1
|
Cui Y, Huang Y, Li X, Zhang X, Zhang Z, Wang P, Lu X. Joint effects of prescription opioid use and physical activity on depressive symptoms. Public Health 2024; 236:430-435. [PMID: 39305661 DOI: 10.1016/j.puhe.2024.08.017] [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: 04/02/2024] [Revised: 06/04/2024] [Accepted: 08/15/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVES The possible interaction of prescription opioid use and physical activity with regard to depressive symptoms has not been well studied. This study aimed to investigate the joint effects of prescription opioid use and physical activity on depressive symptoms. STUDY DESIGN Cross-sectional study. METHODS This cross-sectional study included 29,542 participants from the National Health and Nutrition Examination Survey (2007 to March 2020). Depressive symptoms were evaluated using the Patient Health Questionnaire-9. Multivariable logistic regression models were used to examine the association. RESULTS Of the 29,542 adults, 2598 had depressive symptoms (weighted, 7.7%), 1845 used prescription opioids (weighted, 6.0%), and 18,373 (weighted 67.0%) achieved the recommended physical activity. After multivariable adjustment, the odds ratio (OR) of depressive symptoms was 4.06 (95% confidence interval [CI]: 3.28, 5.02) for both prescription opioid use and inactive physical activity compared to those without either condition. No multiplicative interaction was observed for prescription opioid use and inactive physical activity on depressive symptoms (OR = 1.26 [95% CI: 0.87, 1.81]). However, additive interaction was statistically significant between the 2 exposures (relative excess risk due to interaction = 1.34 [95% CI: 0.31, 2.36]; attributable proportion due to interaction = 0.33 [95% CI: 0.12, 0.54]; synergy index = 1.78 [95% CI: 1.12, 2.83]). CONCLUSIONS Prescription opioid use and inactive physical activity interacted synergistically to affect depressive symptoms.
Collapse
Affiliation(s)
- Yaping Cui
- Department of Anesthesiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Yuanyuan Huang
- Department of Anesthesiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Xilong Li
- Department of Anesthesiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Xindan Zhang
- The 1st Clinical Department, China Medical University, Shenyang 110122, China
| | - Zhen Zhang
- Department of Anesthesiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China.
| | - Peng Wang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, The Key Laboratory of Precision Anesthesia & Perioperative Organ Protection, Guangzhou, Guangdong, 510515, China.
| | - Xihua Lu
- Department of Anesthesiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China.
| |
Collapse
|
2
|
Salas-Estrada L, Provasi D, Qiu X, Kaniskan HÜ, Huang XP, DiBerto JF, Lamim Ribeiro JM, Jin J, Roth BL, Filizola M. De Novo Design of κ-Opioid Receptor Antagonists Using a Generative Deep-Learning Framework. J Chem Inf Model 2023; 63:5056-5065. [PMID: 37555591 PMCID: PMC10466374 DOI: 10.1021/acs.jcim.3c00651] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Indexed: 08/10/2023]
Abstract
Likely effective pharmacological interventions for the treatment of opioid addiction include attempts to attenuate brain reward deficits during periods of abstinence. Pharmacological blockade of the κ-opioid receptor (KOR) has been shown to abolish brain reward deficits in rodents during withdrawal, as well as to reduce the escalation of opioid use in rats with extended access to opioids. Although KOR antagonists represent promising candidates for the treatment of opioid addiction, very few potent selective KOR antagonists are known to date and most of them exhibit significant safety concerns. Here, we used a generative deep-learning framework for the de novo design of chemotypes with putative KOR antagonistic activity. Molecules generated by models trained with this framework were prioritized for chemical synthesis based on their predicted optimal interactions with the receptor. Our models and proposed training protocol were experimentally validated by binding and functional assays.
Collapse
Affiliation(s)
- Leslie Salas-Estrada
- Department
of Pharmacological Sciences, Icahn School
of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Davide Provasi
- Department
of Pharmacological Sciences, Icahn School
of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Xing Qiu
- Department
of Pharmacological Sciences, Icahn School
of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Husnu Ümit Kaniskan
- Department
of Pharmacological Sciences, Icahn School
of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Xi-Ping Huang
- National
Institute of Mental Health, Psychoactive Drug Screening Program, Department
of Pharmacology, University of North Carolina
School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Jeffrey F. DiBerto
- National
Institute of Mental Health, Psychoactive Drug Screening Program, Department
of Pharmacology, University of North Carolina
School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - João Marcelo Lamim Ribeiro
- Department
of Pharmacological Sciences, Icahn School
of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Jian Jin
- Department
of Pharmacological Sciences, Icahn School
of Medicine at Mount Sinai, New York, New York 10029, United States
- Mount
Sinai Center for Therapeutics Discovery, Departments of Oncological
Sciences and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Bryan L. Roth
- National
Institute of Mental Health, Psychoactive Drug Screening Program, Department
of Pharmacology, University of North Carolina
School of Medicine, Chapel Hill, North Carolina 27599, United States
- Division
of Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill Eshelman School of Pharmacy, Chapel Hill, North Carolina 27599, United States
| | - Marta Filizola
- Department
of Pharmacological Sciences, Icahn School
of Medicine at Mount Sinai, New York, New York 10029, United States
| |
Collapse
|
3
|
Salas-Estrada L, Provasi D, Qui X, Kaniskan HÜ, Huang XP, DiBerto JF, Ribeiro JML, Jin J, Roth BL, Filizola M. De Novo Design of κ-Opioid Receptor Antagonists Using a Generative Deep Learning Framework. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.25.537995. [PMID: 37162828 PMCID: PMC10168226 DOI: 10.1101/2023.04.25.537995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Likely effective pharmacological interventions for the treatment of opioid addiction include attempts to attenuate brain reward deficits during periods of abstinence. Pharmacological blockade of the κ-opioid receptor (KOR) has been shown to abolish brain reward deficits in rodents during withdrawal, as well as to reduce the escalation of opioid use in rats with extended access to opioids. Although KOR antagonists represent promising candidates for the treatment of opioid addiction, very few potent selective KOR antagonists are known to date and most of them exhibit significant safety concerns. Here, we used a generative deep learning framework for the de novo design of chemotypes with putative KOR antagonistic activity. Molecules generated by models trained with this framework were prioritized for chemical synthesis based on their predicted optimal interactions with the receptor. Our models and proposed training protocol were experimentally validated by binding and functional assays.
Collapse
|
4
|
Blockade of kappa opioid receptors reduces mechanical hyperalgesia and anxiety-like behavior in a rat model of trigeminal neuropathic pain. Behav Brain Res 2022; 417:113595. [PMID: 34592375 DOI: 10.1016/j.bbr.2021.113595] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 09/12/2021] [Accepted: 09/20/2021] [Indexed: 12/18/2022]
Abstract
It has been shown that kappa opioid receptor (KOR) antagonists, such as nor-binaltorphimine (nor-BNI), have antinociceptive effects in some pain models that affect the trigeminal system. Also, its anxiolytic-like effect has been extensively demonstrated in the literature. The present study aimed to investigate the systemic, local, and central effect of nor-BNI on trigeminal neuropathic pain using the infraorbital nerve constriction model (CCI-ION), as well as to evaluate its effect on anxiety-like behavior associated with this model. Animals received nor-BNI systemically; in the trigeminal ganglion (TG); in the subarachnoid space to target the spinal trigeminal nucleus caudalis (Sp5C) or in the central amygdala (CeA) 14 days after CCI-ION surgery. Systemic administration of nor-BNI caused a significant reduction of facial mechanical hyperalgesia and promoted an anxiolytic-like effect, which was detected in the elevated plus-maze and the light-dark transition tests. When administered in the TG or CeA, the KOR antagonist was able to reduce facial mechanical hyperalgesia induced by CCI-ION, but without changing the anxiety-like behavior. Moreover, no change was observed on nociception and anxiety-like behavior after nor-BNI injection into the Sp5C. The present study demonstrated antinociceptive and anxiolytic-like effects of nor-BNI in a model of trigeminal neuropathic pain. The antinociceptive effect seems to be dissociated from the anxiolytic-like effect, at both the sites involved and at the dose need to achieve the effect. In conclusion, the kappa opioid system may represent a promising target to be explored for the control of trigeminal pain and associated anxiety. However, further studies are necessary to better elucidate its functioning and modulatory role in chronic trigeminal pain states.
Collapse
|
5
|
Alshehri FS, Alghamdi BS, Hakami AY, Alshehri AA, Althobaiti YS. Melatonin attenuates morphine-induced conditioned place preference in Wistar rats. Brain Behav 2021; 11:e2397. [PMID: 34710287 PMCID: PMC8671767 DOI: 10.1002/brb3.2397] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 08/04/2021] [Accepted: 09/06/2021] [Indexed: 02/03/2023] Open
Abstract
PURPOSE Morphine is the predominantly used drug for postoperative and cancer pain management. However, the abuse potential of morphine is the primary disadvantage of using opioids in pain management. Melatonin is a neurohormone synthesized in the pineal gland and is involved in circadian rhythms in mammals, as well as other physiological functions. Melatonin provenly attenuates alcohol-seeking and relapse behaviors in rats. Therefore, we aimed to investigate the involvement of the melatonergic system in attenuating morphine dependence. MATERIALS AND METHODS Male Wistar rats were divided into three groups: control, morphine, and morphine + melatonin. Animals were habituated for 3 days, and the initial preference was evaluated. Following the initial preference, the control group received the vehicle and was placed for a 45-min session in the assigned chamber every day, alternating between the two chambers, for 8 days. The morphine group received a morphine injection (5 mg/kg, IP) and was placed for a 45-min session in the white chamber, for a total of four sessions. The morphine + melatonin group received the morphine injection (5 mg/kg, IP) for a total of four sessions over an 8-day period. In the posttest session, the control and morphine groups received a vehicle injection 30 min before placement in the conditioned place preference (CPP). The morphine + melatonin group received a single injection of melatonin (50 mg/kg, IP) 30 min before the preference test. RESULTS Statistical analysis revealed that repeated administration of morphine for four sessions produced a significant increase in the CPP score in the morphine group compared to the control group. However, a single melatonin injection administered 30 min before the posttest attenuated morphine-seeking behavior and reduced morphine-induced place preference. CONCLUSION These findings provide novel evidence for the role of the melatonergic system as a potential target in modulating morphine-seeking behavior.
Collapse
Affiliation(s)
- Fahad S Alshehri
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Badrah S Alghamdi
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia.,Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Alqassem Y Hakami
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia.,King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Abdullah A Alshehri
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Yusuf S Althobaiti
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia.,College of Pharmacy, Addiction and Neuroscience Research Unit, Taif University, Taif, 21944, Saudi Arabia
| |
Collapse
|
6
|
Wu LX, Dong YP, Zhu QM, Zhang B, Ai BL, Yan T, Zhang GH, Sun L. Effects of dezocine on morphine tolerance and opioid receptor expression in a rat model of bone cancer pain. BMC Cancer 2021; 21:1128. [PMID: 34670518 PMCID: PMC8529774 DOI: 10.1186/s12885-021-08850-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 10/05/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Clinically, the coadministration of opioids to enhance antinociception and decrease tolerance has attracted increasing research attention. We investigated the effects of dezocine, a mu- and kappa-opioid receptor agonist/antagonist, on morphine tolerance and explored the involvement of opioid receptor expression in a rat model of bone cancer pain. METHODS Thermal nociceptive thresholds were measured after the subcutaneous injection of morphine (10 mg/kg) alone or combined with dezocine (10 or 1 mg/kg) for 7 consecutive days. Real-time PCR and western blot analysis were used to examine opioid receptor expression in the periaqueductal gray (PAG) and spinal cord. RESULTS The analgesic effect was significantly decreased after 4 days of morphine administration. We observed that low-dose dezocine significantly attenuated morphine tolerance without reducing the analgesic effect of morphine. Low-dose dezocine coadministration significantly reversed the downregulated expression of mu (MOR) and delta (DOR) opioid receptors in the PAG and the upregulated expression of kappa (KOR) and DOR in the spinal cord induced by morphine. Moreover, low-dose dezocine coadministered with morphine significantly inhibited KOR expression in both the PAG and spinal cord. CONCLUSIONS The combination of low-dose dezocine with morphine may prevent or delay the development of morphine tolerance in a rat model of bone cancer pain. The regulation of opioid receptor expression in the PAG and spinal cord may be part of the mechanism.
Collapse
MESH Headings
- Animals
- Female
- Rats
- Analgesics, Opioid/administration & dosage
- Analgesics, Opioid/pharmacology
- Bone Neoplasms/complications
- Bridged Bicyclo Compounds, Heterocyclic/administration & dosage
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Cancer Pain/drug therapy
- Cancer Pain/metabolism
- Cell Line, Tumor
- Down-Regulation/drug effects
- Drug Interactions
- Drug Therapy, Combination/methods
- Drug Tolerance
- Hot Temperature
- Hyperalgesia/physiopathology
- Morphine/administration & dosage
- Morphine/pharmacology
- Pain Measurement/drug effects
- Pain Threshold
- Periaqueductal Gray/metabolism
- Rats, Wistar
- Receptors, Opioid/drug effects
- Receptors, Opioid/metabolism
- Spinal Cord/metabolism
- Tetrahydronaphthalenes/administration & dosage
- Tetrahydronaphthalenes/pharmacology
- Up-Regulation/drug effects
- Disease Models, Animal
Collapse
Affiliation(s)
- Lin-Xin Wu
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yan-Peng Dong
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qian-Mei Zhu
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Bo Zhang
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Bo-Lun Ai
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tao Yan
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Guo-Hua Zhang
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Li Sun
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518100, China.
| |
Collapse
|
7
|
Blanco-Gandia MC, Montagud-Romero S, Rodríguez-Arias M. Binge eating and psychostimulant addiction. World J Psychiatry 2021; 11:517-529. [PMID: 34631457 PMCID: PMC8475000 DOI: 10.5498/wjp.v11.i9.517] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 05/13/2021] [Accepted: 07/27/2021] [Indexed: 02/06/2023] Open
Abstract
Many of the various factors, characteristics, and variables involved in the addictive process can determine an individual’s vulnerability to develop drug addiction. Hedonic eating, based on pleasure rather than energy needs, modulates the same reward circuits, as do drugs of abuse. According to the last report of the World Health Organization, the worldwide obesity rate has more than doubled since 1980, reaching especially critical levels in children and young people, who are overexposed to high-fat, high-sugar, energy-dense foods. Over the past few decades, there has been an increase in the number of studies focused on how eating disorders can lead to the development of drug addiction and on the comorbidity that exists between the two disorders. Herein, we review the most recent research on the subject, focusing especially on animal models of binge eating disorders and drug addiction. The complex profile of patients with substance use and binge eating disorders requires an integrated response to dually diagnosed patients. Nutritional patterns should be considered an important variable in the treatment of substance use disorders, and future studies need to focus on specific treatments and interventions in individuals who show a special vulnerability to shift from one addiction to the other.
Collapse
Affiliation(s)
| | | | - Marta Rodríguez-Arias
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia 46010, Spain
| |
Collapse
|
8
|
Rosoff DB, Smith GD, Lohoff FW. Prescription Opioid Use and Risk for Major Depressive Disorder and Anxiety and Stress-Related Disorders: A Multivariable Mendelian Randomization Analysis. JAMA Psychiatry 2021; 78:151-160. [PMID: 33175090 PMCID: PMC7658804 DOI: 10.1001/jamapsychiatry.2020.3554] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
IMPORTANCE Growing evidence suggests that prescription opioid use affects depression and anxiety disorders; however, observational studies are subject to confounding, making causal inference and determining the direction of these associations difficult. OBJECTIVE To investigate the potential bidirectional associations between the genetic liability for prescription opioid and other nonopioid pain medications and both major depressive disorder (MDD) and anxiety and stress-related disorders (ASRD) using genetically based methods. DESIGN, SETTING, AND PARTICIPANTS We performed 2-sample mendelian randomization (MR) using summary statistics from genome-wide association studies (GWAS) to assess potential associations of self-reported prescription opioid and nonopioid analgesics, including nonsteroidal anti-inflammatories (NSAIDs) and acetaminophen-like derivatives use with MDD and ASRD. The GWAS data were derived from participants of predominantly European ancestry included in observational cohorts. Data were analyzed February 20, 2020, to May 4, 2020. MAIN OUTCOMES AND MEASURES Major depressive disorder, ASRD, and self-reported pain medications (opioids, NSAIDs, anilides, and salicylic acid). RESULTS The GWAS data were derived from participants of predominantly European ancestry included in the population-based UK Biobank and Lundbeck Foundation Initiative for Integrative Psychiatric Research studies: approximately 54% of the initial UK Biobank sample and 55.6% of the Lundbeck Foundation Initiative for Integrative Psychiatric Research sample selected for the ASRD GWAS were women. In a combined sample size of 737 473 study participants, single-variable MR showed that genetic liability for increased prescription opioid use was associated with increased risk of both MDD (odds ratio [OR] per unit increase in log odds opioid use, 1.14; 95% CI, 1.06-1.22; P < .001) and ASRD (OR, 1.24; 95% CI, 1.07-1.44; P = .004). Using multivariable MR, these opioid use estimates remained after accounting for other nonopioid pain medications (MDD OR, 1.14; 95% CI, 1.04-1.25; P = .005; ASRD OR, 1.30; 95% CI, 1.08-1.46; P = .006), and in separate models, accounting for comorbid pain conditions. Bidirectional analyses showed that genetic liability for MDD but not ASRD was associated with increased prescription opioid use risk (OR, 1.18; 95% CI, 1.08-1.30; P < .001). These estimates were generally consistent across single-variable and multivariable inverse variance-weighted (MV-IVW) and MR-Egger sensitivity analyses. Pleiotropy-robust methods did not indicate bias in any MV-IVW estimates. CONCLUSIONS AND RELEVANCE The findings of this mendelian randomization analysis suggest evidence for potential causal associations between the genetic liability for increased prescription opioid use and the risk for MDD and ASRD. While replication studies are necessary, these findings may inform prevention and intervention strategies directed toward the opioid epidemic and depression.
Collapse
Affiliation(s)
- Daniel B. Rosoff
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - George Davey Smith
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, England
| | - Falk W. Lohoff
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
9
|
Robinson SA, Jones AD, Brynildsen JK, Ehrlich ME, Blendy JA. Neurobehavioral effects of neonatal opioid exposure in mice: Influence of the OPRM1 SNP. Addict Biol 2020; 25:e12806. [PMID: 31267641 PMCID: PMC8450766 DOI: 10.1111/adb.12806] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 06/05/2019] [Accepted: 06/06/2019] [Indexed: 01/09/2023]
Abstract
Opioid use among pregnant women is a growing public health concern in the United States. Infants exposed to opioids in utero are at risk of exhibiting neonatal opioid withdrawal syndrome (NOWS). The biological mechanisms underlying short and long-term consequences of in utero opioid exposure and NOWS are unknown. A potential genetic factor is a single-nucleotide polymorphism (SNP) in the mu-opioid receptor gene (OPRM1 A118G). Opioid exposed infants with the G-allele spend less time in hospitals after birth. To determine whether this SNP modulates the neurobehavioral effects of neonatal opioid exposure and withdrawal, we used mice possessing the equivalent Oprm1 SNP (A112G). Pups were treated chronically with saline or morphine from postnatal days (PNDs) 1 to 14, a developmental period equivalent to the third trimester of a human pregnancy and a sensitive period for opioid exposure in rodents. Morphine treatment produced significant developmental delays regardless of genotype and increased total ultrasonic vocalizations in males during spontaneous withdrawal. Animals were aged and tested for anxiety and drug response during adolescence and adulthood, respectively. AA morphine-treated animals showed reduced activity in the marble burying task compared with saline controls; however, this effect was absent in AG and GG animals. As adults, AA males exposed to morphine from PNDs 1 to 14 exhibited enhanced development of locomotor sensitization to morphine, whereas females showed reduced locomotor sensitization. These data suggest the involvement of the Oprm1 SNP for certain outcomes of neonatal opioid exposure and highlight the importance of considering sex and genetic variability for the prognosis of NOWS.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Behavior, Animal/drug effects
- Brain/drug effects
- Disease Models, Animal
- Female
- Male
- Mice
- Mice, Inbred C57BL
- Opioid-Related Disorders/genetics
- Polymorphism, Single Nucleotide/drug effects
- Polymorphism, Single Nucleotide/genetics
- Pregnancy
- Prenatal Exposure Delayed Effects/genetics
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/genetics
Collapse
Affiliation(s)
- Shivon A. Robinson
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia PA, 19104
| | - Alonzo D. Jones
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia PA, 19104
| | - Julia K. Brynildsen
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia PA, 19104
| | - Michelle E. Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York NY, 10029
| | - Julie A. Blendy
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia PA, 19104
| |
Collapse
|
10
|
Reed B, Butelman ER, Kreek MJ. Kappa Opioid Receptor Antagonists as Potential Therapeutics for Mood and Substance Use Disorders. Handb Exp Pharmacol 2020; 271:473-491. [PMID: 33174064 DOI: 10.1007/164_2020_401] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The kappa opioid receptor (KOR) and its primary cognate ligands, the dynorphin peptides, are involved in diverse physiological processes. Disruptions to the KOR/dynorphin system have been found to likely play a role in multiple neuropsychological disorders, and hence KOR has emerged as a potential therapeutic target. Targeting KOR is complicated by close homology to the mu and delta opioid receptors (MOR and DOR), and many KOR ligands have at least moderate affinity to MOR and/or DOR. Animal models utilizing primarily very long-lasting selective KOR antagonists (>3 weeks following a single dose) have demonstrated that KOR antagonism attenuates certain anxiety-like and depression-like behaviors and blocks stress- and cue-induced reinstatement to drug seeking. Recently, relatively selective KOR antagonists with medication-like pharmacokinetic and pharmacodynamic properties and durations of action have been developed. One of these, JNJ-67953964 (also referred to as CERC-501, LY2456302, OpraKappa or Aticaprant) has been studied in humans, and shown to be safe, relatively KOR selective, and able to substantially attenuate binding of a KOR PET tracer to CNS localized KOR for greater than 24 h. While animal studies have indicated that compounds of this structural class are capable of normalizing withdrawal signs in animal models of cocaine and alcohol dependence and reducing cocaine and alcohol intake/seeking, additional studies are needed to determine the value of these second generation KOR antagonists in treating mood disorders and substance use disorders in humans.
Collapse
Affiliation(s)
- Brian Reed
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA.
| | - Eduardo R Butelman
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA
| | - Mary Jeanne Kreek
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA
| |
Collapse
|
11
|
Listos J, Łupina M, Talarek S, Mazur A, Orzelska-Górka J, Kotlińska J. The Mechanisms Involved in Morphine Addiction: An Overview. Int J Mol Sci 2019; 20:ijms20174302. [PMID: 31484312 PMCID: PMC6747116 DOI: 10.3390/ijms20174302] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/23/2019] [Accepted: 08/29/2019] [Indexed: 12/22/2022] Open
Abstract
Opioid use disorder is classified as a chronic recurrent disease of the central nervous system (CNS) which leads to personality disorders, co-morbidities and premature death. It develops as a result of long-term administration of various abused substances, along with morphine. The pharmacological action of morphine is associated with its stimulation of opioid receptors. Opioid receptors are a group of G protein-coupled receptors and activation of these receptors by ligands induces significant molecular changes inside the cell, such as an inhibition of adenylate cyclase activity, activation of potassium channels and reductions of calcium conductance. Recent data indicate that other signalling pathways also may be involved in morphine activity. Among these are phospholipase C, mitogen-activated kinases (MAP kinases) or β-arrestin. The present review focuses on major mechanisms which currently are considered as essential in morphine activity and dependence and may be important for further studies.
Collapse
Affiliation(s)
- Joanna Listos
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4a St., 20-093 Lublin, Poland.
| | - Małgorzata Łupina
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4a St., 20-093 Lublin, Poland.
| | - Sylwia Talarek
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4a St., 20-093 Lublin, Poland.
| | - Antonina Mazur
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4a St., 20-093 Lublin, Poland.
| | - Jolanta Orzelska-Górka
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4a St., 20-093 Lublin, Poland.
| | - Jolanta Kotlińska
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4a St., 20-093 Lublin, Poland.
| |
Collapse
|
12
|
Carlezon WA, Krystal AD. Kappa-Opioid Antagonists for Psychiatric Disorders: From Bench to Clinical Trials. Depress Anxiety 2016; 33:895-906. [PMID: 27699938 PMCID: PMC5288841 DOI: 10.1002/da.22500] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 03/09/2016] [Accepted: 03/09/2016] [Indexed: 12/15/2022] Open
Abstract
Kappa-opioid receptor (KOR) antagonists are currently being considered for the treatment of a variety of neuropsychiatric conditions, including depressive, anxiety, and substance abuse disorders. A general ability to mitigate the effects of stress, which can trigger or exacerbate these conditions, may explain their putative efficacy across such a broad array of conditions. The discovery of their potentially therapeutic effects evolved from preclinical research designed to characterize the molecular mechanisms by which experience causes neuroadaptations in the nucleus accumbens (NAc), a key element of brain reward circuitry. This research established that exposure to drugs of abuse or stress increases the activity of the transcription factor CREB (cAMP response element binding protein) in the NAc, which leads to elevated expression of the opioid peptide dynorphin that in turn causes core signs of depressive- and anxiety-related disorders. Disruption of KORs-the endogenous receptors for dynorphin-produces antidepressant- and anxiolytic-like actions in screening procedures that identify standard drugs of these classes, and reduces stress effects in tests used to study addiction and stress-related disorders. Although interest in this target is high, prototypical KOR antagonists have extraordinarily persistent pharmacodynamic effects that complicate clinical trials. The development of shorter acting KOR antagonists together with more rapid designs for clinical trials may soon provide insight on whether these drugs are efficacious as would be predicted by preclinical work. If successful, KOR antagonists would represent a unique example in psychiatry where the therapeutic mechanism of a drug class is understood before it is shown to be efficacious in humans.
Collapse
Affiliation(s)
- William A. Carlezon
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont MA
| | - Andrew D. Krystal
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC
| |
Collapse
|
13
|
Effects of the adenosinergic system on the expression and acquisition of sensitization to conditioned place preference in morphine-conditioned rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2015; 389:233-41. [PMID: 26638209 PMCID: PMC4715847 DOI: 10.1007/s00210-015-1190-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 11/05/2015] [Indexed: 10/31/2022]
Abstract
In the presented study, we attempt to investigate if the sensitization to conditioned place preference (CPP) induced by low doses of morphine was developed in rats which have been previously conditioned with morphine. The experiments were performed in the CPP test. Firstly, it has been demonstrated that administration of ineffective dose of morphine on the 9th day induces the increase in time spent of rats at a morphine-paired compartment, confirming that sensitization to CPP has been developed in these animals. Secondly, it has been shown that stimulation of A1 receptor significantly inhibits the expression of morphine-induced of sensitization, and blockade of these receptors produces the opposite effect. Finally, it has been indicated that both stimulation and blockade of A1 and/or A2A receptors inhibit the acquisition of sensitization to CPP. The obtained results have strongly supported the significance of adenosinergic system in both expression and acquisition of studied sensitization. These results seem to be important for the identification of connections in the central nervous system which can help finding new strategies to attenuate rewarding action of morphine.
Collapse
|
14
|
Craig MM, Bajic D. Long-term behavioral effects in a rat model of prolonged postnatal morphine exposure. Behav Neurosci 2015; 129:643-55. [PMID: 26214209 PMCID: PMC4586394 DOI: 10.1037/bne0000081] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Prolonged morphine treatment in neonatal pediatric populations is associated with a high incidence of opioid tolerance and dependence. Despite the clinical relevance of this problem, our knowledge of long-term consequences is sparse. The main objective of this study was to investigate whether prolonged morphine administration in a neonatal rat is associated with long-term behavioral changes in adulthood. Newborn animals received either morphine (10 mg/kg) or equal volume of saline subcutaneously twice daily for the first 2 weeks of life. Morphine-treated animals underwent 10 days of morphine weaning to reduce the potential for observable physical signs of withdrawal. Animals were subjected to nonstressful testing (locomotor activity recording and a novel-object recognition test) at a young age (Postnatal Days [PDs] 27-31) or later in adulthood (PDs 55-56), as well as stressful testing (calibrated forceps test, hot plate test, and forced swim test) only in adulthood. Analysis revealed that prolonged neonatal morphine exposure resulted in decreased thermal but not mechanical threshold. Importantly, no differences were found for total locomotor activity (proxy of drug reward/reinforcement behavior), individual forced swim test behaviors (proxy of affective processing), or novel-object recognition test. Performance on the novel-object recognition test was compromised in the morphine-treated group at the young age, but the effect disappeared in adulthood. These novel results provide insight into the long-term consequences of opioid treatment during an early developmental period and suggest long-term neuroplastic differences in sensory processing related to thermal stimuli.
Collapse
Affiliation(s)
- Michael M. Craig
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA, USA
| | - Dusica Bajic
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA, USA
- Department of Anaesthesia, Harvard Medical School, 25 Shattuck St., Boston, MA, USA
| |
Collapse
|
15
|
Bajic D, Soiza-Reilly M, Spalding AL, Berde CB, Commons KG. Endogenous cholinergic neurotransmission contributes to behavioral sensitization to morphine. PLoS One 2015; 10:e0117601. [PMID: 25647082 PMCID: PMC4315441 DOI: 10.1371/journal.pone.0117601] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 12/29/2014] [Indexed: 12/15/2022] Open
Abstract
Neuroplasticity in the mesolimbic dopaminergic system is critical for behavioral adaptations associated with opioid reward and addiction. These processes may be influenced by cholinergic transmission arising from the laterodorsal tegmental nucleus (LDTg), a main source of acetylcholine to mesolimbic dopaminergic neurons. To examine this possibility we asked if chronic systemic morphine administration affects expression of genes in ventral and ventrolateral periaqueductal gray at the level of the LDTg using rtPCR. Specifically, we examined gene expression changes in the area of interest using Neurotransmitters and Receptors PCR array between chronic morphine and saline control groups. Analysis suggested that chronic morphine administration led to changes in expression of genes associated, in part, with cholinergic neurotransmission. Furthermore, using a quantitative immunofluorescent technique, we found that chronic morphine treatment produced a significant increase in immunolabeling of the cholinergic marker (vesicular acetylcholine transporter) in neurons of the LDTg. Finally, systemic administration of the nonselective and noncompetitive neuronal nicotinic antagonist mecamylamine (0.5 or 2 mg/kg) dose-dependently blocked the expression, and to a lesser extent the development, of locomotor sensitization. The same treatment had no effect on acute morphine antinociception, antinociceptive tolerance or dependence to chronic morphine. Taken together, the results suggest that endogenous nicotinic cholinergic neurotransmission selectively contributes to behavioral sensitization to morphine and this process may, in part, involve cholinergic neurons within the LDTg.
Collapse
Affiliation(s)
- Dusica Bajic
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA, 02115, United States of America
- Department of Anaesthesia, Harvard Medical School, 25 Shattuck St., Boston, MA, 02115, United States of America
| | - Mariano Soiza-Reilly
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA, 02115, United States of America
- Department of Anaesthesia, Harvard Medical School, 25 Shattuck St., Boston, MA, 02115, United States of America
| | - Allegra L. Spalding
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA, 02115, United States of America
| | - Charles B. Berde
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA, 02115, United States of America
- Department of Anaesthesia, Harvard Medical School, 25 Shattuck St., Boston, MA, 02115, United States of America
| | - Kathryn G. Commons
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA, 02115, United States of America
- Department of Anaesthesia, Harvard Medical School, 25 Shattuck St., Boston, MA, 02115, United States of America
| |
Collapse
|
16
|
Prescription opioid analgesics increase the risk of depression. J Gen Intern Med 2014; 29:491-9. [PMID: 24165926 PMCID: PMC3930792 DOI: 10.1007/s11606-013-2648-1] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 07/02/2013] [Accepted: 09/18/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Prescription opioid analgesic use has quintupled recently. Evidence linking opioid use with depression emanates from animal models and studies of persons with co-occurring substance use and major depression. Little is known about depressogenic effects of opioid use in other populations. OBJECTIVE The purpose of this study was to determine whether prescription opioids are associated with increased risk of diagnosed depression. DESIGN Retrospective cohort study, new user design. PATIENTS Medical record data from 49,770 US Department of Veterans Affairs (VA) health care system patients with no recent (24-month) history of opioid use or a diagnosis of depression in 1999 and 2000. MAIN MEASURES Propensity scores were used to control for bias by indication, and the data were weighted to balance the distribution of covariates by duration of incident opioid exposure. Cox proportional hazard models with adjustment for painful conditions were used to estimate the association between duration of prescription opioid use and the subsequent risk of development of depression between 2001 and 2007. KEY RESULTS Of 49,770 patients who were prescribed an opioid analgesic, 91 % had a prescription for < 90 days, 4 % for 90-180 days, and 5 % for > 180 days. Compared to patients whose prescription was for < 90 days, the risk of depression increased significantly as the duration of opioid prescription increased (HR = 1.25; 95 % CI: 1.05-1.46 for 90-180 days, and HR = 1.51; 95 % CI:1.31-1.74 for > 180 days). CONCLUSIONS In this sample of veterans with no recent (24-month) history of depression or opioid analgesic use, the risk of development of depression increased as the duration of opioid analgesic exposure increased. The potential for depressogenic effect should be considered in risk-benefit discussions, and patients initiating opioid treatment should be monitored for development of depression.
Collapse
|
17
|
Abuse-related effects of µ-opioid analgesics in an assay of intracranial self-stimulation in rats: modulation by chronic morphine exposure. Behav Pharmacol 2013; 24:459-70. [PMID: 23881045 DOI: 10.1097/fbp.0b013e328364c0bd] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Intracranial self-stimulation (ICSS) is an operant procedure in which responding is maintained by electrical brain stimulation. Stimulation frequency can be varied rapidly to maintain a wide range of baseline response rates, and drugs' effects can be evaluated simultaneously on both low ICSS rates maintained by low stimulation frequencies and high ICSS rates maintained by high stimulation frequencies. ICSS 'facilitation' indicates drug-induced increases in low ICSS rates and is often considered an abuse-related effect, whereas ICSS 'depression' indicates decreases in high ICSS rates and may indicate abuse-limiting effects. This study examined the roles of µ-agonist efficacy and of previous µ-agonist exposure as determinants of µ-agonist effects on ICSS in rats with electrodes implanted into the medial forebrain bundle. The high-efficacy, intermediate-efficacy, and low-efficacy µ agonists methadone, fentanyl, and nalbuphine were tested during escalating regimens of morphine exposure (vehicle, 3.2, and 18 mg/kg/day). During vehicle treatment, methadone and fentanyl primarily depressed ICSS, whereas nalbuphine produced weak facilitation that was not dose dependent. Chronic morphine produced tolerance to ICSS depression and increased expression of ICSS facilitation. These results suggest that µ-agonist exposure increases the expression of abuse-related ICSS facilitation by µ agonists with a broad range of efficacies at µ receptors.
Collapse
|
18
|
Galaj E, Cruz I, Schachar J, Koziolek M, Ranaldi R. Differential effects on natural reward processing in rats after repeated heroin. Psychopharmacology (Berl) 2013; 229:125-32. [PMID: 23609768 DOI: 10.1007/s00213-013-3087-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 03/25/2013] [Indexed: 10/26/2022]
Abstract
RATIONALE Heroin users report reward deficits as well as reward enhancements (to drug stimuli). To better understand the causal relation between chronic heroin and alterations in natural reward processing, we used experimental techniques in animal models. METHODS Separate groups of rats were trained in several food reward paradigms: conditioned place preference (CPP), food-reinforced lever pressing under a progressive ratio schedule of reinforcement, free feeding, and lever pressing with conditioned reinforcement. After training, the rats were subjected to 10 daily heroin (2 mg/kg) or saline vehicle injections and tested at 3, 15, and 30 days post-treatment. RESULTS Repeated heroin treatment abolished the CPP and significantly reduced break points for food reward at 3, 15, and 30 days post-treatment. Repeated heroin did not affect free feeding. Finally, repeated heroin significantly enhanced responding for a food-based conditioned reinforcer. CONCLUSIONS Repeated heroin decreases the attractiveness of food-associated cues and reduces motivation to work for natural reward. However, it appears to enhance natural conditioned reward processes that involve the acquisition of novel responding. Thus, repeated heroin appears to produce differential effects on natural reward processing depending on the nature of the reward-directed behavior.
Collapse
Affiliation(s)
- Ewa Galaj
- The Graduate Center, City University of New York, New York, NY, USA
| | | | | | | | | |
Collapse
|
19
|
Graves SM, Persons AL, Riddle JL, Napier TC. The atypical antidepressant mirtazapine attenuates expression of morphine-induced place preference and motor sensitization. Brain Res 2012; 1472:45-53. [PMID: 22820297 DOI: 10.1016/j.brainres.2012.07.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 06/24/2012] [Accepted: 07/03/2012] [Indexed: 12/27/2022]
Abstract
Opioid abuse and dependence remains prevalent despite having multiple FDA-approved medications to help maintain abstinence. Mirtazapine is an atypical antidepressant receiving attention for substance abuse pharmacotherapy, and its action includes alterations in monoaminergic transmission. As monoamines are indirectly altered by opioids, the current investigation assessed the ability of mirtazapine to ameliorate morphine-induced behaviors. Conditioned place preference (CPP) is a behavioral assay wherein a rewarding drug is paired with a distinct environmental context resulting in reward-related salience of cues through learning-related neuronal plasticity. A second behavioral assay involved motor sensitization (MSn), wherein repeated administration results in an enhanced motoric response to an acute challenge, also reflecting neuronal plasticity. Attenuation of CPP and/or MSn provides two behavioral measures to suggest therapeutic potential for addiction therapy, and the present study evaluated the effectiveness of mirtazapine to reduce both behaviors. To do so, morphine-induced CPP was established using an eight day conditioning paradigm, and expression of CPP was tested on day 10 following a 24h or 30min mirtazapine pretreatment. To determine if mirtazapine altered the expression of MSn, on day 11, rats received a pretreatment of mirtazapine, followed 30min later by a challenge injection of morphine. Pretreatment with mirtazapine 24h prior to the CPP test had no effect on CPP expression. In contrast, a 30min pretreatment of mirtazapine attenuated the expression of both CPP and MSn. Collectively, these results indicate that mirtazapine may help to maintain abstinence in opioid dependent patients.
Collapse
Affiliation(s)
- Steven M Graves
- Department of Pharmacology, Center for Compulsive Behaviors and Addiction, Rush University Medical Center, Chicago, IL 60657, USA
| | | | | | | |
Collapse
|
20
|
Morrison J, Thornton V, Ranaldi R. Chronic intermittent heroin produces locomotor sensitization and long-lasting enhancement of conditioned reinforcement. Pharmacol Biochem Behav 2011; 99:475-9. [DOI: 10.1016/j.pbb.2011.04.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 04/08/2011] [Accepted: 04/25/2011] [Indexed: 10/18/2022]
|
21
|
Harty SC, Whaley JE, Halperin JM, Ranaldi R. Impulsive choice, as measured in a delay discounting paradigm, remains stable after chronic heroin administration. Pharmacol Biochem Behav 2011; 98:337-40. [PMID: 21315105 DOI: 10.1016/j.pbb.2011.02.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Revised: 01/06/2011] [Accepted: 02/02/2011] [Indexed: 10/18/2022]
Abstract
Heroin addicts display poorer impulse control than non-addicts, however it is not known if high impulsivity is a function of chronic heroin intake or a pre-disposing vulnerability for heroin addiction. Using animal models, relatively few studies have examined changes in impulsive choice as a function of chronic drug. The objective of this study was to measure alterations in impulsive choice through a delay discounting paradigm, as a function of chronic heroin administration. Animals were trained on a series of delay discounting sessions. Each session contained 5 blocks of trials. Blocks started with 2 forced, followed by 6 free choice trials. Pressing one lever resulted in the delivery of a small immediate (1 food pellet) reward and another lever in a large delayed (5 pellets) reward. Sessions consisted of the 3 ascending delay sequences in seconds. On the terminal sequence (0, 10, 20, 40, and 60s) animals exhibited a reversal of reward choice pattern of responding that allowed for the calculation of an indifference point (IP). After animals showed stable IPs they were treated with either heroin or saline for 12 days. Three days after the last injection animals were again placed in operant chambers and experienced the terminal delay discounting sequence at which time IPs were reassessed. Heroin-treated animals exhibited significant progressive increases in locomotor activity. Groups did not differ in IPs or performance across delay conditions during either before or after chronic treatment periods. These results indicate that chronic heroin intake does not impact later impulsive responding for natural (food) reward.
Collapse
Affiliation(s)
- Seth C Harty
- CUNY Graduate Center, Neuropsychology Doctoral Program, USA
| | | | | | | |
Collapse
|
22
|
Pitchers KK, Balfour ME, Lehman MN, Richtand NM, Yu L, Coolen LM. Neuroplasticity in the mesolimbic system induced by natural reward and subsequent reward abstinence. Biol Psychiatry 2010; 67:872-9. [PMID: 20015481 PMCID: PMC2854191 DOI: 10.1016/j.biopsych.2009.09.036] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2009] [Revised: 09/23/2009] [Accepted: 09/26/2009] [Indexed: 12/27/2022]
Abstract
BACKGROUND Natural reward and drugs of abuse converge on the mesolimbic system, where drugs of abuse induce neuronal alterations. Here, we tested plasticity in this system after natural reward and the subsequent impact on drug responses. METHODS Effects of sexual experience in male rats on behavioral sensitization and conditioned place preference associated with d-amphetamine (AMPH) and Golgi-impregnated dendrites and spines of nucleus accumbens (NAc) cells were determined. Moreover, the impact of abstinence from sexual behavior in experienced males on these parameters was tested. RESULTS First, repeated sexual behavior induced a sensitized locomotor response to AMPH compared with sexually naive control subjects observed 1, 7, and 28 days after last mating session. Second, sexually experienced animals formed a conditioned place preference for lower doses of AMPH than sexually naive males, indicative of enhanced reward value of AMPH. Finally, Golgi-Cox analysis demonstrated increased numbers of dendrites and spines in the NAc core and shell with sexual experience. The latter two alterations were dependent on a period of abstinence of 7-10 days. CONCLUSIONS Sexual experience induces functional and morphological alterations in the mesolimbic system similar to repeated exposure to psychostimulants. Moreover, abstinence from sexual behavior after repeated mating was essential for increased reward for drugs and dendritic arbors of NAc neurons, suggesting that the loss of sexual reward might also contribute to neuroplasticity of the mesolimbic system. These results suggest that some alterations in the mesolimbic system are common for natural and drug reward and might play a role in general reinforcement.
Collapse
Affiliation(s)
- Kyle K. Pitchers
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | | | - Michael N. Lehman
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Neil M. Richtand
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, Ohio
- Department of Psychiatry, University of Cincinnati, Cincinnati, Ohio
- Psychiatry Service, Cincinnati Veterans Affairs Medical Center
| | - Lei Yu
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, Ohio
| | - Lique M. Coolen
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada, N6A 5C1
- Department of Physiology & Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada, N6A 5C1
| |
Collapse
|
23
|
Kappa-opioid ligands in the study and treatment of mood disorders. Pharmacol Ther 2009; 123:334-43. [PMID: 19497337 DOI: 10.1016/j.pharmthera.2009.05.008] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Accepted: 05/11/2009] [Indexed: 11/23/2022]
Abstract
The biological basis of mood is not understood. Most research on mood and affective states has focused on the roles of brain systems containing monoamines (e.g., dopamine, norepinephrine, serotonin). However, it is becoming clear that endogenous opioid systems in the brain may also be involved in the regulation of mood. In this review, we focus on the potential utility of kappa-opioid receptor (KOR) ligands in the study and treatment of psychiatric disorders. Research from our group and others suggests that KOR antagonists might be useful for depression, KOR agonists might be useful for mania, and KOR partial agonists might be useful for mood stabilization. Currently available KOR agents have some unfavorable properties that might be addressed through medicinal chemistry. The development of KOR-selective agents with improved drug-like characteristics would facilitate preclinical and clinical studies designed to evaluate the possibility that KORs are a feasible target for new medications.
Collapse
|
24
|
Augmentation of morphine-induced sensitization but reduction in morphine tolerance and reward in delta-opioid receptor knockout mice. Neuropsychopharmacology 2009; 34:887-98. [PMID: 18704097 PMCID: PMC2639630 DOI: 10.1038/npp.2008.128] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Studies in experimental animals have shown that individuals exhibiting enhanced sensitivity to the locomotor-activating and rewarding properties of drugs of abuse are at increased risk for the development of compulsive drug-seeking behavior. The purpose of the present study was to assess the effect of constitutive deletion of delta-opioid receptors (DOPr) on the rewarding properties of morphine as well as on the development of sensitization and tolerance to the locomotor-activating effects of morphine. Locomotor activity testing revealed that mice lacking DOPr exhibit an augmentation of context-dependent sensitization following repeated, alternate injections of morphine (20 mg/kg; s.c.; 5 days). In contrast, the development of tolerance to the locomotor-activating effects of morphine following chronic morphine administration (morphine pellet: 25 mg: 3 days) is reduced relative to WT mice. The conditioned rewarding effects of morphine were reduced significantly in DOPrKO mice as compared to WT controls. Similar findings were obtained in response to pharmacological inactivation of DOPr in WT mice, indicating that observed effects are not due to developmental adaptations that occur as a consequence of constitutive deletion of DOPr. Together, these findings indicate that the endogenous DOPr system is recruited in response to both repeated and chronic morphine administration and that this recruitment serves an essential function in the development of tolerance, behavioral sensitization, and the conditioning of opiate reward. Importantly, they demonstrate that DOPr has a distinct role in the development of each of these drug-induced adaptations. The anti-rewarding and tolerance-reducing properties of DOPr antagonists may offer new opportunities for the treatment and prevention of opioid dependence as well as for the development of effective analgesics with reduced abuse liability.
Collapse
|
25
|
Delta-opioid receptor antagonists prevent sensitization to the conditioned rewarding effects of morphine. Biol Psychiatry 2009; 65:169-74. [PMID: 18950747 PMCID: PMC3832215 DOI: 10.1016/j.biopsych.2008.09.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Revised: 09/03/2008] [Accepted: 09/05/2008] [Indexed: 11/20/2022]
Abstract
BACKGROUND Functional interactions between mu- and delta-opioid receptors (MOPr and DOPr, respectively) are implicated in morphine tolerance and dependence. The contribution of DOPr to the conditioned rewarding effects of morphine and the enhanced conditioned response that occurs after repeated morphine administration is unknown. This issue was addressed with the conditioned place preference procedure (CPP). METHODS Rats received home cage injections of saline or morphine (5.0 mg/kg/day x 5 days) before conditioning. For sensitization studies, DOPr antagonists (DOPr1/2: naltrindole, DOPr2: naltriben, DOPr1: 7-benzylidenenaltrexone) were administered before morphine injections. Conditioning sessions (2 morphine; 2 saline) commenced 3 days later. To assess the influence of acute DOPr blockade on the conditioning of morphine reward in naïve animals, 3 morphine and 3 saline conditioning sessions were employed. Antagonists were administered before morphine conditioning sessions. RESULTS Morphine was ineffective as a conditioning stimulus after two conditioning sessions in naïve rats. However, doses > or = 3.0 mg/kg produced significant CPP in morphine pre-exposed rats, confirming that sensitization develops to the conditioned rewarding effects of morphine. In animals that received morphine pre-exposure with naltrindole or naltriben but not 7-benzylidenenaltrexone, sensitization was prevented. No attenuation of morphine CPP was observed in animals that received DOPr antagonists acutely, before conditioning sessions. CONCLUSIONS These data indicate a critical role of DOPr systems in mediating sensitization to the conditioned rewarding effects of morphine. The efficacy of naltrindole and naltriben in preventing the enhanced response to morphine suggest the specific involvement of DOPr2 in the sensitization process.
Collapse
|
26
|
Zarrindast MR, Hoghooghi V, Rezayof A. Inhibition of morphine-induced amnesia in morphine-sensitized mice: Involvement of dorsal hippocampal GABAergic receptors. Neuropharmacology 2008; 54:569-76. [DOI: 10.1016/j.neuropharm.2007.11.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2007] [Revised: 11/03/2007] [Accepted: 11/08/2007] [Indexed: 11/25/2022]
|
27
|
Zarrindast MR, Heidari-Darvishani A, Rezayof A, Fathi-Azarbaijani F, Jafari-Sabet M, Hajizadeh-Moghaddam A. Morphine-induced sensitization in mice: changes in locomotor activity by prior scheduled exposure to GABAA receptor agents. Behav Pharmacol 2007; 18:303-10. [PMID: 17551323 DOI: 10.1097/fbp.0b013e3282186baa] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This study investigated the effects of a gamma-amino-butyric acid type A (GABAA) receptor agonist and antagonist on morphine-induced locomotor sensitization in male albino mice. Subcutaneous administration to mice of a high dose of morphine (30 mg/kg), but not lower doses (5, 10 and 20 mg/kg) increased locomotion. The maximum locomotor activity was achieved during a 20-min measurement period. The locomotor response to a low dose of morphine (5 mg/kg, subcutaneously) given on day 9 was enhanced in mice pretreated with morphine (7.5, 15 and 30 mg/kg/day x 3 days), indicating that sensitization had developed. Three-day intracerebroventricular (i.c.v.) administration of the GABAA receptor agonist, muscimol (0.025, 0.05, 0.1 and 0.2 microg/mouse/day) significantly decreased both morphine-induced motor stimulation and locomotor sensitization. On the other hand, a 3-day pretreatment with the GABAA-receptor antagonist, bicuculline (0.25, 0.5 and 1 microg/mouse/day) reduced morphine (15 mg/kg)-induced locomotor sensitization. Repeated i.c.v. injections of a lower dose of bicuculline (0.25 microg/mouse/day x 3 days) by itself also decreased morphine-induced locomotion. Furthermore, repeated i.c.v. administration of bicuculline (0.25, 0.5 and 1 microg/mouse/day x 3 days) decreased the effect of i.c.v. injection of muscimol (0.1 microg/mouse/day x 3 days) on locomotor activity induced by morphine (5 mg/kg) in both control and sensitized mice. The magnitude of this response was, however, variable. The results indicate that GABAA receptors might be involved in the acquisition of morphine-induced sensitization.
Collapse
Affiliation(s)
- Mohammad-Reza Zarrindast
- Department of Pharmacology and Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran.
| | | | | | | | | | | |
Collapse
|
28
|
Kotlinska J, Pachuta A, Dylag T, Silberring J. The role of neuropeptide FF (NPFF) in the expression of sensitization to hyperlocomotor effect of morphine and ethanol. Neuropeptides 2007; 41:51-8. [PMID: 17107711 DOI: 10.1016/j.npep.2006.09.048] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2006] [Revised: 09/15/2006] [Accepted: 09/15/2006] [Indexed: 11/24/2022]
Abstract
Neuropeptide FF (NPFF) has been characterized as an endogenous anti-opioid peptide because its intraventricular injection (icv) reversed morphine- and stress-induced analgesia, and precipitates withdrawal syndrome in morphine-dependent rats. The role of NPFF in other aspects of drug dependence is unknown. Therefore, the aim of this study was to determine NPFF influence on the expression of sensitization to the morphine-induced hyperlocomotion. As the opioid system plays a role in ethanol effects, the influence of NPFF on the expression of sensitization to hyperlocomotor effect of ethanol was also investigated. Our study indicated that acute administration of NPFF (5, 10, 20nmol, icv) inhibited the expression of morphine-induced sensitization at doses of 10 (P<0.05) and 20nmol (P<0.01), and also inhibited ethanol-induced sensitization at a dose of 20nmol (P<0.01). Furthermore, NPFF inhibited the acute locomotor effect of morphine (10 and 20nmol) but not that of ethanol. NPFF, given alone, did not change the locomotor activity of mice and did not disturb motor coordination of animals in the rotarod test. In conclusion, our experiments indicated that NPFF attenuated the acute morphine locomotion and the expression of sensitization to locomotion. We anticipate that NPFF may be involved in both of these effects.
Collapse
Affiliation(s)
- Jolanta Kotlinska
- Department of Pharmacology and Pharmacodynamics, Medical University, Staszica Str. 4, 20-081 Lublin, Poland.
| | | | | | | |
Collapse
|
29
|
Gerra G, Fantoma A, Zaimovic A. Naltrexone and buprenorphine combination in the treatment of opioid dependence. J Psychopharmacol 2006; 20:806-14. [PMID: 16401652 DOI: 10.1177/0269881106060835] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Naltrexone treatment has demonstrated some advantages for special populations of heroin addicted individuals, but patients' compliance seems to be very poor, with a low adherence and low retention rate. Kappa-opioid system overdrive seems to contribute to opioid protracted abstinence syndrome, with dysphoria and psychosomatic symptoms during naltrexone treatment. The objective of this observational study was to determine the effectiveness of a functional k antagonist in improving naltrexone treatment outcome. A partial mu agonist/kappa antagonist (buprenorphine) and a mu antagonist (naltrexone) were combined during a 12 weeks protocol, theoretically leaving k antagonism as the major medication effect. Sixty patients were submitted to outpatient rapid detoxification utilizing buprenorphine and opioid antagonists. Starting on the fifth day, 30 patients (group A) received naltrexone alone. Alternatively, 30 patients (group B) received naltrexone (50mg oral dose) plus buprenorphine (4 mg sublingual) for the 12 weeks of the observational study. The endpoints of the study were: retention in treatment, negative urinalyses, changes in psychological symptoms (Symptom Checklist-90 Revised: SCL-90) and craving scores (visual analysis scale (VAS)). Thirty-four subjects (56.67%) completed the 12 weeks study. Twenty-one patients (35.0%) had all urine samples negative for opiates and cocaine. nine subjects (15.0%) had urine samples negative for cocaine and opiates for the last 4 weeks of the study. five subjects (8.3%) continued to use cocaine during the 12 weeks of the study. No significant change in pupillary diameter after buprenorphine administration was evidenced during clinical observations from baseline across the weekly measurements. Retention rates in group A (naltrexone) and group B (naltrexone + buprenorphine) at week 12 were respectively 40% (12 patients) and 73.33% (22 patients), with a significant difference in favour of group B (p= 0.018). Patients treated with naltrexone in combination with buprenorphine (B patients) showed a significantly lower rate of positive urines for morphine (4.45%) and cocaine metabolites (9.09%) than those treated with naltrexone alone (A) (25%, morphine; 33.33% cocaine) (p< 0.05; p< 0.05). Irritability, depression, tiredness, psychosomatic symptoms and craving scores decreased significantly less in Group A patients than in group B patients. The dysfunction of opioid system with kappa receptors hyper-activation provoked by heroin exposure, probably underlying dysphoric and psychosomatic symptoms during naltrexone treatment, seems to be counteracted, at least in part, by buprenorphine. The combination of buprenorphine and naltrexone may significantly improve the outcome of opioid antagonists treatment in terms of retention, negative urinalyses, and reduced dysphoria, mood symptoms and craving.
Collapse
Affiliation(s)
- G Gerra
- National Department on Drug Policy, Rome, Italy.
| | | | | |
Collapse
|
30
|
Metcalf MD, Coop A. Kappa opioid antagonists: past successes and future prospects. AAPS JOURNAL 2005. [PMID: 16353947 DOI: 10.1208/aapsj070371].] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Antagonists of the kappa opioid receptor were initially investigated as pharmacological tools that would reverse the effects of kappa opioid receptor agonists. In the years following the discovery of the first selective kappa opioid antagonists, much information about their chemistry and pharmacology has been elicited and their potential therapeutic uses have been investigated. The review presents the current chemistry, ligand-based structure activity relationships, and pharmacology of the known nonpeptidic selective kappa opioid receptor antagonists. This manuscript endeavors to provide the reader with a useful reference of the investigations made to define the structure-activity relationships and pharmacology of selective kappa opioid receptor antagonists and their potential uses as pharmacological tools and as therapeutic agents in the treatment of disease states.
Collapse
Affiliation(s)
- Matthew D Metcalf
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD 21201, USA
| | | |
Collapse
|
31
|
Metcalf MD, Coop A. Kappa opioid antagonists: past successes and future prospects. AAPS J 2005. [PMID: 16353947 DOI: 10.1208/aapsj070371]] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Antagonists of the kappa opioid receptor were initially investigated as pharmacological tools that would reverse the effects of kappa opioid receptor agonists. In the years following the discovery of the first selective kappa opioid antagonists, much information about their chemistry and pharmacology has been elicited and their potential therapeutic uses have been investigated. The review presents the current chemistry, ligand-based structure activity relationships, and pharmacology of the known nonpeptidic selective kappa opioid receptor antagonists. This manuscript endeavors to provide the reader with a useful reference of the investigations made to define the structure-activity relationships and pharmacology of selective kappa opioid receptor antagonists and their potential uses as pharmacological tools and as therapeutic agents in the treatment of disease states.
Collapse
Affiliation(s)
- Matthew D Metcalf
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD 21201, USA
| | | |
Collapse
|
32
|
Metcalf MD, Coop A. Kappa opioid antagonists: past successes and future prospects. AAPS JOURNAL 2005; 7:E704-22. [PMID: 16353947 PMCID: PMC2751273 DOI: 10.1208/aapsj070371] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Antagonists of the kappa opioid receptor were initially investigated as pharmacological tools that would reverse the effects of kappa opioid receptor agonists. In the years following the discovery of the first selective kappa opioid antagonists, much information about their chemistry and pharmacology has been elicited and their potential therapeutic uses have been investigated. The review presents the current chemistry, ligand-based structure activity relationships, and pharmacology of the known nonpeptidic selective kappa opioid receptor antagonists. This manuscript endeavors to provide the reader with a useful reference of the investigations made to define the structure-activity relationships and pharmacology of selective kappa opioid receptor antagonists and their potential uses as pharmacological tools and as therapeutic agents in the treatment of disease states.
Collapse
Affiliation(s)
- Matthew D. Metcalf
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, 21201 Baltimore, MD
| | - Andrew Coop
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, 21201 Baltimore, MD
| |
Collapse
|
33
|
Shi XD, Wang GB, Ma YY, Ren W, Luo F, Cui CL, Han JS. Repeated peripheral electrical stimulations suppress both morphine-induced CPP and reinstatement of extinguished CPP in rats: accelerated expression of PPE and PPD mRNA in NAc implicated. ACTA ACUST UNITED AC 2004; 130:124-33. [PMID: 15519683 DOI: 10.1016/j.molbrainres.2004.07.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2004] [Indexed: 11/17/2022]
Abstract
Previous studies have shown that peripheral electrical stimulation (PES) can suppress morphine-induced conditioned place preference (CPP) and the reinstatement of extinguished CPP in the rat. The present study was performed to elucidate if preproenkephalin (PPE) and preprodynorphin (PPD) mRNAs in the nucleus accumbens (NAc) play a role in this event. Rats were trained with morphine for 4 days to establish CPP paradigm. They were then given 15-min test once a day for eight consecutive days for extinction trial. Twenty-four hours after the 8th session of extinction trials, rats were given peripheral electrical stimulation (PES) at 2 or 100 Hz once a day for 3 days, then a morphine-priming injection at a dose of 1, 2, or 4 mg/kg to reinstate the extinguished CPP. At the end of the experiment, PPE and PPD mRNA levels in the nucleus acccumbens (NAc) were determined by the semiquantitative RT-PCR technique. The results showed that PES at 2- and 100-Hz administered 30 min a day for 3 days suppressed both the expression of morphine-induced CPP and the reinstatement of extinguished CPP. PES at 2 Hz increased preproenkephalin (PPE) mRNA levels, whereas PES of 100 Hz that of preprodynorphin (PPD) mRNA levels in the NAc. These findings suggest that enkephalin and dynorphin in NAc may play important roles in the mechanisms underlying the inhibitory effect of PES on the expression and reinstatement of morphine-induced CPP in rats.
Collapse
Affiliation(s)
- Xiang-Dang Shi
- Neuroscience Research Institute, Key laboratory for Neuroscience, Peking University, the Ministry of Education, 38 Xueyuan Road, Beijing 100083, PR China
| | | | | | | | | | | | | |
Collapse
|
34
|
Mague SD, Pliakas AM, Todtenkopf MS, Tomasiewicz HC, Zhang Y, Stevens WC, Jones RM, Portoghese PS, Carlezon WA. Antidepressant-like effects of kappa-opioid receptor antagonists in the forced swim test in rats. J Pharmacol Exp Ther 2003; 305:323-30. [PMID: 12649385 DOI: 10.1124/jpet.102.046433] [Citation(s) in RCA: 391] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We showed previously that cAMP response element-binding protein (CREB) within the nucleus accumbens (NAc) of rats regulates immobility in the forced swim test (FST), an assay used to study depression. Because CREB regulates expression of dynorphin (which acts at kappa-opioid receptors) in NAc neurons, these findings raised the possibility that kappa-receptors mediate immobility behaviors in the FST. Here, we report that i.c.v. administration of the kappa-antagonist nor-binaltorphimine dose dependently decreased immobility in the FST, suggesting that it has antidepressant-like effects. Implicating a specific effect at kappa-receptors, similar antidepressant-like effects were seen after treatment with either of two novel, structurally dissimilar kappa-antagonists: 5'-guanidinonaltrindole, which was effective after i.c.v. but not systemic treatment, and 5'-acetamidinoethylnaltrindole (ANTI), which was potent and effective after systemic treatment. The behavioral effects of the kappa-antagonists resembled those of tricyclic antidepressants (desipramine) and selective serotonin reuptake inhibitors (fluoxetine and citalopram). Conversely, systemic administration of the kappa-agonist [5alpha,7alpha,8beta]-N-methyl-N-[7-[1-pyrrolidinyl]-1-oxaspiro[4.5]dec8-yl]-benzenacetamide (U-69593) dose dependently increased immobility in the FST, consistent with prodepressant-like effects. The effects of the kappa-ligands in the FST were not correlated with nonspecific effects on locomotor activity. Furthermore, the most potent and effective kappa-antagonist (ANTI) did not affect the rewarding impact of lateral hypothalamic brain stimulation at a dose with strong antidepressant-like effects. These findings are consistent with the hypothesis that CREB-mediated induction of dynorphin in the NAc "triggers" immobility behavior in the FST. Furthermore, they raise the possibility that kappa-antagonists may have efficacy as antidepressants, but lack stimulant or reward-related effects.
Collapse
Affiliation(s)
- Stephen D Mague
- Behavioral Genetics Laboratory, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, Massachusetts 02478, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Ojanen S, Koistinen M, Bäckström P, Kankaanpää A, Tuomainen P, Hyytiä P, Kiianmaa K. Differential behavioural sensitization to intermittent morphine treatment in alcohol-preferring AA and alcohol-avoiding ANA rats: role of mesolimbic dopamine. Eur J Neurosci 2003; 17:1655-63. [PMID: 12752383 DOI: 10.1046/j.1460-9568.2003.02589.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Alcohol-preferring AA (Alko Alcohol) and alcohol-avoiding ANA (Alko Non-Alcohol) rats have well-documented differences in their voluntary ethanol consumption and brain opioidergic systems. The aim of the present study was to investigate whether these rat lines differ in their susceptibility to morphine-induced behavioural and neurochemical sensitization. The rats were given 15 injections of morphine (10 mg/kg, s.c.) or saline every other day. Locomotor activity and release of dopamine in the nucleus accumbens were monitored after a challenge with additional morphine injections (10 mg/kg) 1 and 5 weeks after withdrawal from the repeated treatment. Morphine increased locomotion more in the previously morphine-treated rats than in the saline-treated controls. Furthermore, AA rats were more sensitive to this effect of morphine than ANA rats. Accumbal morphine-induced dopamine release was significantly higher in the morphine-treated AA than ANA rats after the first challenge injection 1 week from withdrawal, but no differences were observed after the second challenge. The brain and plasma concentrations of morphine were similar among the lines suggesting that the differences in the effects of morphine cannot be explained in terms of differential pharmacokinetics of morphine in these lines. These data show that AA rats are more susceptible to morphine-induced behavioural sensitization than ANA rats. Furthermore, it suggests that mesolimbic dopamine has at best only a transient role in the expression of opioid-induced behavioural sensitization. The relationship between the mechanisms underlying the differential sensitivity of these rat lines to the effects of repeated morphine and voluntary ethanol drinking remains to be determined.
Collapse
Affiliation(s)
- Sami Ojanen
- Department of Mental Health and Alcohol Research, National Public Health Institute, POB 33, 00251 Helsinki, Finland
| | | | | | | | | | | | | |
Collapse
|
36
|
Inhibition of cAMP response element-binding protein or dynorphin in the nucleus accumbens produces an antidepressant-like effect. J Neurosci 2003. [PMID: 12486182 DOI: 10.1523/jneurosci.22-24-10883.2002] [Citation(s) in RCA: 218] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The cAMP response element-binding protein (CREB) is a critical integrator of neural plasticity that is responsive in a brain region-specific manner to a variety of environmental and pharmacological stimuli, including widely prescribed antidepressant medications. We developed inducible transgenic lines of mice that express either CREB or a dominant-negative mutant of CREB (mCREB) in forebrain regions and used these mice to determine the functional significance of this transcription factor in the learned helplessness paradigm, a behavioral model of depression. We also use a complementary viral-mediated gene transfer approach to directly test the effect of mCREB in the nucleus accumbens, a brain region important for motivation and reward. The results demonstrate that blockade of CREB by overexpression of mCREB in transgenic mice or by viral expression of mCREB in the nucleus accumbens produces an antidepressant-like effect, whereas overexpression of CREB in transgenic mice results in the opposite phenotype. In addition, mCREB expression was colocalized with and decreased the expression of prodynorphin in nucleus accumbens medium spiny neurons, and antagonism of dynorphin in the nucleus accumbens was sufficient to produce an antidepressant-like effect similar to that observed after blockade of CREB. Together, the results demonstrate that nucleus accumbens CREB-dynorphin influence behavior in the learned helplessness model and suggest that this signaling cascade may contribute to symptoms of depression.
Collapse
|
37
|
Altered responsiveness to cocaine and increased immobility in the forced swim test associated with elevated cAMP response element-binding protein expression in nucleus accumbens. J Neurosci 2001. [PMID: 11549750 DOI: 10.1523/jneurosci.21-18-07397.2001] [Citation(s) in RCA: 356] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Drugs of abuse regulate the transcription factor cAMP response element-binding protein (CREB) in striatal regions, including the nucleus accumbens (NAc). To explore how regulation of CREB in the NAc affects behavior, we used herpes simplex virus (HSV) vectors to elevate CREB expression in this region or to overexpress a dominant-negative mutant CREB (mCREB) that blocks CREB function. Rats treated with HSV-mCREB in place conditioning studies spent more time in environments associated with cocaine, indicating increased cocaine reward. Conversely, rats treated with HSV-CREB spent less time in cocaine-associated environments, indicating increased cocaine aversion. Studies in which drug-environment pairings were varied to coincide with either the early or late effects of cocaine suggest that CREB-associated place aversions reflect increased cocaine withdrawal. Because cocaine withdrawal can be accompanied by symptoms of depression, we examined how altered CREB function in the NAc affects behavior in the forced swim test (FST). Elevated CREB expression increased immobility in the FST, an effect that is opposite to that caused by standard antidepressants and is consistent with a link between CREB and dysphoria. Conversely, overexpression of mCREB decreased immobility, an effect similar to that caused by antidepressants. Moreover, the kappa opioid receptor antagonist nor-Binaltorphimine decreased immobility in HSV-CREB- and HSV-mCREB-treated rats, suggesting that CREB-mediated induction of dynorphin (an endogenous kappa receptor ligand) contributes to immobility behavior in the FST. Exposure to the FST itself dramatically increased CREB function in the NAc. These findings raise the possibility that CREB-mediated transcription within the NAc regulates dysphoric states.
Collapse
|
38
|
Mao L, Wang JQ. Differentially altered mGluR1 and mGluR5 mRNA expression in rat caudate nucleus and nucleus accumbens in the development and expression of behavioral sensitization to repeated amphetamine administration. Synapse 2001; 41:230-40. [PMID: 11418936 DOI: 10.1002/syn.1080] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Altered glutamatergic transmission in the striatum may be implicated in behavioral sensitization to repeated amphetamine (AMPH) administration. Quantitative in situ hybridization histochemistry was performed to define the effects of acute and chronic AMPH exposures on mRNA expression of Group I metabotropic glutamate receptors (mGluRs) in the striatum. Behavioral ratings indicated that the motor activity of rats was significantly higher after the final of five daily AMPH injections (4 mg/kg, i.p.) than that after the first of five daily AMPH, indicative of the development of behavioral sensitization. Moreover, the motor activity of rats treated with five daily AMPH was significantly greater than that of rats treated with five daily saline in response to a 2 mg/kg challenge dose of AMPH 7, 14, 28, and 60 days after the discontinuation of drug treatments, indicative of the persistent expression of behavioral sensitization. Three hours after acute administration of AMPH to naive rats, mGluR1 and mGluR5 mRNA expression in the dorsal (caudatoputamen) and ventral (nucleus accumbens) striatum showed no change as compared to acute saline injection. In rats that developed behavioral sensitization to repeated AMPH, mGluR1 levels in the dorsal and ventral striatum were increased by 53% and 43%, respectively, 3 h after the final AMPH treatment. However, this change did not persist during withdrawal since it was not observed 7, 14, and 28 days after the discontinuation of AMPH treatment. Conversely, mGluR5 levels were markedly reduced 3 h after the final of five daily AMPH treatments in the entire striatum of sensitized rats (34% and 77% of controls in the dorsal and ventral striatum, respectively). The reduction persisted at 7, 14, and 28 days of withdrawal. These results reveal a close linkage between striatal Group I mGluR gene expression and behavioral sensitization to AMPH. This may indicate functional implications of the two subtypes of Group I mGluRs in the regulation of behavioral sensitization to the dopamine stimulant.
Collapse
Affiliation(s)
- L Mao
- Division of Pharmacology, School of Pharmacy, University of Missouri-Kansas City, Kansas City, Missouri 64108, USA
| | | |
Collapse
|
39
|
Powell KR, Holtzman SG. Parametric evaluation of the development of sensitization to the effects of morphine on locomotor activity. Drug Alcohol Depend 2001; 62:83-90. [PMID: 11173171 DOI: 10.1016/s0376-8716(00)00167-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Animals repeatedly administered drugs of abuse often become more sensitive to their effects. It has been proposed that this behavioral sensitization may serve as a useful model for changes that may underlie the etiology and maintenance of drug-seeking behavior. This study was designed to determine systematically some of the conditions of drug exposure under which sensitization occurs to morphine-induced stimulation of locomotor activity. Groups of rats (n=8 per group) were exposed to a regimen of intermittent morphine or saline injections for 1--4 days and tested at later time points with morphine or saline. The amount of behavioral sensitization observed was related to the number of drug exposures, but not to the dose of morphine used during drug exposures. Sensitization to morphine persisted for as long as 3 months and was completely blocked when naltrexone was administered with the test dose of morphine after the final morphine exposure. Administration of naltrexone with morphine during the exposure regimen did not alter the development of behavioral sensitization. These results indicate a robust behavioral sensitization to morphine that appears to be influenced in an orderly manner within a narrow window of the drug exposure conditions.
Collapse
Affiliation(s)
- K R Powell
- Department of Pharmacology, Emory University School of Medicine, 1510 Clifton Road, Atlanta, GA 30322, USA.
| | | |
Collapse
|
40
|
Szumlinski KK, Maisonneuve IM, Glick SD. The potential anti-addictive agent, 18-methoxycoronaridine, blocks the sensitized locomotor and dopamine responses produced by repeated morphine treatment. Brain Res 2000; 864:13-23. [PMID: 10793182 DOI: 10.1016/s0006-8993(00)02069-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
18-Methoxycoronaridine (18-MC), a novel synthetic iboga congener, attenuates the reinforcing efficacy of morphine, disrupts some signs of morphine withdrawal in physically dependent rats and attenuates the dopamine response in the nucleus accumbens to acute morphine. The present study further investigated the interactions between 18-MC and morphine by examining the effects of 18-MC (40 mg/kg, i.p., 19 h earlier) on the expression of dopamine sensitization in the nucleus accumbens in response to morphine (20 mg/kg, i.p.) and on the dose-effect curves for morphine-induced locomotion (0-30 mg/kg, i.p.) in rats treated either acutely or repeatedly (five, once daily, injections of 20 mg/kg, i.p.) with morphine. Compared to vehicle pretreated controls, 18-MC increased the potency of morphine, shifting the dose-response curve to the left, in acute morphine treated rats; however, 18-MC did not alter the potency of morphine in rats treated repeatedly with morphine. Repeated morphine administration induced locomotor sensitization in approximately 50% of the rats tested; in vehicle pretreated rats, the morphine dose-response curve was shifted to the left in sensitized as compared to non-sensitized rats. In 18-MC pretreated rats, sensitized and non-sensitized rats responded similarly to morphine, revealing a blockade of sensitization by 18-MC. Consistent with this behavioural finding, 18-MC pretreatment completely abolished the sensitized dopamine response in the nucleus accumbens expressed by rats repeatedly treated with morphine. It is suggested that the potential anti-addictive efficacy of 18-MC might be related to an ability to restore normal functioning to a hypersensitive mesolimbic dopamine system produced by previous repeated morphine administration.
Collapse
Affiliation(s)
- K K Szumlinski
- Department of Pharmacology and Neuroscience (MC-136), Albany Medical College, 47 New Scotland Avenue, Albany, NY, USA.
| | | | | |
Collapse
|
41
|
Erdtmann-Vourliotis M, Mayer P, Riechert U, Höllt V. Prior experience of morphine application alters the c-fos response to MDMA ('ecstasy') and cocaine in the rat striatum. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2000; 77:55-64. [PMID: 10814832 DOI: 10.1016/s0169-328x(00)00040-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Repeated morphine application usually leads to the development of tolerance but under certain circumstances sensitization may arise simultaneously. This phenomenon becomes obvious in behavioral tests as increasing locomotor activity and increasing drug self-administration during a course of chronic morphine application. It was suggested recently that sensitization could contribute to addiction. The molecular mechanisms of sensitization may include the long lasting increase in neuronal responsiveness to morphine which was observed in defined brain areas after repeated morphine injections. In this work, we studied whether morphine-sensitized Wistar rats also display an enhanced neuronal activity in response to other drugs of abuse (so called co-sensitization). The substances to be tested were injected as single doses 4 weeks after completion of a 10-day morphine pretreatment. MDMA (3, 4-methylenedioxymethamphetamine, 6 mg/kg) as a single test dose yielded a c-fos response in a wide range of brain areas. In the caudate putamen, the expression pattern of c-fos was clearly altered if the rats had received repeated morphine application previously. In this case, the MDMA-induced c-fos expression was markedly confined to the centromedial, mesolimbic aspect of the striatum whereas it had a diffuse appearance in rats not exposed to the opiate earlier. Cocaine application (50 mg/kg) elicited an intense c-fos expression in the medial striatum if the animals were morphine-pretreated; it was virtually absent in drug-naive rats after the same cocaine dose. Ten mg/kg cocaine had a similar but weaker effect. No difference in the c-fos expression pattern between morphine and saline pretreated animals was observed in the case of a THC (Delta(9)-tetrahydrocannabinol, 25 mg/kg) or an LSD (lysergic acid diethylamide, 1 mg/kg) test application. These findings imply that morphine sensitizes the brain towards other addicting drugs. In consequence, morphine sensitization obviously does not solely reflect alterations in mu-opioid receptor signaling. Rather, it seems to reflect further rearrangements within the mesolimbic system.
Collapse
Affiliation(s)
- M Erdtmann-Vourliotis
- Institute for Pharmacology and Toxicology, Otto-von-Guericke Universität, Leipziger Str. 44, 39120, Magdeburg, Germany
| | | | | | | |
Collapse
|
42
|
Abstract
The effects of buprenorphine (0.0125-0.2 mg kg(-1)) on the locomotor activity of rats were determined 1-2 months after ceasing a chronic treatment with morphine (20 mg kg(-1)for 28 days). In control animals buprenorphine exhibited both depressive and stimulatory actions, as repeatedly described for morphine. In post-dependent animals buprenorphine showed a depressive effect similar to that observed in naive ones. On the contrary, a persistent sensitization to the excitatory effect was observed; the degree of cross-sensitization was similar to that of morphine itself (20 mg kg(-1)). The results are discussed in terms of persistent changes in the locomotor response to opioids, possibly correlated to both drug craving and relapse.
Collapse
Affiliation(s)
- M Bartoletti
- Department of Pharmacology, University of Bologna, Via Irnerio 48, Bologna, I-40126, Italy
| | | | | |
Collapse
|
43
|
Erdtmann-Vourliotis M, Mayer P, Linke R, Riechert U, Höllt V. Long-lasting sensitization towards morphine in motoric and limbic areas as determined by c-fos expression in rat brain. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1999; 72:1-16. [PMID: 10521594 DOI: 10.1016/s0169-328x(99)00184-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Chronic application of morphine leads to the development of tolerance towards several of its effects, e.g., analgesia or respiratory depression. Simultaneously, however, sensitization arises which becomes apparent in behavioral tests as increased locomotion or increased self-application. A human correlate for the latter may be the increasing craving for opioids in addicts. To identify brain areas involved in these long-lasting processes, we studied the expression of the transcription factor c-fos by in situ hybridization in rat brain as a marker for changes in gene expression after single or repeated morphine applications in the animals. The only c-fos signal that exceeded background after a single dose of morphine (50 mg/kg) was a diffuse expression in the lateral septum. In contrast, repeated dosage twice daily for 10 days and ascending from 10 to 50 mg/kg resulted in a sharply delineated morphine-induced c-fos synthesis in the dorsomedial and lateral striatum, lateral septum, medial mammillary nuclei, anterior thalamus and, in part masked by a high background due to injection stress, in the cingulate cortex. Most of these areas belong to the limbic system or are closely associated with it. The c-fos response was inducible by morphine in pretreated animals for up to 8 weeks after finishing the repeated application scheme. Retrograde tracing studies revealed that the dorsomedial part of the striatum, which was strongly labeled with the c-fos probe, received inputs from limbic as well as from motoric parts of the thalamus and cortex. Therefore, the sensitization of morphine-induced c-fos expression in parts of the striatum seems to correlate with the locomotor effects of repeated morphine application, whereas the observed sensitization in several limbic brain areas might reflect emotional phenomena like increased self-administration in rats or drug craving in humans.
Collapse
Affiliation(s)
- M Erdtmann-Vourliotis
- Institute for Pharmacology and Toxicology, Otto-von-Guericke-Universität Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
| | | | | | | | | |
Collapse
|
44
|
Abstract
Addictive drugs like cocaine, ethanol, and morphine activate signal transduction pathways that regulate brain gene expression. Such regulation is modulated by the presence of certain transcription factor proteins present in a given neuron. This article summarizes the effects of several addictive drugs on transcriptional processes contributing to the development of a drug-dependent state. The characterization of drug-induced changes in gene expression shows promise for improving our understanding of drug-addiction phenomena and cellular modes of cocaine, ethanol, and morphine action.
Collapse
Affiliation(s)
- G Torres
- Behavioral Neuroscience Program, Department of Psychology, State University of New York at Buffalo, 14260, USA.
| | | |
Collapse
|
45
|
Carlezon WA, Thome J, Olson VG, Lane-Ladd SB, Brodkin ES, Hiroi N, Duman RS, Neve RL, Nestler EJ. Regulation of cocaine reward by CREB. Science 1998; 282:2272-5. [PMID: 9856954 DOI: 10.1126/science.282.5397.2272] [Citation(s) in RCA: 576] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Cocaine regulates the transcription factor CREB (adenosine 3', 5'-monophosphate response element binding protein) in rat nucleus accumbens, a brain region that is important for addiction. Overexpression of CREB in this region decreases the rewarding effects of cocaine and makes low doses of the drug aversive. Conversely, overexpression of a dominant-negative mutant CREB increases the rewarding effects of cocaine. Altered transcription of dynorphin likely contributes to these effects: Its expression is increased by overexpression of CREB and decreased by overexpression of mutant CREB. Moreover, blockade of kappa opioid receptors (on which dynorphin acts) antagonizes the negative effect of CREB on cocaine reward. These results identify an intracellular cascade-culminating in gene expression-through which exposure to cocaine modifies subsequent responsiveness to the drug.
Collapse
Affiliation(s)
- W A Carlezon
- Division of Molecular Psychiatry, Center for Genes and Behavior, Yale University School of Medicine and Connecticut Mental Health Center, New Haven, CT 06508, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Erdtmann-Vourliotis M, Mayer P, Riechert U, Grecksch G, Höllt V. Identification of brain regions that are markedly activated by morphine in tolerant but not in naive rats. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1998; 61:51-61. [PMID: 9795131 DOI: 10.1016/s0169-328x(98)00197-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The induction of c-fos mRNA in rat brain due to morphine treatment was analyzed by in situ hybridization. A single dose of up to 100 mg/kg given to naive rats elicited only a weak c-fos expression. However, rats that were repeatedly pretreated with morphine displayed a marked c-fos induction in a few brain areas in response to morphine application. These brain areas essentially comprised the dorsal striatum, the shell of the nucleus accumbens, and some cortical areas. The c-fos signal was transient and not due to a residual withdrawal. Naloxone-precipitated withdrawal led to a more intense c-fos expression which also encompassed a greater range of brain areas. A similar but weaker pattern was observed in case of spontaneous withdrawal. A low morphine dose suppressed the c-fos expression nearly completely and was not sufficient to elicit the morphine-like expression pattern of c-fos. The brain areas which responded strongly to withdrawal included the piriform cortex, septal and hypothalamic nuclei and parts of the thalamus. Taken together, our data indicate that in certain circumscribed brain areas including the dorsal striatum and the shell of the nucleus accumbens, a sensitization towards morphine takes place at the molecular level. These areas responded to morphine with an elevated c-fos expression only when morphine was repeatedly given previously. Sensitization processes are thought to be important for opiate dependence, in particular for the increased craving for the drug. Furthermore, our data indicate that in case of repeated application signs of withdrawal appear after each morphine dose at the molecular level. Repeated events of withdrawal were also implicated in the establishment of a drug dependence state.
Collapse
Affiliation(s)
- M Erdtmann-Vourliotis
- Institute for Pharmacology and Toxicology, Otto-von-Guericke-Universität Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
| | | | | | | | | |
Collapse
|
47
|
Shippenberg TS, LeFevour A, Thompson AC. Sensitization to the conditioned rewarding effects of morphine and cocaine: differential effects of the kappa-opioid receptor agonist U69593. Eur J Pharmacol 1998; 345:27-34. [PMID: 9593590 DOI: 10.1016/s0014-2999(97)01614-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The ability of the kappa-opioid receptor agonist U69593 to attenuate the sensitization and cross-sensitization which develops to the conditioned rewarding effects of morphine and cocaine was examined using an unbiased place-preference conditioning procedure. The influence of U69593 treatment upon sensitization and cross-sensitization to cocaine was also assessed. Doses of morphine (1.0-5.0 mg kg(-1)) which failed to produce a conditioned response in drug-naive rats produced marked preferences for the drug-paired place in animals which had previously received once daily injections of morphine (5.0 mg kg(-1); s.c.) or cocaine (10.0 mg kg(-1); i.p.) for 5 days. Morphine-induced place preferences also occurred in animals which had received morphine in combination with U69593 (0.04-0.32 mg kg(-1); s.c.) on either days 3-5 or 1-5 of the morphine treatment regimen. In contrast, morphine failed to produce significant conditioning in animals which had received U69593 with cocaine for 5 days. Doses of cocaine (1.0-5.0 mg kg(-1)) which did not produce a conditioned response in naive rats produced preferences for the drug-paired place in animals which had received once daily injections of cocaine (10.0 mg kg(-1) day(-1) x 5 days; i.p.) or morphine (5.0 mg kg(-1) day(-1) x 5 days; s.c.). No enhancement of cocaine-induced conditioning occurred in animals which had received U69593 on days 3-5 or on days 1-5 of the five-day cocaine treatment. In animals, however, which had received U69593 with morphine for 5 days, an enhanced response to cocaine was still seen. These findings confirm that sensitization and cross-sensitization develop to the conditioned rewarding effects of cocaine and morphine. They also indicate that the ability of a kappa-opioid receptor agonist to prevent the development of these sensitized responses depends on the sensitizing agent employed. U69593 prevents sensitization and cross-sensitization induced by cocaine, but does not modify morphine-induced sensitization or the cross-sensitization which develops to cocaine after morphine administration.
Collapse
Affiliation(s)
- T S Shippenberg
- Integrative Neuroscience Unit, Behavioral Neuroscience Branch, NIDA Division of Intramural Research, Baltimore, MD 21224, USA.
| | | | | |
Collapse
|
48
|
Aguilar MA, Miñarro J, Simón VM. Dose-dependent impairing effects of morphine on avoidance acquisition and performance in male mice. Neurobiol Learn Mem 1998; 69:92-105. [PMID: 9619990 DOI: 10.1006/nlme.1997.3804] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The effects of morphine (6.3, 12.6, and 25.2 mg/kg) on active avoidance behavior of BALB/C mice are explored in three acquisition sessions and in two subsequent performance sessions. Morphine-treated animals showed an increase in avoidance acquisition with respect to control group without differences in performance. However, a dramatical, concomitant rise in the locomotor activity of the animals (increase in the number of crossings during the intertrial intervals) prompted us to transform the data employing a formula with which a measure of actual learning was obtained. Applying this formula, we have observed that morphine administration impairs, dose-dependently, acquisition and performance of avoidance. Thus, the impairing effects of morphine on avoidance could be masked by their stimulant effects on locomotor activity.
Collapse
Affiliation(s)
- M A Aguilar
- Area de Psicobiología, Facultad de Psicología, Universitat de Valencia, Spain.
| | | | | |
Collapse
|
49
|
Inderbitzin S, Lauber ME, Schlumpf M, Lichtensteiger W. Amphetamine-induced preprodynorphin mRNA expression and kappa-opioid receptor binding in basal ganglia of adult rats after prenatal exposure to diazepam. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 1997; 98:114-24. [PMID: 9027410 DOI: 10.1016/s0165-3806(96)00172-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
In order to obtain information on the functional state of basal ganglia following prenatal benzodiazepine exposure, preprodynorphin mRNA expression and kappa-opioid receptors were studied in offspring of timed-pregnant Long Evans rats treated with diazepam (1.25 mg/kg/day) on gestational days 14 to 20. Preprodynorphin mRNA was localised by in situ hybridization using a 33P-labeled oligonucleotide. Relative optical density (ROD) was quantified by image analysis in four quadrants of caudate putamen, in nucleus accumbens and olfactory tubercle of adult male rats. Six hours after functional challenge by injection of D-amphetamine (8 mg/kg s.c.), prenatally vehicle-exposed rats exhibited increased preprodynorphin mRNA (ROD) levels in caudate putamen (dorsolateral 187%, dorsomedial 150%, ventrolateral 153%, ventromedial 140% of control), nucleus accumbens (142%) and olfactory tubercle (213%). Prenatal diazepam exposure attenuated the effect of amphetamine in all regions; statistically significant differences between ROD levels of prenatally vehicle/adult amphetamine-treated and prenatally diazepam/adult amphetamine-treated groups were seen in ventrolateral caudate putamen, nucleus accumbens and olfactory tubercle. Baseline levels and topographical distribution of preprodynorphin mRNA remained unchanged. kappa-opioid receptor binding was analyzed in membrane from nucleus accumbens + olfactory tubercle, caudate putamen, and midbrain of male and female offspring using [3h]U69593. Bmax was reduced in nucleus accumbens + olfactory tubercle, but not in caudate putamen or midbrain of adult, prenatally diazepam-exposed male offspring, This effect was not yet seen at earlier postnatal stages (14 and 28 days), and was also absent in females. These data indicate that prenatal exposure to diazepam results in a delayed change in the functional state of dynorphin-containing neurons in several parts of the basal ganglia of adult male offspring. The decreased responsiveness to enhanced dopaminergic transmissions may impair the function of basal ganglia circuitry.
Collapse
Affiliation(s)
- S Inderbitzin
- Institute of Pharmacology, University of Zürich, Switzerland
| | | | | | | |
Collapse
|
50
|
Tjon GH, Voorn P, Vanderschuren LJ, de Vries TJ, Michiels NH, Jonker AJ, Klop H, Nestby P, Mulder AH, Schoffelmeer AN. Delayed occurrence of enhanced striatal preprodynorphin gene expression in behaviorally sensitized rats: differential long-term effects of intermittent and chronic morphine administration. Neuroscience 1997; 76:167-76. [PMID: 8971769 DOI: 10.1016/s0306-4522(96)00363-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Protracted changes in basal "steady-state" opioid peptide gene expression in the brain may represent adaptations underlying the behavioral effects of drugs of abuse, observed long after drug exposure. Here, we have studied the long-term effects of two distinct regimens of morphine administration ("intermittent" vs "chronic" morphine treatment) on behavioral sensitization and "steady-state" striatal preprodynorphin and preproenkephalin gene expression in rats. Opioid peptide gene expression was investigated using in situ hybridization at three rostrocaudal levels (rostral, intermediate and caudal) of the caudate-putamen and the nucleus accumbens. Behavioral studies showed that the intermittent morphine treatment resulted in a significantly greater enhancement of morphine-induced locomotion than the chronic morphine treatment three weeks after cessation of opiate exposure. The intermittent morphine treatment resulted in an initial decrease of preprodynorphin gene expression of about 5-10% in the caudate-putamen and the nucleus accumbens at the rostral and intermediate levels one day after the last morphine administration. In contrast, a protracted increase of preprodynorphin gene expression of about 20% throughout the caudate-putamen and of about 6% in intermediate sections of the nucleus accumbens was observed 21 days after cessation of intermittent morphine treatment. Although the chronic morphine treatment induced a decrease of preprodynorphin messenger RNA levels one day after the last administration, no significant changes were observed three weeks after cessation of chronic morphine treatment. No long-term changes were observed in preproenkephalin gene expression after either morphine treatment. Since the intermittent morphine administration induced long-term behavioral sensitization much more effectively than the chronic morphine treatment, we tentatively suggest that the protracted increase of preprodynorphin gene expression may play a facilitative role in the long-term character of opiate-induced behavioral sensitization.
Collapse
Affiliation(s)
- G H Tjon
- Research Institute Neurosciences Vrije Universiteit, Faculty of Medicine, Department of Pharmacology, Free University, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|