: 02/06/2023]
|
24
|
Der-Ghazarian TS, Charmchi D, Noudali SN, Scott SN, Holter MC, Newbern JM, Neisewander JL. Neural Circuits Associated with 5-HT 1B Receptor Agonist Inhibition of Methamphetamine Seeking in the Conditioned Place Preference Model. ACS Chem Neurosci 2019; 10:3271-3283. [PMID: 31042352 DOI: 10.1021/acschemneuro.8b00709] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
5-HT1B receptors (5-HT1BRs) modulate psychostimulant reward and incentive motivation in rodents. Here we investigated the effects of the 5-HT1BR agonist CP94253 (10 mg/kg, IP) on the acquisition and expression of methamphetamine (Meth) conditioned place preference (CPP) in C57BL/6 male mice. We subsequently examined the potential brain regions involved in CP94253 effects using FOS as a marker of neural activity. In the acquisition experiment, mice received the agonist 30 min before each of the Meth injections given during conditioning. In the expression experiment, mice that had acquired Meth-CPP were given either saline or CP94253 and were tested for CPP 30 min later. We found that CP94253 attenuated the expression of Meth-CPP, but had no effect on acquisition. Mice expressing Meth-CPP had elevated numbers of FOS+ cells in the ventral tegmental area (VTA) and basolateral amygdala (BlA) and reduced FOS+ cells in the central amygdala (CeA) compared to saline controls. CP94253 given before the expression test, but not acutely in drug-naive mice, enhanced FOS+ cells in the VTA, the nucleus accumbens (NAc) shell and core, and the dorsomedial striatum and reversed the Meth-conditioned changes in FOS in the BlA and CeA. Approximately 50-70% of FOS+ cells in the NAc and VTA were GABAergic regardless of group. By contrast, we did not observe FOS-labeling in dopamine neurons in the VTA. The findings suggest that CP94253 attenuates the motivational effects of the Meth-associated environment and highlight the amygdala, VTA, NAc, and dorsomedial striatum as potential regions involved in this effect.
Collapse
Affiliation(s)
| | - Delaram Charmchi
- School of Life Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Sean N. Noudali
- School of Life Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Samantha N. Scott
- School of Life Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Michael C. Holter
- School of Life Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Jason M. Newbern
- School of Life Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Janet L. Neisewander
- School of Life Sciences, Arizona State University, Tempe, Arizona 85287, United States
| |
Collapse
|
25
|
López AJ, Jia Y, White AO, Kwapis JL, Espinoza M, Hwang P, Campbell R, Alaghband Y, Chitnis O, Matheos DP, Lynch G, Wood MA. Medial habenula cholinergic signaling regulates cocaine-associated relapse-like behavior. Addict Biol 2019; 24:403-413. [PMID: 29430793 PMCID: PMC6087687 DOI: 10.1111/adb.12605] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 12/08/2017] [Accepted: 01/08/2018] [Indexed: 01/19/2023]
Abstract
Propensity to relapse, even following long periods of abstinence, is a key feature in substance use disorders. Relapse and relapse‐like behaviors are known to be induced, in part, by re‐exposure to drug‐associated cues. Yet, while many critical nodes in the neural circuitry contributing to relapse have been identified and studied, a full description of the networks driving reinstatement of drug‐seeking behaviors is lacking. One area that may provide further insight to the mechanisms of relapse is the habenula complex, an epithalamic region composed of lateral and medial (MHb) substructures, each with unique cell and target populations. Although well conserved across vertebrate species, the functions of the MHb are not well understood. Recent research has demonstrated that the MHb regulates nicotine aversion and withdrawal. However, it remains undetermined whether MHb function is limited to nicotine and aversive stimuli or if MHb circuit regulates responses to other drugs of abuse. Advances in circuit‐level manipulations now allow for cell‐type and temporally specific manipulations during behavior, specifically in spatially restrictive brain regions, such as the MHb. In this study, we focus on the response of the MHb to reinstatement of cocaine‐associated behavior, demonstrating that cocaine‐primed reinstatement of conditioned place preference engages habenula circuitry. Using chemogenetics, we demonstrate that MHb activity is sufficient to induce reinstatement behavior. Together, these data identify the MHb as a key hub in the circuitry underlying reinstatement and may serve as a target for regulating relapse‐like behaviors.
Collapse
Affiliation(s)
- Alberto J. López
- Department of Neurobiology and Behavior, Ayala School of Biological Sciences; University of California; Irvine CA USA
- UC Irvine Center for Addiction Neuroscience, Ayala School of Biological Sciences; University of California; Irvine CA USA
- Center for the Neurobiology of Learning and Memory, Ayala School of Biological Sciences; University of California; Irvine CA USA
| | - Yousheng Jia
- Department of Anatomy and Neurobiology, School of Medicine; University of California; Irvine CA USA
| | - André O. White
- Department of Biological Sciences, Neuroscience and Behavior; Mount Holyoke College; South Hadley MA USA
| | - Janine L. Kwapis
- Department of Neurobiology and Behavior, Ayala School of Biological Sciences; University of California; Irvine CA USA
- UC Irvine Center for Addiction Neuroscience, Ayala School of Biological Sciences; University of California; Irvine CA USA
| | - Monica Espinoza
- Department of Neurobiology and Behavior, Ayala School of Biological Sciences; University of California; Irvine CA USA
- UC Irvine Center for Addiction Neuroscience, Ayala School of Biological Sciences; University of California; Irvine CA USA
- Center for the Neurobiology of Learning and Memory, Ayala School of Biological Sciences; University of California; Irvine CA USA
| | - Philip Hwang
- Department of Neurobiology and Behavior, Ayala School of Biological Sciences; University of California; Irvine CA USA
- UC Irvine Center for Addiction Neuroscience, Ayala School of Biological Sciences; University of California; Irvine CA USA
- Center for the Neurobiology of Learning and Memory, Ayala School of Biological Sciences; University of California; Irvine CA USA
| | - Rianne Campbell
- Department of Neurobiology and Behavior, Ayala School of Biological Sciences; University of California; Irvine CA USA
- UC Irvine Center for Addiction Neuroscience, Ayala School of Biological Sciences; University of California; Irvine CA USA
- Center for the Neurobiology of Learning and Memory, Ayala School of Biological Sciences; University of California; Irvine CA USA
| | - Yasaman Alaghband
- Department of Neurobiology and Behavior, Ayala School of Biological Sciences; University of California; Irvine CA USA
- UC Irvine Center for Addiction Neuroscience, Ayala School of Biological Sciences; University of California; Irvine CA USA
- Center for the Neurobiology of Learning and Memory, Ayala School of Biological Sciences; University of California; Irvine CA USA
| | - Om Chitnis
- Department of Neurobiology and Behavior, Ayala School of Biological Sciences; University of California; Irvine CA USA
- UC Irvine Center for Addiction Neuroscience, Ayala School of Biological Sciences; University of California; Irvine CA USA
- Center for the Neurobiology of Learning and Memory, Ayala School of Biological Sciences; University of California; Irvine CA USA
| | - Dina P. Matheos
- Department of Neurobiology and Behavior, Ayala School of Biological Sciences; University of California; Irvine CA USA
- UC Irvine Center for Addiction Neuroscience, Ayala School of Biological Sciences; University of California; Irvine CA USA
- Center for the Neurobiology of Learning and Memory, Ayala School of Biological Sciences; University of California; Irvine CA USA
| | - Gary Lynch
- Department of Anatomy and Neurobiology, School of Medicine; University of California; Irvine CA USA
| | - Marcelo A. Wood
- Department of Neurobiology and Behavior, Ayala School of Biological Sciences; University of California; Irvine CA USA
- UC Irvine Center for Addiction Neuroscience, Ayala School of Biological Sciences; University of California; Irvine CA USA
- Center for the Neurobiology of Learning and Memory, Ayala School of Biological Sciences; University of California; Irvine CA USA
| |
Collapse
|
26
|
Marion‐Poll L, Besnard A, Longueville S, Valjent E, Engmann O, Caboche J, Hervé D, Girault J. Cocaine conditioned place preference: unexpected suppression of preference due to testing combined with strong conditioning. Addict Biol 2019; 24:364-375. [PMID: 29318708 DOI: 10.1111/adb.12600] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 11/09/2017] [Accepted: 12/19/2017] [Indexed: 12/30/2022]
Abstract
Conditioned place preference (CPP) is widely used for evaluating the rewarding effects of drugs. Like other memories, CPP is proposed to undergo reconsolidation during which it is unstable and sensitive to pharmacological inhibition. Previous studies have shown that cocaine CPP can be apparently erased by extracellular signal-regulated kinase (ERK) pathway inhibition during cocaine reconditioning (re-exposure to the drug-paired environment in the presence of the drug). Here, we show that blockade of D1 receptors during reconditioning prevented ERK activation and induced a loss of CPP. However, we also unexpectedly observed a CPP disappearance in mice that underwent testing and reconditioning with cocaine alone, specifically in strong conditioning conditions. The loss was due to the intermediate test. CPP was not recovered with reconditioning or priming in the short term, but it spontaneously reappeared after a month. When we challenged the D1 antagonist-mediated erasure, we observed that both a high dose of cocaine and a first CPP test were required for this effect. Our results also suggest a balance between D1-dependent ERK pathway activation and an A2a-dependent mechanism in D2 striatal neurons in controlling CPP expression. Our data reveal that, paradoxically, a simple CPP test can induce a complete (but transient) loss of place preference following strong but not weak cocaine conditioning. This study emphasizes the complex nature of CPP memory and the importance of multiple parameters that must be taken into consideration when investigating reconsolidation.
Collapse
Affiliation(s)
- Lucile Marion‐Poll
- Inserm UMR‐S839 France
- Sorbonne Université, Université Pierre et Marie Curie (UPMC) France
- Institut du Fer à Moulin France
| | - Antoine Besnard
- Sorbonne Université, Université Pierre et Marie Curie (UPMC) France
- Inserm UMR‐S 1130, Neurosciences Paris Seine France
- CNRS UMR 8246 France
| | - Sophie Longueville
- Inserm UMR‐S839 France
- Sorbonne Université, Université Pierre et Marie Curie (UPMC) France
- Institut du Fer à Moulin France
| | | | - Olivia Engmann
- Inserm UMR‐S839 France
- Sorbonne Université, Université Pierre et Marie Curie (UPMC) France
- Institut du Fer à Moulin France
| | - Jocelyne Caboche
- Sorbonne Université, Université Pierre et Marie Curie (UPMC) France
- Inserm UMR‐S 1130, Neurosciences Paris Seine France
- CNRS UMR 8246 France
| | - Denis Hervé
- Inserm UMR‐S839 France
- Sorbonne Université, Université Pierre et Marie Curie (UPMC) France
- Institut du Fer à Moulin France
| | - Jean‐Antoine Girault
- Inserm UMR‐S839 France
- Sorbonne Université, Université Pierre et Marie Curie (UPMC) France
- Institut du Fer à Moulin France
| |
Collapse
|
27
|
He Y, Lu Y, Shen Y, Wu F, Xu X, Kong E, Huang Z, Sun Y, Yu W. Transgenic increase in the β-endorphin concentration in cerebrospinal fluid alleviates morphine-primed relapse behavior through the μ opioid receptor in rats. J Med Virol 2019; 91:1158-1167. [PMID: 30701563 PMCID: PMC6593851 DOI: 10.1002/jmv.25415] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/06/2018] [Accepted: 11/28/2018] [Indexed: 11/11/2022]
Abstract
BACKGROUND Opioid-primed relapse is a global burden. Although current strategies have improved, optimal therapy is urgently needed. METHODS A recombinant adenovirus (Ad-NEP) expressing β-endorphin (β-EP) was designed and injected intracerebroventricularly (icv) into the right lateral ventricle in rats. Spatial and temporal β-EP expression in the lateral ventricle wall, subventricular zone and adjacent choroid plexus and the β-EP concentration in the cerebrospinal fluid (CSF) were observed during a 21-day period. A morphine priming-induced conditioned place preference (CPP) rat model was established. The β-EP-ir neuron counts, CSF β-EP concentration, and CPP score, which were used to evaluate morphine-primed reinstatement following extinction, were recorded 7 days after the icv injection. Additionally, the rats were pretreated with the irreversible μ opioid receptor antagonist β-funaltrexamine (β-FNA) and the selective κ opioid receptor antagonist nor-binaltorphimine (nor-BNI) to identify the receptor-dependent mechanism. RESULTS Both peak β-EP expression in target neurons and the peak CSF β-EP concentration occurred 7 to 8 days after Ad-NEP icv injection. The sustainable increase in the CSF β-EP concentration was correlated with a decrease in the CPP score 7 days after the Ad-NEP icv injection. Furthermore, reinstatement was almost reversed by β-FNA pretreatment 24 hours before the behavioral test, but nor-BNI had little effect. CONCLUSION The increasing cerebrospinal fluid β-endorphin concentrations showed that the therapeutic effect on opioid relapse occurred predominantly through a μ opioid receptor-dependent mechanism. The Ad-NEP adenovirus can be considered an alternative therapy for opioid relapse.
Collapse
Affiliation(s)
- Yan He
- Department of Anesthesiology & Intensive Care, Eastern Hepatobiliary Hospital, Second Military Medical University, Shanghai, China.,Department of Anesthesiology, Fuzhou General Hospital of PLA, Fuzhou, Fujian, China
| | - Yugang Lu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yang Shen
- Drug and Equipment Section, 442 Clinic Department of Fuzhous General Hospital of PLA, Ningde, Fujian, China
| | - Feixiang Wu
- Department of Anesthesiology & Intensive Care, Eastern Hepatobiliary Hospital, Second Military Medical University, Shanghai, China
| | - Xuewu Xu
- Department of Anesthesiology, 306 Hospital of PLA, Beijing, China
| | - Erliang Kong
- Department of Anesthesiology & Intensive Care, Eastern Hepatobiliary Hospital, Second Military Medical University, Shanghai, China
| | - Zhangxiang Huang
- Pain Clinic of First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yuming Sun
- Department of Anesthesiology & Intensive Care, Eastern Hepatobiliary Hospital, Second Military Medical University, Shanghai, China
| | - Weifeng Yu
- Department of Anesthesiology & Intensive Care, Eastern Hepatobiliary Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
28
|
Taslimi Z, Komaki A, Sarihi A, Haghparast A. Effect of acute and chronic restraint stress on electrical activity of prefrontal cortex neurons in the reinstatement of extinguished methamphetamine-induced conditioned place preference: An electrophysiological study. Brain Res Bull 2019; 146:237-243. [PMID: 30660715 DOI: 10.1016/j.brainresbull.2019.01.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 01/09/2019] [Accepted: 01/12/2019] [Indexed: 12/29/2022]
Abstract
Increased vulnerability to drug abuse has been observed after exposure to stress and the prefrontal cortex (PFC) plays a major role in the control of the stress response and reward pathway. The current study was conducted to clarify the effects of acute and chronic restraint stress on PFC neural activity during the reinstatement of methamphetamine (METH)-induced conditioned place preference (CPP) in rats. Following the establishment of CPP (METH 0.5 mg/kg; s.c. for 3 days) and the extinction phase, male Wistar rats were divided into threshold (0.25 mg/kg; s.c.) and sub-threshold (0.125 mg/kg; s.c.) METH-treated super groups to induce reinstatement. Each super group contained control (non-stressed), acute restraint stress (ARS) and chronic restraint stress (CRS) groups. in vivo single unit recordings were performed on the urethane-anesthetized rats in these groups. After baseline recordings (10-min period) of the neurons in the PFC, their firing activity was recorded for 50 min during the reinstatement phase after injection of METH. The results showed that the threshold dose, but not the sub-threshold dose, of METH significantly increased PFC neural activity in the non-stressed animals. The sub-threshold dose of METH notably changed this activity in both the ARS and CRS groups. These changes in the excited neurons after the sub-threshold dose in the ARS and CRS groups were significantly higher than those in the non-stressed group. It appears that the PFC is implicated in the associated reward pathway and stress functions. METH affected the firing rate of PFC neurons and stress amplified the effect of METH on changes in the neuronal firing rate in the PFC.
Collapse
Affiliation(s)
- Zahra Taslimi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Abdolrahman Sarihi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, 19615-1178, Tehran, Iran.
| |
Collapse
|
29
|
Althobaiti YS, Alshehri FS, Hakami AY, Hammad AM, Sari Y. Effects of Clavulanic Acid Treatment on Reinstatement to Methamphetamine, Glial Glutamate Transporters, and mGluR 2/3 Expression in P Rats Exposed to Ethanol. J Mol Neurosci 2018; 67:1-15. [PMID: 30471010 DOI: 10.1007/s12031-018-1194-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 10/01/2018] [Indexed: 11/24/2022]
Abstract
Evidence demonstrated that the glutamatergic system is implicated in mediating relapse to several drugs of abuse, including methamphetamine (METH). Glutamate homeostasis is maintained by a number of glutamate transporters, such as glutamate transporter type 1 (GLT-1), cystine/glutamate transporter (xCT), and glutamate aspartate transporter (GLAST). In addition, group II metabotropic glutamate receptors (mGluR2/3) were found to be implicated in relapse-seeking behavior. Ample evidence showed that β-lactam antibiotics are effective in upregulating GLT-1 and xCT expression, thus improving glutamate homeostasis and attenuating relapse to drugs of abuse. In this study, we investigated the reinstatement of METH using conditioned place preference (CPP) in male alcohol-preferring (P) rats exposed to home-cage free choice ethanol drinking. Here, we tested the effect of clavulanic acid (CA), a β-lactam, on the reinstatement of METH-seeking and ethanol drinking. In addition, we examined the expression of GLT-1, xCT, and GLAST as well as metabotropic glutamate receptor (mGluR2/3) in the nucleus accumbens (NAc) shell, NAc core, and dorsomedial prefrontal cortex (dmPFC). A priming i.p. injection of METH reinstated preference in METH-paired chamber following extinction. Chronic exposure to ethanol decreased the expression of GLT-1 and xCT in the NAc shell, but not in the NAc core or dmPFC. CA treatment blocked the reinstatement of METH-seeking, decreased ethanol intake, and restored the expression of GLT-1 and xCT in the NAc shell. In addition, the expression of mGluR2/3 was increased by CA treatment in the NAc shell and dmPFC. These findings suggest that these glutamate transporters and mGluR2/3 might be potential therapeutic targets for the attenuation of reinstatement to METH-seeking.
Collapse
Affiliation(s)
- Yusuf S Althobaiti
- College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology and Experimental Therapeutics, University of Toledo, Health Science Campus, 3000 Arlington Avenue, HEB 282G, Toledo, OH, 43614, USA.,College of Pharmacy, Department of Pharmacology and Toxicology, Taif University, Taif, Saudi Arabia
| | - Fahad S Alshehri
- College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology and Experimental Therapeutics, University of Toledo, Health Science Campus, 3000 Arlington Avenue, HEB 282G, Toledo, OH, 43614, USA
| | - Alqassem Y Hakami
- College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology and Experimental Therapeutics, University of Toledo, Health Science Campus, 3000 Arlington Avenue, HEB 282G, Toledo, OH, 43614, USA
| | - Alaa M Hammad
- College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology and Experimental Therapeutics, University of Toledo, Health Science Campus, 3000 Arlington Avenue, HEB 282G, Toledo, OH, 43614, USA
| | - Youssef Sari
- College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology and Experimental Therapeutics, University of Toledo, Health Science Campus, 3000 Arlington Avenue, HEB 282G, Toledo, OH, 43614, USA.
| |
Collapse
|
30
|
Akhiary M, Purvis EM, Klein AK, Ettenberg A. Methamphetamine self-administration in a runway model of drug-seeking behavior in male rats. Pharmacol Biochem Behav 2018; 175:27-32. [PMID: 30196087 DOI: 10.1016/j.pbb.2018.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 09/05/2018] [Accepted: 09/06/2018] [Indexed: 12/27/2022]
Abstract
Cocaine administration has been shown to produce immediate positive (rewarding) and subsequent negative (anxiogenic) effects in humans and animals. These dual and opposing affective responses have been more difficult to demonstrate with administration of methamphetamine (meth). While animal studies have reliably demonstrated the positive reinforcing effects of the drug, reports of negative aftereffects following acute exposure have been few in number and contradictory in nature. The current research was devised to assess the effects of acute meth using a runway model of self-administration that is uniquely sensitive to both the positive and negative effects of a drug reinforcer in the same animal on the same trial. Male rats were allowed to traverse a straight alley once a day for 16 consecutive days/trials where entry into the goal box resulted in a single IV injection of meth (0.25, 0.5 or 1.0 mg/kg/inj.). The chosen doses were confirmed to be psychoactive as they produced dose-dependent increases in motoric/locomotor activation in these same subjects. The results demonstrated a U-shaped dose-response curve for the reinforcing effects of meth in that the intermediate dose group (0.5 mg/kg) produced the strongest approach behavior in the runway. Unlike other psychomotor stimulants, like cocaine, animals running for IV meth exhibited no evidence of any significant approach-avoidance behaviors reflective of the drug's negative anxiogenic effects. These results suggest that the abuse potential for meth is likely higher than for other shorter-acting psychomotor stimulants and reaffirms the utility of the runway procedure as a screen for a substance's abuse potential.
Collapse
Affiliation(s)
- Mona Akhiary
- Behavioral Pharmacology Laboratory, Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA 93109-9660, USA
| | - Erin M Purvis
- Behavioral Pharmacology Laboratory, Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA 93109-9660, USA
| | - Adam K Klein
- Behavioral Pharmacology Laboratory, Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA 93109-9660, USA
| | - Aaron Ettenberg
- Behavioral Pharmacology Laboratory, Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA 93109-9660, USA.
| |
Collapse
|
31
|
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
|
32
|
Abstract
Interest for the use of oxytocin as a treatment for addiction began over 40years ago. Better known for its roles in parturition, lactation and pair bonding, oxytocin also has anxiolytic properties, reduces immune and inflammatory responses, and has a role in learning and memory. In this chapter, oxytocin effects on addiction processes are described by highlighting research findings that have used oxytocin within current preclinical animal models of addiction, relapse, or craving. First, we provide a brief background of the endogenous oxytocin system followed by descriptions of the behavioral models used to study addiction, including models of drug taking and seeking. Then we review recent preclinical studies that have used oxytocin as a therapeutic intervention throughout multiple stages of the addiction cycle from a behavioral and neurobiological perspective. These models encompass the entire range of the addiction cycle including acquisition and maintenance of drug taking, withdrawal and craving during periods of drug abstinence, and ultimately relapse. We then posit several theories about how oxytocin interacts with both drug and social reward, as well as presenting a mechanistic account of how specific oxytocin receptor localization may contribute to oxytocin's efficacy as an addiction therapeutic.
Collapse
|
33
|
Wang R, Shen YL, Hausknecht KA, Chang L, Haj-Dahmane S, Vezina P, Shen RY. Prenatal ethanol exposure increases risk of psychostimulant addiction. Behav Brain Res 2018; 356:51-61. [PMID: 30076855 DOI: 10.1016/j.bbr.2018.07.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/04/2018] [Accepted: 07/31/2018] [Indexed: 01/04/2023]
Abstract
Prenatal ethanol exposure (PE) causes many cognitive and behavioral deficits including increased drug addiction risk, demonstrated by enhanced ethanol intake and behavioral phenotypes associated with addiction risk. Additionally, preclinical studies show that PE persistently changes the function of dopaminergic neurons in the ventral tegmental area, a major neural substrate for addiction, and alters these neurons' responses to psychostimulants. Accordingly, PE could also lead to increased risk of addiction to drugs of abuse, other than ethanol. In the present study, addiction risk was examined utilizing paradigms of amphetamine conditioned place preference (CPP) and intravenous self-administration. Ethanol was administered to pregnant dams via intragastric gavage (6 g/kg, during gestational days 8-20). Behavioral tests were conducted in adult male offspring. Amphetamine at a low dose (0.3 mg/kg, i.p.) induced CPP in PE but not control rats, whereas at a higher dose (0.6 mg/kg, i.p.) both groups acquired CPP. There was no group difference in amphetamine-induced CPP reinstatement. Furthermore, PE rats self-administered more amphetamine at a low dose (0.02 mg/kg/infusion) than controls, while no group differences were observed at a higher dose (0.1 mg/kg/infusion). Rats with PE also exhibited greater reactivity to contextual drug cues after extended abstinence and amphetamine-induced reinstatement of drug seeking. These results support that PE persistently leads to increased psychostimulant addiction risk later in life, manifested in many elements of addictive behavior following limited psychostimulant exposure. The observations provide insights into prevention strategies for drug addiction in individuals with fetal alcohol spectrum disorders.
Collapse
Affiliation(s)
- Ruixiang Wang
- Research Institute on Addictions and Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 1021 Main Street, Buffalo, NY, 14203, USA; Department of Psychology, Park Hall Room 204, University at Buffalo, Buffalo, NY, 14260, USA
| | - Ying-Ling Shen
- Research Institute on Addictions and Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 1021 Main Street, Buffalo, NY, 14203, USA; Graduate Institute of Humanities in Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Kathryn A Hausknecht
- Research Institute on Addictions and Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 1021 Main Street, Buffalo, NY, 14203, USA
| | - Lawrence Chang
- Research Institute on Addictions and Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 1021 Main Street, Buffalo, NY, 14203, USA
| | - Samir Haj-Dahmane
- Research Institute on Addictions and Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 1021 Main Street, Buffalo, NY, 14203, USA
| | - Paul Vezina
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, 5841 South Maryland Avenue MC 3077, Chicago, IL, 60637, USA
| | - Roh-Yu Shen
- Research Institute on Addictions and Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 1021 Main Street, Buffalo, NY, 14203, USA.
| |
Collapse
|
34
|
Doncheck EM, Hafenbreidel M, Ruder SA, Fitzgerald MK, Torres L, Mueller D. bFGF expression is differentially regulated by cocaine seeking versus extinction in learning-related brain regions. Learn Mem 2018; 25:361-368. [PMID: 30012881 PMCID: PMC6049391 DOI: 10.1101/lm.047530.118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 06/11/2018] [Indexed: 11/24/2022]
Abstract
In cocaine use disorder, relapse can be elicited by drug-associated cues despite long periods of abstinence. The persistence of drug-associated cues in eliciting drug seeking suggests enduring changes in structural and functional plasticity, which may be mediated by basic fibroblast growth factor (bFGF, FGF2). Stimulant drug use increases bFGF expression in reward- and learning-related brain regions, such as the infralimbic medial-prefrontal cortex (IL-mPFC), and we previously found that this increase was reversed by extinction. However, whether bFGF expression is similarly modified in other brain regions is unknown. Therefore, we used the conditioned place preference (CPP) paradigm to assess bFGF expression following cocaine-associated CPP or extinction of that CPP within the mPFC, nucleus accumbens (NAc), hippocampus (Hipp), and basolateral amygdala (BLA). bFGF expression was increased in IL-mPFC and NAc-Core and -Shell following a cocaine-associated CPP, an effect reversed by extinction. Conversely, bFGF expression was increased in BLA following extinction, but no significant changes were observed in PL-mPFC or either dorsal or ventral Hipp. These results demonstrate differential regulation of bFGF following cocaine-associated CPP or extinction of that CPP in discrete brain regions. Changes in bFGF expression may regulate long-lasting drug-induced plasticity that underlies persistent drug-associated memories, and therefore present potential prophylactic targets.
Collapse
Affiliation(s)
- Elizabeth M Doncheck
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53201-0413, USA
| | - Madalyn Hafenbreidel
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53201-0413, USA
| | - Sarah A Ruder
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53201-0413, USA
| | - Michael K Fitzgerald
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53201-0413, USA
| | - Lilith Torres
- Department of Basic Sciences, Ponce Health Sciences University-School of Medicine/Ponce Research Institute, Ponce, Puerto Rico, 00732, USA
| | - Devin Mueller
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53201-0413, USA
- Department of Basic Sciences, Ponce Health Sciences University-School of Medicine/Ponce Research Institute, Ponce, Puerto Rico, 00732, USA
| |
Collapse
|
35
|
Noye Tuplin EW, Lightfoot SHM, Holahan MR. Comparison of the Time-Dependent Changes in Immediate Early Gene Labeling and Spine Density Following Abstinence From Contingent or Non-contingent Chocolate Pellet Delivery. Front Behav Neurosci 2018; 12:144. [PMID: 30061817 PMCID: PMC6055009 DOI: 10.3389/fnbeh.2018.00144] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 06/25/2018] [Indexed: 12/23/2022] Open
Abstract
Rationale: Incubation of craving is a phenomenon whereby responding for cues associated with a reward increases over extended periods of abstinence. Both contingent and non-contingent behavioral designs have been used to study the incubation of craving phenomenon with differing results. The present study directly compares behavioral and neural changes following contingent or non-contingent administration of chocolate flavored pellets. Objective: The current study examined whether an incubation of craving response would be observed at the behavioral and neural levels following delays of abstinence from chocolate pellets in a contingent or non-contingent reinforcement design. Methods: Rats were trained for 10 days to bar press for chocolate pellets (contingent) or received chocolate pellets in a non-contingent design (classical conditioning). Groups were then subjected to abstinence from the reward for 24 h, 7, 14 or 28 days at which point they were tested for responding for reward associated cues. Following the test, brains from all rats were processed and assessed for c-Fos and FosB labeling as well as dendritic spine density in the nucleus accumbens (NAc). Results: Behavioral measures during the test (lever presses, food hopper entries and locomotor activity) revealed similar behavioral outcomes across all delays indicating the lack of an incubation of craving response on both the contingent and non-contingent designs. Overall, labeling of c-Fos in the NAc was lower for the non-contingent group compared to the operant-trained and food restricted control. Compared to the operant-trained and non-trained control groups, a significantly reduced FosB labeling was noted in the NAc of the classically conditioned groups across all abstinence periods. Spine density in the NAc was elevated in both the classically and operant conditioned compared to the food-restricted, non-trained controls. Conclusions: Chocolate pellet reward did not result in incubation of craving but did produce behavioral learning that was associated with increased spine density. This suggests that chocolate pellet administration results in long-term structural and functional changes that are present for at least 28 days following abstinence. Contingent and non-contingent administration resulted in differential immediate early gene labeling in the NAc, but the functional significance of this has yet to be elucidated.
Collapse
|
36
|
Blanco-Gandía MC, Aguilar MA, Miñarro J, Rodríguez-Arias M. Reinstatement of Drug-seeking in Mice Using the Conditioned Place Preference Paradigm. J Vis Exp 2018:56983. [PMID: 29939175 PMCID: PMC6101638 DOI: 10.3791/56983] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The present protocol describes the Conditioned Place Preference (CPP) as a model of relapse in drug addiction. In this model, animals are first trained to acquire a conditioned place preference in a drug-paired compartment, and after the post-conditioning test, they perform several sessions to extinguish the established preference. The CPP permits the evaluation of the conditioned rewarding effects of drugs related to environmental cues. Then, the extinguished CPP can be robustly reinstated by the non-contingent administration of a priming dose of the drug, and by exposure to stressful stimuli. Both methods will be explained here. When the animal reinitiates the behavioral response, a reinstatement of the conditioned reward is considered to have taken place. The main advantages of this protocol are that it is non-invasive, inexpensive, and simple with good validity criteria. In addition, it allows the study of different environmental manipulations, such as stress or diet, which can modulate relapse into drug seeking behaviors. However, one limitation is that if the researcher aims to explore the motivation and primary reinforcing effects of the drug, it should be complemented with self-administration procedures, as they involve operant responses of animals.
Collapse
Affiliation(s)
- M Carmen Blanco-Gandía
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de València
| | - María A Aguilar
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de València
| | - José Miñarro
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de València
| | - Marta Rodríguez-Arias
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de València;
| |
Collapse
|
37
|
Ball KT, Stone E, Best O, Collins T, Edson H, Hagan E, Nardini S, Neuciler P, Smolinsky M, Tosh L, Woodlen K. Chronic restraint stress during withdrawal increases vulnerability to drug priming-induced cocaine seeking via a dopamine D1-like receptor-mediated mechanism. Drug Alcohol Depend 2018; 187:327-334. [PMID: 29705546 PMCID: PMC5959797 DOI: 10.1016/j.drugalcdep.2018.03.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 02/22/2018] [Accepted: 03/15/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND A major obstacle in the treatment of individuals with cocaine addiction is their high propensity for relapse. Although the clinical scenario of acute stress-induced relapse has been well studied in animal models, few pre-clinical studies have investigated the role of chronic stress in relapse or the interaction between chronic stress and other relapse triggers. METHODS We tested the effect of chronic restraint stress on cocaine seeking in rats using both extinction- and abstinence-based animal relapse models. Rats were trained to press a lever for I.V. cocaine infusions (0.50 mg/kg/infusion) paired with a discrete tone + light cue in daily 3-h sessions. Following self-administration, rats were exposed to a chronic restraint stress procedure (3 h/day) or control procedure (unstressed) during the first seven days of a 13-day extinction period during which lever presses had no programmed consequences. This was followed by cue- and cocaine priming-induced drug seeking tests. In a separate group of rats, cocaine seeking was assessed during forced abstinence both before and after the same chronic stress procedure. RESULTS A history of chronic restraint stress was associated with increased cocaine priming-induced drug seeking, an effect attenuated by co-administration of SCH-23390 (10.0 μg/kg; i.p.), a dopamine D1-like receptor antagonist, with daily restraint. Repeated SCH-23390 administration but not stress during extinction increased cue-induced reinstatement. CONCLUSIONS Exposure to chronic stress during early withdrawal may confer lasting vulnerability to some types of relapse, and dopamine D1-like receptors appear to mediate both chronic stress effects on cocaine seeking and extinction of cocaine seeking.
Collapse
Affiliation(s)
- Kevin T. Ball
- Department of Psychology, Bloomsburg University of Pennsylvania, 400 E. 2nd St., Bloomsburg, PA, 17815, USA
| | - Eric Stone
- Department of Psychology, Bloomsburg University of Pennsylvania, 400 E. 2nd St., Bloomsburg, PA, 17815, USA
| | - Olivia Best
- Department of Psychology, Bloomsburg University of Pennsylvania, 400 E. 2nd St., Bloomsburg, PA, 17815, USA
| | - Tyler Collins
- Department of Psychology, Bloomsburg University of Pennsylvania, 400 E. 2nd St., Bloomsburg, PA, 17815, USA
| | - Hunter Edson
- Department of Psychology, Bloomsburg University of Pennsylvania, 400 E. 2nd St., Bloomsburg, PA, 17815, USA
| | - Erin Hagan
- Department of Psychology, Bloomsburg University of Pennsylvania, 400 E. 2nd St., Bloomsburg, PA, 17815, USA
| | - Salvatore Nardini
- Department of Psychology, Bloomsburg University of Pennsylvania, 400 E. 2nd St., Bloomsburg, PA, 17815, USA
| | - Phelan Neuciler
- Department of Psychology, Bloomsburg University of Pennsylvania, 400 E. 2nd St., Bloomsburg, PA, 17815, USA
| | - Michael Smolinsky
- Department of Psychology, Bloomsburg University of Pennsylvania, 400 E. 2nd St., Bloomsburg, PA, 17815, USA
| | - Lindsay Tosh
- Department of Psychology, Bloomsburg University of Pennsylvania, 400 E. 2nd St., Bloomsburg, PA, 17815, USA
| | - Kristin Woodlen
- Department of Psychology, Bloomsburg University of Pennsylvania, 400 E. 2nd St., Bloomsburg, PA, 17815, USA
| |
Collapse
|
38
|
Farahimanesh S, Karimi S, Haghparast A. Role of orexin-1 receptors in the dorsal hippocampus (CA1 region) in expression and extinction of the morphine-induced conditioned place preference in the rats. Peptides 2018; 101:25-31. [PMID: 29269074 DOI: 10.1016/j.peptides.2017.12.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 11/16/2017] [Accepted: 12/18/2017] [Indexed: 11/23/2022]
Abstract
Orexinergic system is involved in reward processing and drug addiction. Objectives here, we investigated the effect of intra-hippocampal CA1 administration of orexin-1 receptor (OX1r) antagonist on the expression, and extinction of morphine-induced place preference in rats. Conditioned place preference (CPP) was induced by subcutaneous injection of morphine (5 mg/kg) during a 3-day conditioning phase. Two experimental plots were designed; SB334867 as a selective OX1r antagonist was dissolved in 12% DMSO, prepared in solutions with different concentrations (3, 30, and 300 nM), and microinjected into the CA1 and some neighboring regions (0.5 μl/side), bilaterally. CPP score and locomotor activity were recorded during the CPP test. Results demonstrated that intra-CA1 administration of the OX1r antagonist attenuates the expression of morphine-induced CPP. Furthermore, higher concentrations of SB334867 facilitated the extinction period of morphine-induced CPP and reduced its latency. Nevertheless, solely administration of DMSO did not have any influence on the CPP scores and locomotion in both phases. Our findings suggest that OX1rs in the CA1 region of the hippocampus are involved in the expression of morphine CPP. Moreover, blockade of OX1rs could facilitate extinction and may extinguish the ability of drug-related cues. It seems that the antagonist might be considered as a propitious therapeutic agent in suppressing drug-seeking behaviors.
Collapse
Affiliation(s)
- Sharareh Farahimanesh
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Institute for Cognitive Science Studies (ICSS), Tehran, Iran
| | - Sara Karimi
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
39
|
Regional Differences in Striatal Neuronal Ensemble Excitability Following Cocaine and Extinction Memory Retrieval in Fos-GFP Mice. Neuropsychopharmacology 2018; 43:718-727. [PMID: 28540927 PMCID: PMC5809776 DOI: 10.1038/npp.2017.101] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/03/2017] [Accepted: 05/18/2017] [Indexed: 11/08/2022]
Abstract
Learned associations between drugs of abuse and the drug administration environment have an important role in addiction. In rodents, exposure to a drug-associated environment elicits conditioned psychomotor activation, which may be weakened following extinction (EXT) learning. Although widespread drug-induced changes in neuronal excitability have been observed, little is known about specific changes within neuronal ensembles activated during the recall of drug-environment associations. Using a cocaine-conditioned locomotion (CL) procedure, the present study assessed the excitability of neuronal ensembles in the nucleus accumbens core and shell (NAccore and NAcshell), and dorsal striatum (DS) following cocaine conditioning and EXT in Fos-GFP mice that express green fluorescent protein (GFP) in activated neurons (GFP+). During conditioning, mice received repeated cocaine injections (20 mg/kg) paired with a locomotor activity chamber (Paired) or home cage (Unpaired). Seven to 13 days later, both groups were re-exposed to the activity chamber under drug-free conditions and Paired, but not Unpaired, mice exhibited CL. In a separate group of mice, CL was extinguished by repeatedly exposing mice to the activity chamber under drug-free conditions. Following the expression and EXT of CL, GFP+ neurons in the NAccore (but not NAcshell and DS) displayed greater firing capacity compared to surrounding GFP- neurons. This difference in excitability was due to a generalized decrease in GFP- excitability following CL and a selective increase in GFP+ excitability following its EXT. These results suggest a role for both widespread and ensemble-specific changes in neuronal excitability following recall of drug-environment associations.
Collapse
|
40
|
Steiner N, Rossetti C, Sakurai T, Yanagisawa M, de Lecea L, Magistretti PJ, Halfon O, Boutrel B. Hypocretin/orexin deficiency decreases cocaine abuse liability. Neuropharmacology 2018; 133:395-403. [PMID: 29454841 DOI: 10.1016/j.neuropharm.2018.02.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 01/12/2018] [Accepted: 02/12/2018] [Indexed: 12/13/2022]
Abstract
Compelling evidence indicates that hypocretin/orexin signaling regulates arousal, stress and reward-seeking behaviors. However, most studies on drug reward-related processes have so far described the effects of pharmacological blockers disrupting hypocretin/orexin transmission. We report here an extensive study on cocaine-related behaviors in hypocretin/orexin-deficient mice (KO) and their heterozygous (HET) and wildtype (WT) littermates. We evaluated behavioral sensitization following repeated administrations and preference for an environment repeatedly paired with cocaine injections (15 mg/kg). Mice were also trained to self-administer cocaine (0.5-1.5 mg/kg/infusion). Our observations show that whereas all mice exhibited quite similar responses to acute administration of cocaine, only Hcrt KO mice exhibited reduced cocaine-seeking behaviors following a period of abstinence or extinction, and reduced cocaine incubation craving. Further, if the present findings confirm that Hcrt deficient mice may display a hypoactive phenotype, possibly linked to a reduced alertness concomitant to a decreased exploration of their environment, hypocretin/orexin defiency did not cause any attentional deficit. We thus report that innate disruption of hypocretin/orexin signaling moderately alters cocaine reward but significantly reduces long-term affective dependence that may explain the lack of relapse for cocaine seeking seen in Hcrt KO mice. Overall, with blunted cocaine intake at the highest concentration and reduced responsiveness to cocaine cues after prolonged abstinence, our findings suggest that hypocretin deficient mice may display signs of resilience to cocaine addiction.
Collapse
Affiliation(s)
- Nadia Steiner
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Switzerland
| | - Clara Rossetti
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Switzerland
| | - Takeshi Sakurai
- Department of Molecular Neuroscience and Integrative Physiology, Faculty of Medicine, Kanazawa University, Japan; International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Japan
| | - Luis de Lecea
- Dept. of Psychiatry and Behavioral Sciences, Stanford University, USA
| | - Pierre J Magistretti
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Switzerland
| | - Olivier Halfon
- Division of Adolescent and Child Psychiatry, Department of Psychiatry, Lausanne University Hospital, Switzerland
| | - Benjamin Boutrel
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Switzerland; Division of Adolescent and Child Psychiatry, Department of Psychiatry, Lausanne University Hospital, Switzerland.
| |
Collapse
|
41
|
Pena-Bravo JI, Reichel CM, Lavin A. Abstinence from Cocaine-Induced Conditioned Place Preference Produces Discrete Changes in Glutamatergic Synapses onto Deep Layer 5/6 Neurons from Prelimbic and Infralimbic Cortices. eNeuro 2017; 4:ENEURO.0308-17.2017. [PMID: 29242822 PMCID: PMC5729037 DOI: 10.1523/eneuro.0308-17.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/10/2017] [Accepted: 11/22/2017] [Indexed: 01/28/2023] Open
Abstract
Glutamatergic signaling in the medial prefrontal cortex (mPFC) plays a critical role in drug addiction and relapse. The mPFC is functionally subdivided into dorsal (prelimbic, PL) and ventral (infralimbic, IL) regions, and evidence suggests a differential role of these two divisions in the control of drug seeking and taking; however, there is a dearth of information on the cocaine-induced adaptations in PL- and IL-mPFC synaptic glutamate transmission and their regulation of behavioral responses to cocaine-associated stimuli. We tested male rats in a cocaine-induced conditioned place preference (CPP) paradigm. In vitro whole-cell recordings were performed at different abstinence intervals to investigate the neuroadaptations in synaptic glutamate transmission in PL- and IL-mPFC deep layer (5/6) pyramidal neurons. Our results show that in naïve animals, PL-mPFC neurons expressed higher frequency of spontaneous events (sEPSCs) than IL-mPFC neurons. Following cocaine-CPP and a short abstinence (SA) period (8 d), we observed decreases in the amplitude of sEPSCs in both mPFC regions. Longer abstinence periods (30 d), resulted in a sustained decrease in the frequency of sEPSCs and an increase in AMPA receptor rectification only in PL-mPFC neurons. In addition, PL-mPFC neurons expressed a decrease in the area under the curve of sEPSCs, suggesting altered receptor activation dynamics. Synaptic glutamate transmission was not significantly different between retested and naïve rats. These results suggest that retention of cocaine-CPP requires differential modulation of glutamate transmission between PL- and IL-mPFC neurons and that these adaptations are dependent on the abstinence interval and reexposure to the cocaine context.
Collapse
Affiliation(s)
- José I Pena-Bravo
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - Carmela M Reichel
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - Antonieta Lavin
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| |
Collapse
|
42
|
Otis JM, Mueller D. Reversal of Cocaine-Associated Synaptic Plasticity in Medial Prefrontal Cortex Parallels Elimination of Memory Retrieval. Neuropsychopharmacology 2017; 42:2000-2010. [PMID: 28466871 PMCID: PMC5561348 DOI: 10.1038/npp.2017.90] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/26/2017] [Accepted: 04/27/2017] [Indexed: 01/24/2023]
Abstract
Addiction is characterized by abnormalities in prefrontal cortex that are thought to allow drug-associated cues to drive compulsive drug seeking and taking. Identification and reversal of these pathologic neuroadaptations are therefore critical for treatment of addiction. Previous studies using rodents reveal that drugs of abuse cause dendritic spine plasticity in prelimbic medial prefrontal cortex (PL-mPFC) pyramidal neurons, a phenomenon that correlates with the strength of drug-associated memories in vivo. Thus, we hypothesized that cocaine-evoked plasticity in PL-mPFC may underlie cocaine-associated memory retrieval, and therefore disruption of this plasticity would prevent retrieval. Indeed, using patch clamp electrophysiology we find that cocaine place conditioning increases excitatory presynaptic and postsynaptic transmission in rat PL-mPFC pyramidal neurons. This was accounted for by increases in excitatory presynaptic release, paired-pulse facilitation, and increased AMPA receptor transmission. Noradrenergic signaling is known to maintain glutamatergic plasticity upon reactivation of modified circuits, and we therefore next determined whether inhibition of noradrenergic signaling during memory reactivation would reverse the cocaine-evoked plasticity and/or disrupt the cocaine-associated memory. We find that administration of the β-adrenergic receptor antagonist propranolol before memory retrieval, but not after (during memory reconsolidation), reverses the cocaine-evoked presynaptic and postsynaptic modifications in PL-mPFC and causes long-lasting memory impairments. Taken together, these data reveal that cocaine-evoked synaptic plasticity in PL-mPFC is reversible in vivo, and suggest a novel strategy that would allow normalization of prefrontal circuitry in addiction.
Collapse
Affiliation(s)
- James M Otis
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA,Department of Psychiatry, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Devin Mueller
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA,Department of Basic Sciences, Neuroscience Division, Ponce Health Sciences University-School of Medicine, Ponce Research Institute, Ponce, Puerto Rico,Department of Basic Sciences, Neuroscience Division, Ponce Health Sciences University-School of Medicine, Ponce Research Institute, P.O. Box 7004, Ponce 00732-7004, Puerto Rico, Tel: +1 787 840 2575 Ext. 2588, Fax: +1 787 844 1980, E-mail:
| |
Collapse
|
43
|
Hammad AM, Alasmari F, Althobaiti YS, Sari Y. Modulatory effects of Ampicillin/Sulbactam on glial glutamate transporters and metabotropic glutamate receptor 1 as well as reinstatement to cocaine-seeking behavior. Behav Brain Res 2017. [PMID: 28624317 DOI: 10.1016/j.bbr.2017.06.017] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Glutamatergic system has an important role in cocaine-seeking behavior. Studies have reported that chronic exposure to cocaine induces downregulation of glutamate transporter-1 (GLT-1) and cystine/glutamate exchanger (xCT) in the central reward brain regions. Ceftriaxone, a β-lactam antibiotic, restored GLT-1 expression and consequently reduced cue-induced reinstatement of cocaine-seeking behavior. In this study, we investigated the reinstatement to cocaine (20mg/kg, i.p.) seeking behavior using a conditioned place preference (CPP) paradigm in male alcohol-preferring (P) rats. In addition, we investigated the effects of Ampicillin/Sulbactam (AMP/SUL) (200mg/kg, i.p.), a β-lactam antibiotic, on cocaine-induced reinstatement. We also investigated the effects of AMP/SUL on the expression of glial glutamate transporters and metabotropic glutamate receptor 1 (mGluR1) in the nucleus accumbens (NAc) core and shell and the dorsomedial prefrontal cortex (dmPFC). We found that AMP/SUL treatment reduced cocaine-triggered reinstatement. This effect was associated with a decrease in locomotor activity. Moreover, GLT-1 and xCT were downregulated in the NAc core and shell, but not in the dmPFC, following cocaine-primed reinstatement. However, cocaine exposure increased the expression of mGluR1 in the NAc core, but not in the NAc shell or dmPFC. Importantly, AMP/SUL treatment normalized GLT-1 and xCT expression in the NAc core and shell; however, the drug normalized mGluR1 expression in the NAc core only. Additionally, AMP/SUL increased the expression of GLT-1 and xCT in the dmPFC as compared to the water naïve group. These findings demonstrated that glial glutamate transporters and mGluR1 in the mesocorticolimbic area could be potential therapeutic targets for the attenuation of reinstatement to cocaine-seeking behavior.
Collapse
Affiliation(s)
- Alaa M Hammad
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Fawaz Alasmari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Yusuf S Althobaiti
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA.
| |
Collapse
|
44
|
Relapse to cocaine seeking in an invertebrate. Pharmacol Biochem Behav 2017; 157:41-46. [PMID: 28455125 DOI: 10.1016/j.pbb.2017.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 04/07/2017] [Accepted: 04/21/2017] [Indexed: 12/21/2022]
Abstract
Addiction is characterised by cycles of compulsive drug taking, periods of abstinence and episodes of relapse. The extinction/reinstatement paradigm has been extensively used in rodents to model human relapse and explore underlying mechanisms and therapeutics. However, relapse to drug seeking behaviour has not been previously demonstrated in invertebrates. Here, we used a cocaine conditioned place preference (CPP) paradigm in the flatworm, planarian, followed by extinction and reinstatement of drug seeking. Once baseline preference was established for one of two distinctly textured environments (i.e. compartments with a coarse or smooth surface), planarian received pairings of cocaine (5μM) in the non-preferred, and vehicle in the most preferred, environment, and were tested for conditioning thereafter. Cocaine produced robust CPP, measured as a significant increase in the time spent in the cocaine-paired compartment. Subsequently, planarian underwent extinction training, reverting back to their original preference within three sessions. Brief exposure to cocaine (5μM) or methamphetamine (5μM) reinstated cocaine-seeking behaviour. By contrast, the high affinity dopamine transporter inhibitor, (N-(n-butyl)-3α-[bis (4-fluorophenyl) methoxy]-tropane) (JHW007), which in rodents exhibits a neurochemical and behavioural profile distinct from cocaine, was ineffective. The present findings demonstrate for the first time reinstatement of extinguished cocaine seeking in an invertebrate model and suggest that the long-term adaptations underlying drug conditioning and relapse are highly conserved through evolution.
Collapse
|
45
|
Nesbit MO, Dias C, Phillips AG. The effects of d -govadine on conditioned place preference with d -amphetamine or food reward. Behav Brain Res 2017; 321:223-231. [DOI: 10.1016/j.bbr.2016.12.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 12/29/2016] [Accepted: 12/31/2016] [Indexed: 10/20/2022]
|
46
|
Counterconditioning During Reconsolidation Prevents Relapse of Cocaine Memories. Neuropsychopharmacology 2017; 42:716-726. [PMID: 27468918 PMCID: PMC5240172 DOI: 10.1038/npp.2016.140] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 07/19/2016] [Accepted: 07/25/2016] [Indexed: 11/08/2022]
Abstract
Relapse to drug abuse is often caused by exposure to drug-associated cues that evoke craving. Therefore, disruption of the cue-drug memory can prevent relapse. Memories destabilize and become temporarily labile upon their retrieval, and re-stabilize in a process termed reconsolidation. Pharmacological disruption of reconsolidation prevents relapse in animal models, yet may evoke side effects. Therefore, behavioral procedures capable of preventing cue-induced craving and relapse are extremely valuable. Aversion therapies, in which drug-paired cues are re-associated (counterconditioned) with aversive consequences, have limited success, because the previous cue-drug memory may recover, triggering relapse. Here, we prevented the memory recovery and relapse to cocaine seeking by applying aversive counterconditioning during memory reconsolidation. Mice were trained to seek cocaine in a conditioned place preference procedure. The cocaine-associated compartment was then counterconditioned with lithium chloride (LiCl)-induced malaise, preceded by a brief exposure to the compartment (memory retrieval). Relapse was assessed in a reinstatement test. We found that aversive counterconditioning conducted shortly after memory retrieval (during reconsolidation) induced a long-lasting prevention of relapse to cocaine seeking. However, mice relapsed when counterconditioned without, before, or long after memory retrieval, or when receiving LiCl without place counterconditioning. Our findings suggest that post-retrieval aversive counterconditioning leads to relapse prevention, possibly by replacing the cue-drug with a cue-aversion memory, thereby the cue ceases to evoke craving. Moreover, we found that a similar memory replacement procedure prevented relapse of conditioned place aversion. Hence, this novel procedure can also prevent relapse of aversive memories, providing a safe approach to alter various maladaptive behaviors.
Collapse
|
47
|
Golden SA, Aleyasin H, Heins R, Flanigan M, Heshmati M, Takahashi A, Russo SJ, Shaham Y. Persistent conditioned place preference to aggression experience in adult male sexually-experienced CD-1 mice. GENES, BRAIN, AND BEHAVIOR 2017; 16:44-55. [PMID: 27457669 PMCID: PMC5243174 DOI: 10.1111/gbb.12310] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 07/12/2016] [Accepted: 07/22/2016] [Indexed: 01/26/2023]
Abstract
We recently developed a conditioned place preference (CPP) procedure, commonly used to study rewarding drug effects, to demonstrate that dominant sexually-experienced CD-1 male mice form CPP to contexts previously associated with defeating subordinate male C57BL/6J mice. Here we further characterized conditioned and unconditioned aggression behavior in CD-1 mice. In Exp. 1 we used CD-1 mice that displayed a variable spectrum of unconditioned aggressive behavior toward younger subordinate C57BL/6J intruder mice. We then trained the CD-1 mice in the CPP procedure where one context was intruder-paired, while a different context was not. We then tested for aggression CPP 1 day after training. In Exp. 2, we tested CD-1 mice for aggression CPP 1 day and 18 days after training. In Exp. 3-4, we trained the CD-1 mice to lever-press for palatable food and tested them for footshock punishment-induced suppression of food-reinforced responding. In Exp. 5, we characterized unconditioned aggression in hybrid CD-1 × C57BL/6J D1-Cre or D2-Cre F1 generation crosses. Persistent aggression CPP was observed in CD-1 mice that either immediately attacked C57BL/6J mice during all screening sessions or mice that gradually developed aggressive behavior during the screening phase. In contrast, CD-1 mice that did not attack the C57BL/6J mice during screening did not develop CPP to contexts previously paired with C57BL/6J mice. The aggressive phenotype did not predict resistance to punishment-induced suppression of food-reinforced responding. CD-1 × D1-Cre or D2-Cre F1 transgenic mice showed strong unconditioned aggression. Our study demonstrates that aggression experience causes persistent CPP and introduces transgenic mice for circuit studies of aggression.
Collapse
Affiliation(s)
- Sam A. Golden
- Behavioral Neuroscience Branch, Intramural Research Program, NIDA, NIH, Baltimore MD
| | - Hossein Aleyasin
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY
| | - Robert Heins
- Behavioral Neuroscience Branch, Intramural Research Program, NIDA, NIH, Baltimore MD
| | - Meghan Flanigan
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY
| | - Mitra Heshmati
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY
| | - Aki Takahashi
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba, Japan
| | - Scott J. Russo
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY
| | - Yavin Shaham
- Behavioral Neuroscience Branch, Intramural Research Program, NIDA, NIH, Baltimore MD
| |
Collapse
|
48
|
Delis F, Polissidis A, Poulia N, Justinova Z, Nomikos GG, Goldberg SR, Antoniou K. Attenuation of Cocaine-Induced Conditioned Place Preference and Motor Activity via Cannabinoid CB2 Receptor Agonism and CB1 Receptor Antagonism in Rats. Int J Neuropsychopharmacol 2016; 20:269-278. [PMID: 27994006 PMCID: PMC5408977 DOI: 10.1093/ijnp/pyw102] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 11/07/2016] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Studies have shown the involvement of cannabinoid (CB) receptors in the behavioral and neurobiological effects of psychostimulants. Most of these studies have focused on the role of CB1 receptors in the psychostimulant effects of cocaine, while very few have investigated the respective role of CB2 receptors. Further studies are warranted to elucidate the extent of CB receptor involvement in the expression of cocaine-induced effects. METHODS The role of CB1 and CB2 receptors in the rewarding and motor properties of cocaine was assessed in conditioned place preference, conditioned motor activity, and open field activity in rats. RESULTS The CB1 receptor antagonist rimonabant (3 mg/kg) decreased the acquisition and the expression of conditioned place preference induced by cocaine (20 mg/kg). Rimonabant inhibited cocaine-elicited conditioned motor activity when administered during the expression of cocaine-induced conditioned place preference. Rimonabant decreased ambulatory and vertical activity induced by cocaine. The CB2 receptor agonist JWH-133 (10 mg/kg) decreased the acquisition and the expression of cocaine-induced conditioned place preference. JWH-133 inhibited cocaine-elicited conditioned motor activity when administered during the acquisition and the expression of cocaine-induced conditioned place preference. JWH-133 decreased ambulatory activity and abolished vertical activity induced by cocaine. The effects of JWH-133 on cocaine conditioned and stimulated responses were abolished when the CB2 receptor antagonist/inverse agonist AM630 (5 mg/kg) was preadministered. CONCLUSIONS Cannabinoid CB1 and CB2 receptors modulate cocaine-induced rewarding behavior and appear to have opposite roles in the regulation of cocaine's reinforcing and psychomotor effects.
Collapse
Affiliation(s)
- Foteini Delis
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece (Dr Delis, Dr Polissidis, Ms Poulia, and Dr Anoniou)
| | - Alexia Polissidis
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece (Dr Delis, Dr Polissidis, Ms Poulia, and Dr Anoniou);,Laboratory of Neurodegenerative Diseases, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece (Dr Polissidis)
| | - Nafsika Poulia
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece (Dr Delis, Dr Polissidis, Ms Poulia, and Dr Anoniou)
| | - Zuzana Justinova
- Preclinical Pharmacology Section, Behavioral Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, DHHS, Baltimore, MD (Drs Justinova and Goldberg)
| | - George G. Nomikos
- Global Clinical Science, Takeda Development Center Americas, Inc, Deerfield, IL (Dr Nomikos)
| | - Steven R. Goldberg
- Preclinical Pharmacology Section, Behavioral Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, DHHS, Baltimore, MD (Drs Justinova and Goldberg)
| | - Katerina Antoniou
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece (Dr Delis, Dr Polissidis, Ms Poulia, and Dr Anoniou)
| |
Collapse
|
49
|
Yu YJ, Huang CH, Chang CH, Gean PW. Involvement of protein phosphatases in the destabilization of methamphetamine-associated contextual memory. ACTA ACUST UNITED AC 2016; 23:486-93. [PMID: 27531839 PMCID: PMC4986857 DOI: 10.1101/lm.039941.115] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 07/08/2016] [Indexed: 01/20/2023]
Abstract
Destabilization refers to a memory that becomes unstable when reactivated and is susceptible to disruption by amnestic agents. Here we delineated the cellular mechanism underlying the destabilization of drug memory. Mice were conditioned with methamphetamine (MeAM) for 3 d, and drug memory was assessed with a conditioned place preference (CPP) protocol. Anisomycin (ANI) was administered 60 min after the CPP retrieval to disrupt reconsolidation. We found that destabilization of MeAM CPP after the application of ANI was blocked by the N-methyl-d-aspartate receptor (NMDAR) antagonist MK-801 and the NR2B antagonist ifenprodil (IFN) but not by the NR2A antagonist NVP-AAM077 (NVP). In addition, decrease in the phosphorylation of GluR1 at Serine845 (p-GluR1-Ser845), decrease in spine density, and a reduction in the AMPAR/NMDAR ratio in the basolateral amygdala (BLA) were reversed after the MK-801 treatment. The effect of ANI on destabilization was prevented by the protein phosphatase 2B (calcineurin, CaN) inhibitors cyclosporine A (CsA) and FK-506 and the protein phosphatase 1 (PP1) inhibitors calyculin A (CA) and okadaic acid (OA). These results suggest that memory destabilization involves the activation of NR2B-containing NMDARs, which in turn allows the influx of Ca2+. Increased intracellular Ca2+ stimulates CaN, leading to the dephosphorylation and inactivation of inhibitor 1 and the activation of PP1. PP1 then dephosphorylates p-GluR1-Ser845 to elicit AMPA receptor (AMPAR) endocytosis and destabilization of the drug memory.
Collapse
Affiliation(s)
- Yang-Jung Yu
- Institute of Basic Medical Sciences and Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan, Taiwan 701
| | - Chien-Hsuan Huang
- Institute of Basic Medical Sciences and Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan, Taiwan 701
| | - Chih-Hua Chang
- Institute of Basic Medical Sciences and Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan, Taiwan 701
| | - Po-Wu Gean
- Institute of Basic Medical Sciences and Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan, Taiwan 701
| |
Collapse
|
50
|
NMDA antagonist MK 801 in nucleus accumbens core but not shell disrupts the restraint stress-induced reinstatement of extinguished cocaine-conditioned place preference in rats. Behav Brain Res 2016; 315:150-9. [PMID: 27506656 DOI: 10.1016/j.bbr.2016.08.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 08/04/2016] [Accepted: 08/05/2016] [Indexed: 12/27/2022]
Abstract
Relapse is a common feature of cocaine addiction. In rodents, it can be elicited by cues, stress or the drug. Restraint stress-induced reinstatement of cocaine-conditioned place preference (CPP) is a useful model to study the mechanisms involved in stress-induced relapse of drug-seeking behavior. There is evidence that the glutamate NMDA receptors are critically involved in drug- and cue-induced reinstatement of seeking behavior and drug-CPP responses. The aim of this study was to investigate the contribution of NMDA receptors within core vs. shell nucleus accumbens (NAc) subregions to restraint stress-induced reinstatement of extinguished cocaine-CPP. After extinction of cocaine-conditioned preference, animals were administered MK 801 systemically or directly into intra-core or intra-shell, and restrained for 30min or left undisturbed in their home-cages. First, we demonstrated that restraint stress-induced reinstatement of extinguished cocaine-CPP depends on the duration of restraint as well as on the context in which it is applied. Second, this effect was blocked by systemic MK 801 administration either before or after restraint. Third, intra-core but not intra-shell administration abrogated the restraint stress-induced reinstatement. These findings show that NMDA receptors within NAc core, but not shell, play a critical role in restraint stress-induced reinstatement of cocaine-CPP.
Collapse
|