1
|
Lopes CR, Lourenço VS, Tomé ÂR, Cunha RA, Canas PM. Use of knockout mice to explore CNS effects of adenosine. Biochem Pharmacol 2020; 187:114367. [PMID: 33333075 DOI: 10.1016/j.bcp.2020.114367] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/09/2020] [Accepted: 12/09/2020] [Indexed: 12/11/2022]
Abstract
The initial exploration using pharmacological tools of the role of adenosine receptors in the brain, concluded that adenosine released as such acted on A1R to inhibit excitability and glutamate release from principal neurons throughout the brain and that adenosine A2A receptors (A2AR) were striatal-'specific' receptors controlling dopamine D2R. This indicted A1R as potential controllers of neurodegeneration and A2AR of psychiatric conditions. Global knockout of these two receptors questioned the key role of A1R and instead identified extra-striatal A2AR as robust controllers of neurodegeneration. Furthermore, transgenic lines with altered metabolic sources of adenosine revealed a coupling of ATP-derived adenosine to activate A2AR and a role of A1R as a hurdle to initiate neurodegeneration. Additionally, cell-selective knockout of A2AR unveiled the different roles of A2AR in different cell types (neurons/astrocytes) in different portions of the striatal circuits (dorsal versus lateral) and in different brain areas (hippocampus/striatum). Finally, a new transgenic mouse line with deletion of all adenosine receptors seems to indicate a major allostatic rather than homeostatic role of adenosine and may allow isolating P2R-mediated responses to unravel their role in the brain, a goal close to heart of Geoffrey Burnstock, to whom we affectionately dedicate this review.
Collapse
Affiliation(s)
- Cátia R Lopes
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Vanessa S Lourenço
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Ângelo R Tomé
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3000-456 Coimbra, Portugal
| | - Rodrigo A Cunha
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal.
| | - Paula M Canas
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| |
Collapse
|
2
|
Loi B, Sahai MA, De Luca MA, Shiref H, Opacka-Juffry J. The Role of Dopamine in the Stimulant Characteristics of Novel Psychoactive Substances (NPS)-Neurobiological and Computational Assessment Using the Case of Desoxypipradrol (2-DPMP). Front Pharmacol 2020; 11:806. [PMID: 32670057 PMCID: PMC7289955 DOI: 10.3389/fphar.2020.00806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 05/18/2020] [Indexed: 11/13/2022] Open
Abstract
Stimulant drugs, including novel psychoactive substances (NPS, formerly “legal highs”) have addictive potential which their users may not realize. Stimulants increase extracellular dopamine levels in the brain, including the reward and addiction pathways, through interacting with dopamine transporter (DAT). This work aimed to assess the molecular and atomistic mechanisms of stimulant NPS actions at DAT, which translate into biological outcomes such as dopamine release in the brain’s reward pathway. We applied combined in vitro, in vivo, and in silico methods and selected 2-diphenylmethylpiperidine (2-DPMP) as an example of stimulant NPS for this study. We measured in vitro binding of 2-DPMP to rat striatum and accumbens DAT by means of quantitative autoradiography with a selective DAT-radioligand [125I]RTI-121. We evaluated the effects of intravenously administered 2-DPMP on extracellular dopamine in the accumbens-shell and striatum using in vivo microdialysis in freely moving rats. We used dynamic modeling to investigate the interactions of 2-DPMP within DAT, in comparison with cocaine and amphetamine. 2-DPMP potently displaced the radioligand in the accumbens and striatum showing dose-dependence from 0.3 to 30 μM. IC50 values were: 5.65 × 10-7M for accumbens shell and 6.21 × 10-7M for dorsal striatum. Dose-dependent responses were also observed in accumbens-shell and striatum in vivo, with significant increases in extracellular dopamine levels. Molecular dynamics simulations identified contrasting conformational changes of DAT for inhibitors (cocaine) and releasers (amphetamine). 2-DPMP led to molecular rearrangements toward an outward-facing DAT conformation that suggested a cocaine-type effect. The present combination of molecular modeling with experimental neurobiological procedures allows for extensive characterization of the mechanisms of drug actions at DAT as the main molecular target of stimulants, and provides an insight into the role of dopamine in the molecular and neurobiological mechanisms of brain responses to stimulant NPS that have addictive potential. Such knowledge reveals the risk of addiction related to NPS use. The research presented here can be adapted for other psychostimulants that act at their membrane protein targets.
Collapse
Affiliation(s)
- Barbara Loi
- Psychopharmacology, Drug Misuse and Novel Psychoactive Substances Research Unit, Department of Pharmacy, Postgraduate Medicine and Pharmacology, University of Hertfordshire, Hatfield, United Kingdom.,Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Michelle A Sahai
- Department of Life Sciences, University of Roehampton, London, United Kingdom
| | | | - Hana Shiref
- Department of Life Sciences, University of Roehampton, London, United Kingdom
| | | |
Collapse
|
3
|
Adenosine A 2AReceptors in Substance Use Disorders: A Focus on Cocaine. Cells 2020; 9:cells9061372. [PMID: 32492952 PMCID: PMC7348840 DOI: 10.3390/cells9061372] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 05/28/2020] [Accepted: 05/28/2020] [Indexed: 12/31/2022] Open
Abstract
Several psychoactive drugs can evoke substance use disorders (SUD) in humans and animals, and these include psychostimulants, opioids, cannabinoids (CB), nicotine, and alcohol. The etiology, mechanistic processes, and the therapeutic options to deal with SUD are not well understood. The common feature of all abused drugs is that they increase dopamine (DA) neurotransmission within the mesocorticolimbic circuitry of the brain followed by the activation of DA receptors. D2 receptors were proposed as important molecular targets for SUD. The findings showed that D2 receptors formed heteromeric complexes with other GPCRs, which forced the addiction research area in new directions. In this review, we updated the view on the brain D2 receptor complexes with adenosine (A)2A receptors (A2AR) and discussed the role of A2AR in different aspects of addiction phenotypes in laboratory animal procedures that permit the highly complex syndrome of human drug addiction. We presented the current knowledge on the neurochemical in vivo and ex vivo mechanisms related to cocaine use disorder (CUD) and discussed future research directions for A2AR heteromeric complexes in SUD.
Collapse
|
4
|
Che X, Liu P, Wu C, Song W, An N, Yu L, Bai Y, Xing Z, Cai J, Wang X, Yang J. Potential role of the ecto-5'-nucleotidase in morphine-induced uridine release and neurobehavioral changes. Neuropharmacology 2018; 141:1-10. [PMID: 30071207 DOI: 10.1016/j.neuropharm.2018.07.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 07/24/2018] [Accepted: 07/29/2018] [Indexed: 12/21/2022]
Abstract
There is growing evidence that uridine may act as an endogenous neuromodulator with a potential signaling role in the central nervous system in addition to its function in pyrimidine metabolism. We previously found that acute morphine treatment significantly increased uridine release in the dorsal striatum of mice, indicating that uridine may contribute to morphine-induced neurobehavioral changes. In the present study, we analyzed the mechanism involved in morphine-induced uridine release and the role of uridine in morphine-induced neurobehavioral changes. Uridine release in the dorsal striatum of mice was assessed by in vivo microdialysis coupled with high performance liquid chromatography (HPLC) after morphine treatment. Western blotting and immunofluorescence were used to evaluate the expression of uridine-related proteins. Morphine-induced neurobehavioral changes were assessed by locomotor activity, behavioral sensitization and conditioned place preference (CPP) test. The expression of NT5E, an extracellular enzyme involved in formation of nucleosides, including uridine, was specifically knocked down in the dorsal striatum of mice using adeno-associated virus (AAV)-mediated short hairpin RNA (shRNA). The results indicated that both acute and chronic morphine administration significantly increased uridine release in the dorsal striatum, and this was associated with upregulation of NT5E but not other uridine-related proteins. Inhibition of NT5E with APCP or shRNA markedly inhibited morphine-induced uridine release in the dorsal striatum and related neurobehavioral changes, including hyperlocomotor activity, behavioral sensitization and CPP. Our data give a better understanding of the contribution of NT5E to morphine-induced uridine release and neurobehavioral changes, and identify NT5E as a potential target for treating morphine abuse.
Collapse
Affiliation(s)
- Xiaohang Che
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China
| | - Ping Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China
| | - Chunfu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China
| | - Wu Song
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China
| | - Nina An
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China
| | - Lisha Yu
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China
| | - Yijun Bai
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China
| | - Zheng Xing
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China
| | - Jialing Cai
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China
| | - Xiaomin Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China
| | - Jingyu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China.
| |
Collapse
|
5
|
Furlong TM, Supit AS, Corbit LH, Killcross S, Balleine BW. Pulling habits out of rats: adenosine 2A receptor antagonism in dorsomedial striatum rescues meth-amphetamine-induced deficits in goal-directed action. Addict Biol 2017; 22:172-183. [PMID: 26515740 DOI: 10.1111/adb.12316] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 08/06/2015] [Accepted: 09/02/2015] [Indexed: 11/29/2022]
Abstract
Addiction is characterized by a persistent loss of behavioral control resulting in insensitivity to negative feedback and abnormal decision-making. Here, we investigated the influence of methamphetamine (METH)-paired contextual cues on decision-making in rats. Choice between goal-directed actions was sensitive to outcome devaluation in a saline-paired context but was impaired in the METH-paired context, a deficit that was also found when negative feedback was provided. Reductions in c-Fos-related immunoreactivity were found in dorsomedial striatum (DMS) but not dorsolateral striatum after exposure to the METH context suggesting this effect reflected a loss specifically in goal-directed control in the METH context. This reduction in c-Fos was localized to non-enkephalin-expressing neurons in the DMS, likely dopamine D1-expressing direct pathway neurons, suggesting a relative change in control by the D1-direct versus D2-indirect pathways originating in the DMS may have been induced by METH-context exposure. To test this suggestion, we infused the adenosine 2A receptor antagonist ZM241385 into the DMS prior to test to reduce activity in D2 neurons relative to D1 neurons in the hope of reducing the inhibitory output from this region of the striatum. We found that this treatment fully restored sensitivity to negative feedback in a test conducted in the METH-paired context. These results suggest that drug exposure alters decision-making by downregulation of the circuitry mediating goal-directed action, an effect that can be ameliorated by acute A2A receptor inhibition in this circuit.
Collapse
Affiliation(s)
- Teri M. Furlong
- Brain & Mind Research Institute; University of Sydney; Australia
| | - Alva S.A. Supit
- Brain & Mind Research Institute; University of Sydney; Australia
| | | | | | | |
Collapse
|
6
|
Chesworth R, Brown RM, Kim JH, Ledent C, Lawrence AJ. Adenosine 2A receptors modulate reward behaviours for methamphetamine. Addict Biol 2016; 21:407-21. [PMID: 25612195 DOI: 10.1111/adb.12225] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Addiction to methamphetamine (METH) is a global health problem for which there are no approved pharmacotherapies. The adenosine 2A (A2 A ) receptor presents a potential therapeutic target for METH abuse due to its modulatory effects on striatal dopamine and glutamate transmission. Notably, A2 A receptor signalling has been implicated in the rewarding effects of alcohol, cocaine and opiates; yet, the role of this receptor in METH consumption and seeking is essentially unknown. Therefore, the current study used A2 A knockout (KO) mice to assess the role of A2 A in behaviours relevant to METH addiction. METH conditioned place preference was absent in A2 A KO mice compared with wild-type (WT) littermates. Repeated METH treatment produced locomotor sensitization in both genotypes; however, sensitization was attenuated in A2 A KO mice in a dose-related manner. METH intravenous self-administration was intact in A2 A KO mice over a range of doses and schedules of reinforcement. However, the motivation to self-administer was reduced in A2 A KO mice. Regression analysis further supported the observation that the motivation to self-administer METH was reduced in A2 A KO mice even when self-administration was similar to WT mice. Sucrose self-administration was also reduced in A2 A KO mice but only at higher schedules of reinforcement. Collectively, these data suggest that A2 A signalling is critically required to integrate rewarding and motivational properties of both METH and natural rewards.
Collapse
Affiliation(s)
- Rose Chesworth
- Behavioural Neuroscience Division; Florey Institute of Neuroscience and Mental Health; Australia
- Florey Department of Neuroscience and Mental Health; University of Melbourne; Australia
| | - Robyn M. Brown
- Behavioural Neuroscience Division; Florey Institute of Neuroscience and Mental Health; Australia
- Florey Department of Neuroscience and Mental Health; University of Melbourne; Australia
- Department of Neurosciences; Medical University of South Carolina; Charleston SC USA
| | - Jee Hyun Kim
- Behavioural Neuroscience Division; Florey Institute of Neuroscience and Mental Health; Australia
- Florey Department of Neuroscience and Mental Health; University of Melbourne; Australia
| | - Catherine Ledent
- Institut de Recherche Interdisciplinaire; Faculté de Médecine; Université de Bruxelles; Belgium
| | - Andrew J. Lawrence
- Behavioural Neuroscience Division; Florey Institute of Neuroscience and Mental Health; Australia
- Florey Department of Neuroscience and Mental Health; University of Melbourne; Australia
| |
Collapse
|
7
|
Simola N, Costa G, Morelli M. Activation of adenosine A₂A receptors suppresses the emission of pro-social and drug-stimulated 50-kHz ultrasonic vocalizations in rats: possible relevance to reward and motivation. Psychopharmacology (Berl) 2016; 233:507-19. [PMID: 26564233 DOI: 10.1007/s00213-015-4130-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 10/18/2015] [Indexed: 12/31/2022]
Abstract
RATIONALE Rats emit 50-kHz ultrasonic vocalizations (USVs) in response to pleasurable stimuli, and these USVs are considered a tool for investigating reward and motivation. OBJECTIVES This study aimed to clarify how activity of adenosine A2A receptors, which modulate reward and motivation, influences 50-kHz USV emission in rats. METHODS Rats received one of the following treatments in a test cage: (1) acute administration of the A2A receptor agonist CGS 21680 (0.05-0.2 mg/kg, i.p.) during social interactions; (2) long-term amphetamine (1 or 2 mg/kg, i.p.) or morphine (7.5 mg/kg, s.c.) administration on alternate days, alone or with CGS 21680, followed after 7 days of discontinuation by test cage re-exposure, to assess drug-conditioning effects, and thereafter drug challenge; (3) acute administration of the D1/D2 receptor agonist apomorphine (4 mg/kg, i.p.), alone or with CGS 21680; and (4) long-term administration of the non-selective A1/A2A receptor antagonist caffeine (15 mg/kg, i.p.), on alternate days. USVs and locomotor activity were evaluated throughout the treatments. RESULTS CGS 21680 attenuated 50-kHz USV emission stimulated by social interactions, amphetamine, apomorphine, and morphine, and rats administered CGS 21680 with amphetamine or morphine emitted fewer conditioned 50-kHz USVs upon test cage re-exposure, compared with rats administered amphetamine or morphine alone. Moreover, CGS 21680 administration prevented long-term changes in locomotor activity in amphetamine- and morphine-treated rats. Finally, caffeine had no effect on 50-kHz USVs. CONCLUSIONS These results indicate that activation of A2A receptors attenuates 50-kHz USV emission in rats and further elucidate how these receptors modulate the motivational properties of natural and pharmacological stimuli.
Collapse
Affiliation(s)
- Nicola Simola
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale, 72, 09124, Cagliari, Italy.
| | - Giulia Costa
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale, 72, 09124, Cagliari, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale, 72, 09124, Cagliari, Italy.,CNR, National Research Council of Italy, Neuroscience Institute, Cagliari, Italy
| |
Collapse
|
8
|
Karami M, Rahimpour M, Karimi S, Sahraei H. Nitric oxide in central amygdala potentiates expression of conditioned withdrawal induced by morphine. Indian J Pharmacol 2014; 46:57-62. [PMID: 24550586 PMCID: PMC3912809 DOI: 10.4103/0253-7613.125169] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 09/04/2013] [Accepted: 11/21/2013] [Indexed: 12/14/2022] Open
Abstract
Objective: The aim of this study was to evaluate if nitric oxide (NO) in the central amygdala (CeA) is involved in the expression of withdrawal aspects induced by morphine. Materials and Methods: Male Wistar rats (weighing 200-250 g) were bilaterally cannulated in the CeA and conditioned to morphine using an unbiased paradigm. Morphine (2.5-10 mg/kg) was subcutaneously injected once a day throughout the conditioning phase of the procedure. This phase also included 3-saline paired sessions. Naloxone (0.1-0.4 mg/kg, intraperitoneally [i.p.]), an antagonist of opioid receptors, was administered i.p. 10 min prior to testing of morphine-induced withdrawal features. The NO precursor, L-arginine (0.3-3 μg/rat) was intra-CeA injected prior to testing of naloxone response. To evaluate the involvement of NO system an inhibitor of NO synthase (NOS), NG-nitro-L-arginine methyl ester (L-NAME) (0.3-3 μg/rat), was injected ahead of L-arginine. Control group received saline solely instead of drug. As a complementary study, the activation of NOS was studied by nicotinamide adenine dinucleotide phosphate-diaphorase (NADPH-d). Results: Morphine induced a significant increase in wet dog shaking and grooming behaviors compared with controls. Injection of naloxone pre-testing of morphine response significantly reversed the response to morphine. However, pre-microinjection of L-arginine intra-CeA recovered the response to morphine. Injection of L-NAME intra-CeA ahead of L-arginine though had no effect behaviorally, but, inhibited the NOS which has been evidenced by NADPH-d. Conclusion: The present study shows that NO in the CeA potentiates the expression of conditioned withdrawal induced by morphine paired with naloxone.
Collapse
Affiliation(s)
- Manizheh Karami
- Neurophysiology Research Center, Shahed University, Tehran, Iran ; Department of Biology, Faculty of Basic Sciences, Shahed University, Tehran, Iran
| | - Mahnaz Rahimpour
- Department of Biology, Faculty of Basic Sciences, Shahed University, Tehran, Iran
| | - Sara Karimi
- Department of Biology, Faculty of Basic Sciences, Shahed University, Tehran, Iran
| | - Hedayat Sahraei
- Department of Physiology, Baghiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Clark PJ, Ghasem PR, Mika A, Day HE, Herrera JJ, Greenwood BN, Fleshner M. Wheel running alters patterns of uncontrollable stress-induced cfos mRNA expression in rat dorsal striatum direct and indirect pathways: A possible role for plasticity in adenosine receptors. Behav Brain Res 2014; 272:252-63. [PMID: 25017571 DOI: 10.1016/j.bbr.2014.07.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/22/2014] [Accepted: 07/03/2014] [Indexed: 01/22/2023]
Abstract
Emerging evidence indicates that adenosine is a major regulator of striatum activity, in part, through the antagonistic modulation of dopaminergic function. Exercise can influence adenosine and dopamine activity, which may subsequently promote plasticity in striatum adenosine and dopamine systems. Such changes could alter activity of medium spiny neurons and impact striatum function. The purpose of this study was twofold. The first was to characterize the effect of long-term wheel running on adenosine 1 (A1R), adenosine 2A (A2AR), dopamine 1 (D1R), and dopamine 2 (D2R) receptor mRNA expression in adult rat dorsal and ventral striatum structures using in situ hybridization. The second was to determine if changes to adenosine and dopamine receptor mRNA from running are associated with altered cfos mRNA induction in dynorphin- (direct pathway) and enkephalin- (indirect pathway) expressing neurons of the dorsal striatum following stress exposure. We report that chronic running, as well as acute uncontrollable stress, reduced A1R and A2AR mRNA levels in the dorsal and ventral striatum. Running also modestly elevated D2R mRNA levels in striatum regions. Finally, stress-induced cfos was potentiated in dynorphin and attenuated in enkephalin expressing neurons of running rats. These data suggest striatum adenosine and dopamine systems are targets for neuroplasticity from exercise, which may contribute to changes in direct and indirect pathway activity. These findings may have implications for striatum mediated motor and cognitive processes, as well as exercise facilitated stress-resistance.
Collapse
Affiliation(s)
- Peter J Clark
- Integrative Physiology, University of Colorado Boulder, 354 UCB, Boulder, CO 80309, United States.
| | - Parsa R Ghasem
- Integrative Physiology, University of Colorado Boulder, 354 UCB, Boulder, CO 80309, United States
| | - Agnieszka Mika
- Integrative Physiology, University of Colorado Boulder, 354 UCB, Boulder, CO 80309, United States
| | - Heidi E Day
- Integrative Physiology, University of Colorado Boulder, 354 UCB, Boulder, CO 80309, United States; Department of Psychology & Neuroscience, University of Colorado Boulder, Muenzinger D244, 345 UCB, Boulder, CO 80309, United States
| | - Jonathan J Herrera
- Integrative Physiology, University of Colorado Boulder, 354 UCB, Boulder, CO 80309, United States
| | - Benjamin N Greenwood
- Integrative Physiology, University of Colorado Boulder, 354 UCB, Boulder, CO 80309, United States
| | - Monika Fleshner
- Integrative Physiology, University of Colorado Boulder, 354 UCB, Boulder, CO 80309, United States
| |
Collapse
|
10
|
Wu M, Sahbaie P, Zheng M, Lobato R, Boison D, Clark JD, Peltz G. Opiate-induced changes in brain adenosine levels and narcotic drug responses. Neuroscience 2012; 228:235-42. [PMID: 23098802 DOI: 10.1016/j.neuroscience.2012.10.031] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Revised: 10/10/2012] [Accepted: 10/13/2012] [Indexed: 10/27/2022]
Abstract
We have very little information about the metabolomic changes that mediate neurobehavioral responses, including addiction. It was possible that opioid-induced metabolomic changes in brain could mediate some of the pharmacodynamic effects of opioids. To investigate this, opiate-induced brain metabolomic responses were profiled using a semi-targeted method in C57BL/6 and 129Sv1 mice, which exhibit extreme differences in their tendency to become opiate dependent. Escalating morphine doses (10-40 mg/kg) administered over a 4-day period selectively induced a twofold decrease (p<0.00005) in adenosine abundance in the brainstem of C57BL/6 mice, which exhibited symptoms of narcotic drug dependence; but did not decrease adenosine abundance in 129Sv1 mice, which do not exhibit symptoms of dependence. Based on this finding, the effect of adenosine on dependence was investigated in genetically engineered mice with alterations in adenosine tone in the brain and in pharmacologic experiments. Morphine withdrawal behaviors were significantly diminished (p<0.0004) in genetically engineered mice with reduced adenosine tone in the brainstem, and by treatment with an adenosine receptor(1) (A(1)) agonist (2-chloro-N6-cyclopentyladenosine, 0.5mg/kg) or an A(2a) receptor (A(2a)) antagonist (SCH 58261, 1mg/kg). These results indicate that adenosine homeostasis plays a crucial role in narcotic drug responses. Opiate-induced changes in brain adenosine levels may explain many important neurobehavioral features associated with opiate addiction and withdrawal.
Collapse
Affiliation(s)
- M Wu
- Department of Anesthesia, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | | | |
Collapse
|
11
|
Varani AP, Moutinho LM, Bettler B, Balerio GN. Acute behavioural responses to nicotine and nicotine withdrawal syndrome are modified in GABA(B1) knockout mice. Neuropharmacology 2012; 63:863-72. [PMID: 22727822 DOI: 10.1016/j.neuropharm.2012.06.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 05/18/2012] [Accepted: 06/05/2012] [Indexed: 02/01/2023]
Abstract
Nicotine is the main active component of tobacco, and has both acute and chronic pharmacological effects that can contribute to its abuse potential in humans. The aim of the present study was to evaluate a possible role of GABA(B) receptors in acute and chronic responses to nicotine administration, by comparing GABA(B1) knockout mice and their wild-type littermates. In wild-type mice, acute nicotine administration (0.5, 1, 3 and 6 mg/kg, sc) dose-dependently decreased locomotor activity, and induced antinociceptive responses in the tail-immersion and hot-plate tests. In GABA(B1) knockout mice, the hypolocomotive effect was observed only with the highest dose of nicotine, and the antinociceptive responses in both tests were significantly reduced in GABA(B1) knockout mice compared to their wild-type littermate. Additionally, nicotine elicited anxiolytic- (0.05 mg/kg) and anxiogenic-like (0.8 mg/kg) responses in the elevated plus-maze test in wild-type mice, while selectively the anxiolytic-like effect was abolished in GABA(B1) knockout mice. We further investigated nicotine withdrawal in mice chronically treated with nicotine (25 mg/kg/day, sc). Mecamylamine (1 mg/kg, sc) precipitated several somatic signs of nicotine withdrawal in wild-type mice. However, signs of nicotine withdrawal were missing in GABA(B1) knockout mice. Finally, there was a decreased immunoreactivity of Fos-positive nuclei in the bed nucleus of the stria terminalis, basolateral amygdaloid nucleus and hippocampal dentate gyrus in abstinent wild-type but not in GABA(B1) knockout mice. These results reveal an interaction between the GABA(B) system and the neurochemical systems through which nicotine exerts its acute and long-term effects.
Collapse
Affiliation(s)
- Andrés P Varani
- Instituto de Investigaciones Farmacológicas-CONICET, Junín 956, 5° Piso, Buenos Aires C1113AAD, Argentina
| | | | | | | |
Collapse
|
12
|
Akula KK, Kulkarni SK. Adenosinergic system: an assorted approach to therapeutics for drug addiction. FUTURE NEUROLOGY 2012. [DOI: 10.2217/fnl.12.19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Adenosine is an endogenous purine nucleoside and it is extensively present in the brain. It exerts several metabolic and neuromodulatory roles in the body. Adenosine also acts as an important messenger molecule for extracellular signaling and shows a homeostatic neuromodulatory function at the synaptic level. Extracellular adenosine exerts a wide variety of biological actions through four cell surface G-protein-coupled receptor subtypes, namely A1, A2A, A2B and A3 adenosine receptors. The extracellular levels of adenosine have been found to be enhanced in several neuropathological conditions, including drug addiction, and thus a neuroprotective role of adenosine was perceived by various experimental studies. The aversive withdrawal symptoms emanating from drug discontinuation provokes rebound drug intake patterns. In addition, alteration of neurotransmitter(s) release and changes in receptor expression contribute to the behavioral changes of drug withdrawal. Furthermore, the abuse of major drugs such as alcohol and opioids are reported to modulate extracellular adenosine levels. In this context, the neuromodulatory functions of adenosine would be valuable if projected to the clinical applications and thus, an increasing attention is currently given to the functional role of adenosine in human addictive disorders. This review will focus on recent clinical and experimental studies that reveal the actions of adenosine and related ligands in drug addiction and various drug-withdrawal syndromes. The evidence and reports provided in this review highlight the looming therapeutic potential of purinergic drugs, with a hope that new therapeutic interventions based on the adenosinergic concept will emerge in the coming years for the management of drug withdrawal syndrome.
Collapse
Affiliation(s)
- Kiran Kumar Akula
- R.S. Dow Neurobiology Laboratories, Legacy Research, 1225 NE 2nd Avenue, Portland, OR 97232, USA
| | - SK Kulkarni
- Pharmacology Division, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh-160014, India
| |
Collapse
|
13
|
Wells L, Opacka-Juffry J, Fisher D, Ledent C, Hourani S, Kitchen I. In vivo dopaminergic and behavioral responses to acute cocaine are altered in adenosine A(2A) receptor knockout mice. Synapse 2012; 66:383-90. [PMID: 22213208 DOI: 10.1002/syn.21527] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Accepted: 12/14/2011] [Indexed: 11/07/2022]
Abstract
Adenosine, acting on adenosine A(2A) receptors (A2ARs), regulates addictive processes induced by drugs of abuse. This study investigates the role of A(2A) adenosine receptors in neurochemical and behavioral responses to an acute cocaine challenge. Changes in the extracellular levels of dopamine (DA) in the nucleus accumbens (NAc) of mice lacking A(2A) adenosine receptors and wild type (WT) littermates after an acute cocaine (20 mg/kg) administration were evaluated by in vivo microdialysis studies. Locomotor effects induced by cocaine were measured during the microdialysis procedure. Cocaine-evoked increases in extracellular DA were not sustained in mice lacking A(2A) Rs in comparison with wild-type mice (P < 0.05). Cocaine administration significantly increased ambulatory activity in both genotypes. However, overall locomotor activity was further increased, whereas rest and small local movement measures were significantly attenuated in the A(2A) R knockout mice compared with WT littermates (P < 0.05). Our findings support an important role for adenosine A(2A) R in modulating the acute effects of cocaine, as demonstrated by the decrease in cocaine-evoked dopaminergic transmission in the NAc. Furthermore, the results support an important antagonistic role of A(2A) R in vivo in regulating psychostimulant-induced hyperlocomotion.
Collapse
Affiliation(s)
- Lisa Wells
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey GU2 7XH, United Kingdom.
| | | | | | | | | | | |
Collapse
|
14
|
Micioni Di Bonaventura MV, Cifani C, Lambertucci C, Volpini R, Cristalli G, Froldi R, Massi M. Effects of A₂A adenosine receptor blockade or stimulation on alcohol intake in alcohol-preferring rats. Psychopharmacology (Berl) 2012; 219:945-57. [PMID: 21833502 DOI: 10.1007/s00213-011-2430-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Accepted: 07/22/2011] [Indexed: 10/17/2022]
Abstract
RATIONALE A(2A) adenosine receptors (A(2A)ARs) have been proposed to be involved in drug addiction; however, preclinical studies about the effects of A(2A)AR ligands on alcohol consumption have provided inconsistent results. OBJECTIVES The present study evaluated the effect of intraperitoneal injections of the A(2A)AR antagonist ANR 94, and the A(2A)AR agonists CGS 21680 and VT 7 on voluntary drinking and operant self-administration of 10% ethanol in Marchigian Sardinian alcohol-preferring (msP) rats. RESULTS Voluntary ethanol drinking was increased by ANR 94 in acute and subchronic experiments, while it was reduced by A(2A)AR agonists. The effect of CGS 21680 was abolished by a low dose of ANR 94, confirming its mediation by A(2A)ARs. Ethanol self-administration was reduced by CGS 21680 and VT 7, while ANR 94 slightly but significantly increased it. Blood alcohol levels were not modified by A(2A)AR agonists, indicating that their effect is not related to ethanol pharmacokinetics. The effect of VT 7 on ethanol drinking was behaviourally selective; ethanol and food intake were reduced, but water intake was increased, and total fluid intake was not different from that of controls. Moreover, VT 7 did not affect locomotor activity. CGS 21680 (0.1 mg/kg) did not modify total fluid intake, but 0.2 and 0.3 mg/kg reduced total fluid intake and locomotor activity. CONCLUSION These results provide evidence that A(2A)AR agonists reduce ethanol consumption in msP rats, which represent an animal model of alcohol abuse related to stress, anxiety and depression. A(2A)ARs may represent a potential target for treatment of alcohol abuse.
Collapse
|
15
|
Listos J, Talarek S, Poleszak E, Wróbel A, Fidecka S. Attenuating effect of adenosine receptor agonists on the development of behavioral sensitization induced by sporadic treatment with morphine. Pharmacol Biochem Behav 2011; 98:356-61. [DOI: 10.1016/j.pbb.2011.01.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Revised: 01/17/2011] [Accepted: 01/24/2011] [Indexed: 10/18/2022]
|
16
|
Wei CJ, Li W, Chen JF. Normal and abnormal functions of adenosine receptors in the central nervous system revealed by genetic knockout studies. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2010; 1808:1358-79. [PMID: 21185258 DOI: 10.1016/j.bbamem.2010.12.018] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2010] [Revised: 12/14/2010] [Accepted: 12/15/2010] [Indexed: 12/17/2022]
Abstract
Endogenous adenosine is a widely distributed upstream regulator of a broad spectrum of neurotransmitters, receptors, and signaling pathways that converge to contribute to the expression of an array of important brain functions. Over the past decade, the generation and characterization of genetic knockout models for all four G-protein coupled adenosine receptors, the A1 and A2A receptors in particular, has confirmed and extended the neuromodulatory and integrated role of adenosine receptors in the control of a broad spectrum of normal and abnormal brain functions. After a brief introduction of the available adenosine receptor knockout models, this review focuses on findings from the genetic knockout approach, placing particular emphasis on the most recent findings. This review is organized into two sections to separately address (i) the role of adenosine receptors in normal brain processes including neuroplasticity, sleep-wake cycle, motor function, cognition, and emotion-related behaviors; and (ii) their role in the response to various pathologic insults to brain such as ischemic stroke, neurodegeneration, or brain dysfunction/disorders. We largely limit our overview to the prominent adenosine receptor subtypes in brain-the A1 and A2A receptors-for which numerous genetic knockout studies on brain function are available. A1 and A2A receptor knockouts have provided significant new insights into adenosine's control of complex physiologic (e.g., cognition) and pathologic (e.g., neuroinflammation) phenomena. These findings extend and strengthen the support for A1 and A2A receptors in brain as therapeutic targets in several neurologic and psychiatric diseases. However, they also emphasize the importance of considering the disease context-dependent effect when developing adenosine receptor-based therapeutic strategies.
Collapse
Affiliation(s)
- Catherine J Wei
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
| | | | | |
Collapse
|
17
|
Adams CL, Short JL, Lawrence AJ. Cue-conditioned alcohol seeking in rats following abstinence: involvement of metabotropic glutamate 5 receptors. Br J Pharmacol 2010; 159:534-42. [PMID: 20067474 DOI: 10.1111/j.1476-5381.2009.00562.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE The current study was designed to: (i) examine whether functional interactions occur between receptors known to regulate alcohol self-administration; and (ii) characterize relapse to alcohol seeking following abstinence. EXPERIMENTAL APPROACH The selective cannabinoid CB(1) receptor antagonist SR141716A (0.03-1.0 mg.kg(-1) i.p.) resulted in a dose-dependent reduction in ethanol self-administration in ethanol-preferring Indiana-preferring rats. SR141716A was then co-administered with either the selective glutamate metabotropic glutamate 5 (mGlu(5)) receptor antagonist 3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]pyridine (MTEP) or the selective adenosine A(2A) receptor antagonist SCH58261. KEY RESULTS When administered at individually sub-threshold doses, a combination of SR141716A (0.1 mg.kg(-1)) and SCH58261 (0.5 mg.kg(-1) i.p.) produced a reduction (28%) in ethanol self-administration. Combinations of threshold doses of SR141716A (0.3 mg.kg(-1)) and SCH58261 (2.0 mg.kg(-1), i.p.) caused an essentially additive reduction (68%) in alcohol self-administration. A combination of individually sub-threshold doses of CB(1) and mGlu(5) receptor antagonists did not affect alcohol self-administration; however, combined threshold doses of SR141716A (0.3 mg.kg(-1)) and MTEP (1.0 mg.kg(-1) i.p.) did reduce ethanol self-administration markedly (80%). Cue-conditioned alcohol seeking was attenuated by pretreatment with MTEP (1.0 mg.kg(-1)) co-administered with SR141716A (0.3 mg.kg(-1) i.p.). In contrast, SCH58261 (2.0 mg.kg(-1)) co-administered with SR141716A (0.3 mg.kg(-1) i.p.) did not reduce cue-conditioned alcohol seeking. CONCLUSIONS AND IMPLICATIONS Adenosine A(2A) and cannabinoid CB(1) receptors regulated alcohol self-administration additively, but combined low-dose antagonism of these receptors did not prevent cue-conditioned alcohol seeking after abstinence. In contrast, combined low-dose antagonism of mGlu(5) and CB(1) receptors did prevent relapse-like alcohol seeking after abstinence, suggesting a prominent role for mGlu(5) receptors in this paradigm.
Collapse
Affiliation(s)
- C L Adams
- Howard Florey Institute, Parkville, Vic., Australia
| | | | | |
Collapse
|
18
|
Jupp B, Lawrence AJ. New horizons for therapeutics in drug and alcohol abuse. Pharmacol Ther 2010; 125:138-68. [DOI: 10.1016/j.pharmthera.2009.11.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Accepted: 11/03/2009] [Indexed: 11/25/2022]
|
19
|
Abstract
This paper is the 31st consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2008 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurologic disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration and thermoregulation (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, 65-30 Kissena Blvd, Flushing, NY 11367, United States.
| |
Collapse
|