1
|
Gómez-Acero L, Sánchez-Fernández N, Subirana P, Ciruela F, Aso E. Striatal dopamine D 2, adenosine A 2A and cannabinoid CB 1 receptors balance as a target against non-cognitive symptoms in a mouse model of Alzheimer's disease. Pharmacol Biochem Behav 2025; 249:173970. [PMID: 39956277 DOI: 10.1016/j.pbb.2025.173970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 12/03/2024] [Accepted: 02/07/2025] [Indexed: 02/18/2025]
Abstract
Behavioral and psychological symptoms of dementia are almost ubiquitous in Alzheimer's disease (AD) but current therapies are not fully effective and safe. In this study, we aim to evaluate the role played by the interplay among striatal D2, adenosine A2A (A2AR) and cannabinoid CB1 (CB1R) receptors in some of these non-cognitive impairments in a well-established animal model of AD, the double transgenic APP/PS1 mice. Our results reveal that the alterations existing in the ratios between these three receptors significantly correlate with the sensorimotor gating and the social interaction impairments occurring in APP/PS1 mice at 12 months of age. Moreover, the pharmacological stimulation of A2AR and CB1R blunted the sensorimotor gating deficiencies in APP/PS1 mice. To note, we observed some age-dependent differences among male and female mice. In conclusion, the present study provides evidence for the contribution of an altered interplay between dopaminergic, adenosinergic and endocannabinoid systems in the sensorimotor gating deficits and social withdrawal occurring in AD and points to A2AR and CB1R as a potential target to reverse these non-cognitive symptoms in AD patients.
Collapse
Affiliation(s)
- Laura Gómez-Acero
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain; Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, Spain
| | - Nuria Sánchez-Fernández
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain; Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, Spain
| | - Paula Subirana
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain; Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, Spain
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain; Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, Spain
| | - Ester Aso
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain; Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, Spain.
| |
Collapse
|
2
|
You B, Shi G, Zhang Y, Fu X, Li Q, Wang Y, Huang G, Fang Y, Li R. The interaction of adenosine and dopamine in modulating the consequences of central nervous system oxygen toxicity. J Appl Physiol (1985) 2025; 138:169-181. [PMID: 39601779 DOI: 10.1152/japplphysiol.00500.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/03/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
Hyperbaric oxygen (HBO) refers to pure oxygen with a pressure greater than 1 atmospheres absolute (ATA), and when the pressure is too high, it can cause convulsive attacks. Adenosine and dopamine have been shown to be closely associated with HBO-induced convulsion seizures, and their receptors exhibited a coexisting relationship of mutual antagonism on the membrane of nerve cells. We explored the influence of adenosine and dopamine interplay on the occurrence of oxygen convulsion. Rats were individually exposed to HBO of 6 ATA and treated with adenosine, dopamine, and their receptor modulators separately and jointly, with the latency of convulsion onset recorded. In addition, after administering adenosine to rats and exposing them to HBO for 30 min, the content of dopamine and its metabolites and the activity of enzymes related to their metabolism were measured. The results revealed that dopamine was effective in resisting convulsion (>60 min vs. 32.53 ± 5.31 min, P = 0.000), and low-dose adenosine partially counteracted its effect (>60 min vs. 28.18 ± 6.24 min, P = 0.002). The combined use of adenosine A1 and dopamine D1 receptor modulators significantly impacted the incidence of convulsion. The activation or inhibition of the A2A receptor had a particularly significant impact on convulsion, whereas modulating the D2 receptor did not affect their effects. The combination of A1 agonist and D2 agonist was highly effective in resisting convulsion (>60 min vs. 32.53 ± 5.31 min, P = 0.000). Exposure to HBO accelerated the metabolism of dopamine to its end products, which may be related to the enhanced activity of monoamine oxidase (MAO). Adenosine can inhibit MAO activity (0.0766 ± 0.0150 U/mg.prot vs. 0.1055 ± 0.0086 U/mg.prot, P = 0.004), maintaining a higher level of dopamine (1.820 ± 0.379 mg/g vs. 0.602 ± 0.087 mg/g, P = 0.000). The study demonstrated that dopamine plays a significant role in oxygen convulsion and adenosine can affect dopamine metabolism. The interaction between them can have a crucial impact on the occurrence of oxygen convulsion. The findings offer a novel perspective for further investigating the mechanism of oxygen convulsion and exploring effective preventive strategies.NEW & NOTEWORTHY The interaction between adenosine and dopamine is critically important in determining the incidence of oxygen convulsion. Simultaneous regulation of both adenosine and dopamine offers a superior approach to counteract oxygen convulsion, achieving a synergistic effect exceeding the sum of their individual impacts. These findings provide new directions and insights for future in-depth and systematic exploration of the pathogenesis of central nervous system oxygen toxicity.
Collapse
Affiliation(s)
- Benming You
- Department of Pharmacy, Changhai Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Guorong Shi
- Department of Pharmacy, Shidong Hospital of Shanghai Yangpu District, Shanghai, People's Republic of China
| | - Yanan Zhang
- Department of Diving and Hyperbaric Medicine, PLA Naval Medical Center, Naval Medical University, Shanghai, People's Republic of China
| | - Xiang Fu
- Department of Pharmacy, Shidong Hospital of Shanghai Yangpu District, Shanghai, People's Republic of China
| | - Qian Li
- Department of Pharmacy, Changhai Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Yu Wang
- Department of Pharmacy, Changhai Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Guoyang Huang
- Department of Diving and Hyperbaric Medicine, PLA Naval Medical Center, Naval Medical University, Shanghai, People's Republic of China
| | - Yiqun Fang
- Department of Diving and Hyperbaric Medicine, PLA Naval Medical Center, Naval Medical University, Shanghai, People's Republic of China
| | - Runping Li
- Department of Diving and Hyperbaric Medicine, PLA Naval Medical Center, Naval Medical University, Shanghai, People's Republic of China
| |
Collapse
|
3
|
Abeledo-Machado A, Argerich J, Yaneff A, Vidal N, García-Roca C, Bornancini D, Peña-Zanoni M, Gironacci MM, Shayo C, Ciruela F, Díaz-Torga G. B2R-D2R Interaction in Prolactinomas and Nonfunctional Adenomas: Impact on Dopamine Resistance. Endocrinology 2024; 165:bqae144. [PMID: 39487739 DOI: 10.1210/endocr/bqae144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/15/2024] [Accepted: 10/31/2024] [Indexed: 11/04/2024]
Abstract
Prolactinomas, the most common pituitary-secreting adenomas, can be effectively treated with dopamine D2 receptor (D2R) agonists. However, a subset of them (∼20%) are resistant to dopamine-based therapies and require extirpation. The molecular mechanisms underlying their escape from dopaminergic regulation are not fully elucidated and may include alterations in D2R signaling. D2R can heteromerize with other G protein-coupled receptors, resulting in modulation of dopaminergic signaling. Because the bradykinin receptor type 2 (B2R) is overexpressed in prolactinomas, we interrogated whether this dopaminergic dysregulation observed in some prolactinomas may depend on a physical and functional interaction between D2R and B2R. The formation of B2R-D2R complexes in cultured cells transiently expressing both receptors was validated using NanoBiT technology. Interestingly, although D2R stimulation did not alter B2R-induced intracellular calcium mobilization, B2R stimulation abolished D2R signaling through modulation of cAMP. The existence of B2R-D2R complexes in pituitary adenomas biopsies was evaluated using an ALPHALisa approach. Importantly, B2R-D2R complexes were detected in human prolactinomas and nonfunctioning pituitary adenomas, but not in mixed (prolactin + growth hormone)-secreting adenomas. These results suggest that overexpression of B2R in resistant prolactinomas may promote the formation of B2R-D2R complexes, with B2R precluding D2R signaling, thus generating resistance to D2R agonists.
Collapse
Affiliation(s)
| | - Josep Argerich
- Department of Pathology and Experimental Therapeutics, Pharmacology Unit, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 Barcelona, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, 08907 Barcelona, Spain
| | - Agustín Yaneff
- Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Universidad de Buenos Aires, C1113AAD Buenos Aires, Argentina
| | - Noemi Vidal
- Neurohatology, Servei d´Anatomia Patològica, Hospital Universitari de Bellvitge, 08907 Barcelona, Spain
| | - Claudia García-Roca
- Neurohatology, Servei d´Anatomia Patològica, Hospital Universitari de Bellvitge, 08907 Barcelona, Spain
| | - Dana Bornancini
- Laboratorio de Fisiopatología Hormonal, IBYME-CONICET, C1428ADN Buenos Aires, Argentina
| | - Milagros Peña-Zanoni
- Laboratorio de Fisiopatología Hormonal, IBYME-CONICET, C1428ADN Buenos Aires, Argentina
| | - Mariela M Gironacci
- Facultad de Farmacia y Bioquímica, Departamento de Química Biológica and IQUIFIB (UBA-CONICET), Universidad de Buenos Aires, C1113AAD Buenos Aires, Argentina
| | - Carina Shayo
- Laboratorio de Patología y Farmacología Molecular, IBYME-CONICET, C1428ADN Buenos Aires, Argentina
| | - Francisco Ciruela
- Department of Pathology and Experimental Therapeutics, Pharmacology Unit, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 Barcelona, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, 08907 Barcelona, Spain
| | - Graciela Díaz-Torga
- Laboratorio de Fisiopatología Hormonal, IBYME-CONICET, C1428ADN Buenos Aires, Argentina
| |
Collapse
|
4
|
Ballardin D, Makrini-Maleville L, Seper A, Valjent E, Rebholz H. 5-HT4R agonism reduces L-DOPA-induced dyskinesia via striatopallidal neurons in unilaterally 6-OHDA lesioned mice. Neurobiol Dis 2024; 198:106559. [PMID: 38852753 DOI: 10.1016/j.nbd.2024.106559] [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: 03/25/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024] Open
Abstract
Parkinson's disease is caused by a selective vulnerability and cell loss of dopaminergic neurons of the Substantia Nigra pars compacta and, consequently, striatal dopamine depletion. In Parkinson's disease therapy, dopamine loss is counteracted by the administration of L-DOPA, which is initially effective in ameliorating motor symptoms, but over time leads to a burdening side effect of uncontrollable jerky movements, termed L-DOPA-induced dyskinesia. To date, no efficient treatment for dyskinesia exists. The dopaminergic and serotonergic systems are intrinsically linked, and in recent years, a role has been established for pre-synaptic 5-HT1a/b receptors in L-DOPA-induced dyskinesia. We hypothesized that post-synaptic serotonin receptors may have a role and investigated the effect of modulation of 5-HT4 receptor on motor symptoms and L-DOPA-induced dyskinesia in the unilateral 6-OHDA mouse model of Parkinson's disease. Administration of RS 67333, a 5-HT4 receptor partial agonist, reduces L-DOPA-induced dyskinesia without altering L-DOPA's pro-kinetic effect. In the dorsolateral striatum, we find 5-HT4 receptor to be predominantly expressed in D2R-containing medium spiny neurons, and its expression is altered by dopamine depletion and L-DOPA treatment. We further show that 5-HT4 receptor agonism not only reduces L-DOPA-induced dyskinesia, but also enhances the activation of the cAMP-PKA pathway in striatopallidal medium spiny neurons. Taken together, our findings suggest that agonism of the post-synaptic serotonin receptor 5-HT4 may be a novel therapeutic approach to reduce L-DOPA-induced dyskinesia.
Collapse
Affiliation(s)
- Demetra Ballardin
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Laboratory of Signaling mechanisms in neurological disorders, 75014 Paris, France
| | | | - Alexander Seper
- Center of Neurodegeneration, Faculty of Medicine, Danube Private University, Krems, Austria
| | - Emmanuel Valjent
- IGF, Univ. Montpellier, CNRS, INSERM, F-34094 Montpellier, France
| | - Heike Rebholz
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Laboratory of Signaling mechanisms in neurological disorders, 75014 Paris, France; Center of Neurodegeneration, Faculty of Medicine, Danube Private University, Krems, Austria; GHU-Paris Psychiatrie et Neuroscience, Hôpital Sainte Anne, F-75014 Paris, France.
| |
Collapse
|
5
|
Mercante F, Micioni Di Bonaventura E, Pucci M, Botticelli L, Cifani C, D'Addario C, Micioni Di Bonaventura MV. Repeated binge-like eating episodes in female rats alter adenosine A 2A and dopamine D2 receptor genes regulation in the brain reward system. Int J Eat Disord 2024; 57:1433-1446. [PMID: 38650547 DOI: 10.1002/eat.24216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/22/2024] [Accepted: 04/01/2024] [Indexed: 04/25/2024]
Abstract
OBJECTIVE Binge-eating disorder is an eating disorder characterized by recurrent binge-eating episodes, during which individuals consume excessive amounts of highly palatable food (HPF) in a short time. This study investigates the intricate relationship between repeated binge-eating episode and the transcriptional regulation of two key genes, adenosine A2A receptor (A2AAR) and dopamine D2 receptor (D2R), in selected brain regions of rats. METHOD Binge-like eating behavior on HPF was induced through the combination of food restrictions and frustration stress (15 min exposure to HPF without access to it) in female rats, compared to control rats subjected to only restriction or only stress or none of these two conditions. After chronic binge-eating episodes, nucleic acids were extracted from different brain regions, and gene expression levels were assessed through real-time quantitative PCR. The methylation pattern on genes' promoters was investigated using pyrosequencing. RESULTS The analysis revealed A2AAR upregulation in the amygdala and in the ventral tegmental area (VTA), and D2R downregulation in the nucleus accumbens in binge-eating rats. Concurrently, site-specific DNA methylation alterations at gene promoters were identified in the VTA for A2AAR and in the amygdala and caudate putamen for D2R. DISCUSSION The alterations on A2AAR and D2R genes regulation highlight the significance of epigenetic mechanisms in the etiology of binge-eating behavior, and underscore the potential for targeted therapeutic interventions, to prevent the development of this maladaptive feeding behavior. These findings provide valuable insights for future research in the field of eating disorders. PUBLIC SIGNIFICANCE Using an animal model with face, construct, and predictive validity, in which cycles of food restriction and frustration stress evoke binge-eating behavior, we highlight the significance of epigenetic mechanisms on adenosine A2A receptor (A2AAR) and dopamine D2 receptor (D2R) genes regulation. They could represent new potential targets for the pharmacological management of eating disorders characterized by this maladaptive feeding behavior.
Collapse
Affiliation(s)
- Francesca Mercante
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | | | - Mariangela Pucci
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Luca Botticelli
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Carlo Cifani
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Claudio D'Addario
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
6
|
de Moraes Santos Corrêa É, Christofoletti G, de Souza AS. Effects of Intracerebral Aminophylline Dosing on Catalepsy and Gait in an Animal Model of Parkinson's Disease. Int J Mol Sci 2024; 25:5191. [PMID: 38791229 PMCID: PMC11120906 DOI: 10.3390/ijms25105191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
Parkinson's disease (PD) is a progressive disorder characterized by the apoptosis of dopaminergic neurons in the basal ganglia. This study explored the potential effects of aminophylline, a non-selective adenosine A1 and A2A receptor antagonist, on catalepsy and gait in a haloperidol-induced PD model. Sixty adult male Swiss mice were surgically implanted with guide cannulas that targeted the basal ganglia. After seven days, the mice received intraperitoneal injections of either haloperidol (experimental group, PD-induced model) or saline solution (control group, non-PD-induced model), followed by intracerebral infusions of aminophylline. The assessments included catalepsy testing on the bar and gait analysis using the Open Field Maze. A two-way repeated-measures analysis of variance (ANOVA), followed by Tukey's post hoc tests, was employed to evaluate the impact of groups (experimental × control), aminophylline (60 nM × 120 nM × saline/placebo), and interactions. Significance was set at 5%. The results revealed that the systemic administration of haloperidol in the experimental group increased catalepsy and dysfunction of gait that paralleled the observations in PD. Co-treatment with aminophylline at 60 nM and 120 nM reversed catalepsy in the experimental group but did not restore the normal gait pattern of the animals. In the non-PD induced group, which did not present any signs of catalepsy or motor dysfunctions, the intracerebral dose of aminophylline did not exert any interference on reaction time for catalepsy but increased walking distance in the Open Field Maze. Considering the results, this study highlights important adenosine interactions in the basal ganglia of animals with and without signs comparable to those of PD. These findings offer valuable insights into the neurobiology of PD and emphasize the importance of exploring novel therapeutic strategies to improve patient's catalepsy and gait.
Collapse
Affiliation(s)
| | | | - Albert Schiaveto de Souza
- Faculty of Medicine, Institute of Health, Federal University of Mato Grosso do Sul, UFMS, Campo Grande 79060-900, Brazil; (É.d.M.S.C.); (G.C.)
| |
Collapse
|
7
|
Bechard AR, McElderry S. Environmental interventions reduced repetitive behavior in a mouse model. Physiol Behav 2024; 273:114386. [PMID: 37884109 DOI: 10.1016/j.physbeh.2023.114386] [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: 05/17/2023] [Revised: 10/06/2023] [Accepted: 10/20/2023] [Indexed: 10/28/2023]
Abstract
Repetitive motor behaviors are associated with several neurodevelopmental disorders including autism spectrum disorder. Non-invasive environmental interventions that can ameliorate repetitive behavior and be introduced in early development could benefit many. In Experiment 1, we characterized the development of repetitive circling in mice reared in standard and enriched environments. Environmental enrichment was associated with reduced repetitive behavior. In Experiment 2, two weekly injections of an A2A adenosine receptor agonist reduced repetitive behavior in mice fed a ketogenic diet. Together, these two approaches modified the environment and reduced repetitive behavior with potential implications for increased functioning of the indirect basal ganglia pathway.
Collapse
|
8
|
Ferré S, Köfalvi A, Ciruela F, Justinova Z, Pistis M. Targeting corticostriatal transmission for the treatment of cannabinoid use disorder. Trends Pharmacol Sci 2023; 44:495-506. [PMID: 37331914 PMCID: PMC10524660 DOI: 10.1016/j.tips.2023.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 06/20/2023]
Abstract
It is generally assumed that the rewarding effects of cannabinoids are mediated by cannabinoid CB1 receptors (CB1Rs) the activation of which disinhibits dopaminergic neurons in the ventral tegmental area (VTA). However, this mechanism cannot fully explain novel results indicating that dopaminergic neurons also mediate the aversive effects of cannabinoids in rodents, and previous results showing that preferentially presynaptic adenosine A2A receptor (A2AR) antagonists counteract self-administration of Δ-9-tetrahydrocannabinol (THC) in nonhuman primates (NHPs). Based on recent experiments in rodents and imaging studies in humans, we propose that the activation of frontal corticostriatal glutamatergic transmission constitutes an additional and necessary mechanism. Here, we review evidence supporting the involvement of cortical astrocytic CB1Rs in the activation of corticostriatal neurons and that A2AR receptor heteromers localized in striatal glutamatergic terminals mediate the counteracting effects of the presynaptic A2AR antagonists, constituting potential targets for the treatment of cannabinoid use disorder (CUD).
Collapse
Affiliation(s)
- Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA.
| | - Attila Köfalvi
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Spain; Neuroscience Program, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, Spain
| | - Zuzana Justinova
- Division of Pharmacology, Physiology, and Biological Chemistry (PPBC), National Institute of General Medical Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Marco Pistis
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy; Neuroscience Institute, Section of Cagliari, National Research Council of Italy (CNR), Cagliari, Italy
| |
Collapse
|
9
|
Eun Kim G, Jun Kim H, Jun Jin H. The association between Adenosine A 2A receptor gene polymorphisms and attention deficit hyperactivity disorder in Korean children. Gene 2023; 876:147503. [PMID: 37220831 DOI: 10.1016/j.gene.2023.147503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 05/06/2023] [Accepted: 05/19/2023] [Indexed: 05/25/2023]
Abstract
Attention deficit hyperactivity disorder (ADHD) is a common and heritable neurodevelopmental disorder. Particularly, ADHD is known to be related to the dopaminergic system. ADHD symptoms can appear when the dopamine binding affinity diminishes due to dopamine receptor abnormalities, such as the dopamine D2 receptor (D2R). This receptor interacts with the adenosine A2A receptor (A2AR). The A2AR acts as an antagonist of D2R, that is, the increased binding of adenosine with A2AR inhibits the D2R activity. Furthermore, it is found that the single nucleotide polymorphisms of the adenosine A2A receptor gene (ADORA2A) revealed a significant relationship with ADHD in various populations. Therefore, we examined the genetic relationship between ADORA2A polymorphisms (rs2297838, rs5751876, and rs4822492) and Korean ADHD children. A case-control study was performed for 150 cases and 322 controls. Genotyping of ADORA2A polymorphisms was conducted by PCR-RFLP. The results demonstrated that the rs5751876 TC genotype was associated with children with ADHD (p = 0.018). The rs2298383 CC genotype was significantly associated with children with ADHD/HI (p = 0.026). However, when Bonferroni correction was used, the significance vanished (padjusted = 0.054 and padjusted = 0.078, respectively). Haplotype analysis showed that TTC, TCC, and CTG demonstrated a significant difference between ADHD/C children and control groups (padjusted = 0.006, padjusted = 0.011, and padjusted = 0.028, respectively). In conclusion, we propose a possible association between ADORA2A polymorphisms with Korean children having ADHD.
Collapse
Affiliation(s)
- Ga Eun Kim
- Department of Biological Sciences, College of Science & Technology, Dankook University, Cheonan, South Korea
| | - Hyung Jun Kim
- Department of Biological Sciences, College of Science & Technology, Dankook University, Cheonan, South Korea
| | - Han Jun Jin
- Department of Biological Sciences, College of Science & Technology, Dankook University, Cheonan, South Korea.
| |
Collapse
|
10
|
Jenner P, Kanda T, Mori A. How and why the adenosine A 2A receptor became a target for Parkinson's disease therapy. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 170:73-104. [PMID: 37741697 DOI: 10.1016/bs.irn.2023.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2023]
Abstract
Dopaminergic therapy for Parkinson's disease has revolutionised the treatment of the motor symptoms of the illness. However, it does not alleviate all components of the motor deficits and has only limited effects on non-motor symptoms. For this reason, alternative non-dopaminergic approaches to treatment have been sought and the adenosine A2A receptor provided a novel target for symptomatic therapy both within the basal ganglia and elsewhere in the brain. Despite an impressive preclinical profile that would indicate a clear role for adenosine A2A antagonists in the treatment of Parkinson's disease, the road to clinical use has been long and full of difficulties. Some aspects of the drugs preclinical profile have not translated into clinical effectiveness and not all the clinical studies undertaken have had a positive outcome. The reasons for this will be explored and suggestions made for the further development of this drug class in the treatment of Parkinson's disease. However, one adenosine A2A antagonist, namely istradefylline has been introduced successfully for the treatment of late-stage Parkinson's disease in two major areas of the world and has become a commercial success through offering the first non-dopaminergic approach to the treatment of unmet need to be introduced in several decades.
Collapse
Affiliation(s)
- Peter Jenner
- Institute of Pharmaceutical Sciences, King's College London, London, United Kingdom.
| | - Tomoyuki Kanda
- Kyowa Kirin Co., Ltd., Otemachi. Chiyoda-ku, Tokyo, Japan
| | | |
Collapse
|
11
|
Chen JF, Choi DS, Cunha RA. Striatopallidal adenosine A 2A receptor modulation of goal-directed behavior: Homeostatic control with cognitive flexibility. Neuropharmacology 2023; 226:109421. [PMID: 36634866 PMCID: PMC10132052 DOI: 10.1016/j.neuropharm.2023.109421] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/30/2022] [Accepted: 01/08/2023] [Indexed: 01/11/2023]
Abstract
Dysfunction of goal-directed behaviors under stressful or pathological conditions results in impaired decision-making and loss of flexibility of thoughts and behaviors, which underlie behavioral deficits ranging from depression, obsessive-compulsive disorders and drug addiction. Tackling the neuromodulators fine-tuning this core behavioral element may facilitate the development of effective strategies to control these deficits present in multiple psychiatric disorders. The current investigation of goal-directed behaviors has concentrated on dopamine and glutamate signaling in the corticostriatal pathway. In accordance with the beneficial effects of caffeine intake on mood and cognitive dysfunction, we now propose that caffeine's main site of action - adenosine A2A receptors (A2AR) - represent a novel target to homeostatically control goal-directed behavior and cognitive flexibility. A2AR are abundantly expressed in striatopallidal neurons and colocalize and interact with dopamine D2, NMDA and metabotropic glutamate 5 receptors to integrate dopamine and glutamate signaling. Specifically, striatopallidal A2AR (i) exert an overall "break" control of a variety of cognitive processes, making A2AR antagonists a novel strategy for improving goal-directed behavior; (ii) confer homeostatic control of goal-directed behavior by acting at multiple sites with often opposite effects, to enhance cognitive flexibility; (iii) integrate dopamine and adenosine signaling through multimeric A2AR-D2R heterocomplexes allowing a temporally precise fine-tuning in response to local signaling changes. As the U.S. Food and Drug Administration recently approved the A2AR antagonist Nourianz® (istradefylline) to treat Parkinson's disease, striatal A2AR-mediated control of goal-directed behavior may offer a new and real opportunity for improving deficits of goal-directed behavior and enhance cognitive flexibility under various neuropsychiatric conditions. This article is part of the Special Issue on "Purinergic Signaling: 50 years".
Collapse
Affiliation(s)
- Jiang-Fan Chen
- Molecular Neuropharmacology Laboratory, Wenzhou Medical University, Wenzhou, China; Department of Neurology, School of Medicine, Boston University, Boston, MA, USA.
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, USA; Department of Psychiatry and Psychology, Mayo Clinic College of Medicine, Rochester, MN, USA.
| | - Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; FMUC-Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
12
|
Emtage JA, Shipman ML, Corbit LH. The role of dorsomedial striatum adenosine 2A receptors in the loss of goal-directed behaviour. Psychopharmacology (Berl) 2023; 240:547-559. [PMID: 36129491 DOI: 10.1007/s00213-022-06220-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/23/2022] [Indexed: 11/30/2022]
Abstract
RATIONALE Adenosine A2A receptors (A2AR) in the dorsal striatum have been implicated in goal-directed behaviour. While activation of these receptors with several methods has resulted in an insensitivity to outcome devaluation, particular explanations for how they disrupt behaviour have not been explored. We both confirm a role for A2A receptors in goal-directed responding and evaluate additional behavioural aspects of goal-directed control to more fully understand the role of A2A receptors in instrumental behaviour. OBJECTIVES To examine the effects of the adenosine A2A agonist CGS-21680 in the DMS on response-outcome encoding, updating representations of outcome value and on the ability to inhibit behaviour when reward is not available. METHODS Male rats were trained to lever press for food reward. The A2AR agonist CGS-21680 was infused into the dorsomedial striatum either before an outcome devaluation test, prior to training with two distinct response-outcome associations or prior to a test of discriminative stimulus control over instrumental performance. RESULTS Intra-DMS administration of CGS-21680 impaired sensitivity to outcome devaluation. CGS-21680 treatment did not impair acquisition of specific response-outcome associations, selective control of responding based on the presence of stimuli that signaled when reward was or was not available, discrimination between stimuli or lever choices nor did it influence the effect of devaluation on the amounts of food eaten in a consumption test. CONCLUSIONS CGS-21680 impairs the ability to modulate responding based on recent changes to outcome value, an effect that is not accounted for by impairments in behavioural inhibition, discrimination, encoding the specific outcome of a response or the effectiveness of specific satiety.
Collapse
Affiliation(s)
- Jaec A Emtage
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON, M5S 3G5, Canada
| | - Megan L Shipman
- Department of Psychology, University of Toronto, 100 St. George Street, Toronto, ON, M5S 3G3, Canada
| | - Laura H Corbit
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON, M5S 3G5, Canada. .,Department of Psychology, University of Toronto, 100 St. George Street, Toronto, ON, M5S 3G3, Canada.
| |
Collapse
|
13
|
Ferré S, Sarasola LI, Quiroz C, Ciruela F. Presynaptic adenosine receptor heteromers as key modulators of glutamatergic and dopaminergic neurotransmission in the striatum. Neuropharmacology 2023; 223:109329. [PMID: 36375695 DOI: 10.1016/j.neuropharm.2022.109329] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 11/12/2022]
Abstract
Adenosine plays a very significant role in modulating striatal glutamatergic and dopaminergic neurotransmission. In the present essay we first review the extensive evidence that indicates this modulation is mediated by adenosine A1 and A2A receptors (A1Rs and A2ARs) differentially expressed by the components of the striatal microcircuit that include cortico-striatal glutamatergic and mesencephalic dopaminergic terminals, and the cholinergic interneuron. This microcircuit mediates the ability of striatal glutamate release to locally promote dopamine release through the intermediate activation of cholinergic interneurons. A1Rs and A2ARs are colocalized in the cortico-striatal glutamatergic terminals, where they form A1R-A2AR and A2AR-cannabinoid CB1 receptor (CB1R) heteromers. We then evaluate recent findings on the unique properties of A1R-A2AR and A2AR-CB1R heteromers, which depend on their different quaternary tetrameric structure. These properties involve different allosteric mechanisms in the two receptor heteromers that provide fine-tune modulation of adenosine and endocannabinoid-mediated striatal glutamate release. Finally, we evaluate the evidence supporting the use of different heteromers containing striatal adenosine receptors as targets for drug development for neuropsychiatric disorders, such as Parkinson's disease and restless legs syndrome, based on the ability or inability of the A2AR to demonstrate constitutive activity in the different heteromers, and the ability of some A2AR ligands to act preferentially as neutral antagonists or inverse agonists, or to have preferential affinity for a specific A2AR heteromer.
Collapse
Affiliation(s)
- Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes on Drug Abuse, Baltimore, MD, USA.
| | - Laura I Sarasola
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907, L'Hospitalet de Llobregat, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, 08907, L'Hospitalet de Llobregat, Spain
| | - César Quiroz
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes on Drug Abuse, Baltimore, MD, USA
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907, L'Hospitalet de Llobregat, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, 08907, L'Hospitalet de Llobregat, Spain.
| |
Collapse
|
14
|
Dogra N, Jakhmola-Mani R, Potshangbam AM, Buch S, Pande Katare D. CXCR4 as possible druggable target linking inflammatory bowel disease and Parkinson's disease. Metab Brain Dis 2023; 38:1079-1096. [PMID: 36656397 DOI: 10.1007/s11011-022-01155-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/21/2022] [Indexed: 01/20/2023]
Abstract
Parkinson's disease (PD) is a chronic, progressive, and second most prevalent neurological disorder affecting the motor system. It has been found that people suffering with inflammatory bowel disease (IBD) are at 22% more risk for PD. In the current study, we have established a molecular link between gut and brain. The microarray gene expression datasets of Homo sapiens were obtained from Gene Expression Omnibus Database. Major genes involved in gut-brain connection were found to be CXCR4, LRRK2, APOE, SNCA, IL6, HIF-1α, ABCA1 etc. The common biological pathways linking both the pathologies were found to be HIF-signaling, cytokines interactions, JAK-STAT pathway, cholesterol metabolism, apoptosis and CXCR4 signaling which modulates the synaptic function and neuronal survival in the mature brain. It is known that flavonoid-rich foods throughout life hold the potential to limit the inflammation, neurodegeneration and, to prevent the age-dependent cognitive impairment. Therefore, the potential receptor, CXCR4 was used further for docking with twenty-seven phytochemicals from 5 different classes of Flavonoids found in several dietary items. Docking studies of the top scoring compounds were compared with a known inhibitor (BPRCX807) of receptor CXCR4 (IC50 = 40.4 ± 8.0 nM). The study indicates that Flavan-3-ol families of flavonoids are the best fit and finest dietary supplements for improving brain health. Hence the food items like Pistachio nuts, hazelnuts, Green Tea, walnuts, etc. should be incorporated more in the diet of healthy people as well as in IBD and PD patients to prevent inflammation in gut and brain damage from oxidative stress.
Collapse
Affiliation(s)
- Nitu Dogra
- Proteomics and Translational Research Lab, Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, India, 201301
| | - Ruchi Jakhmola-Mani
- Proteomics and Translational Research Lab, Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, India, 201301
| | - Angamba Meetei Potshangbam
- Department of Biotechnology, Manipur University, Canchipur, Imphal, Manipur, India, 795003
- Research Boulevard Technologies, Greater Noida, India, 201301
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Deepshikha Pande Katare
- Proteomics and Translational Research Lab, Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, India, 201301.
| |
Collapse
|
15
|
Lin YS, Weibel J, Landolt HP, Santini F, Slawik H, Borgwardt S, Cajochen C, Reichert CF. Brain activity during a working memory task after daily caffeine intake and caffeine withdrawal: a randomized double-blind placebo-controlled trial. Sci Rep 2023; 13:1002. [PMID: 36653409 PMCID: PMC9849460 DOI: 10.1038/s41598-022-26808-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 12/20/2022] [Indexed: 01/19/2023] Open
Abstract
Acute caffeine intake has been found to increase working memory (WM)-related brain activity in healthy adults without improving behavioral performances. The impact of daily caffeine intake-a ritual shared by 80% of the population worldwide-and of its discontinuation on working memory and its neural correlates remained unknown. In this double-blind, randomized, crossover study, we examined working memory functions in 20 young healthy non-smokers (age: 26.4 ± 4.0 years; body mass index: 22.7 ± 1.4 kg/m2; and habitual caffeine intake: 474.1 ± 107.5 mg/day) in a 10-day caffeine (150 mg × 3 times/day), a 10-day placebo (3 times/day), and a withdrawal condition (9-day caffeine followed by 1-day placebo). Throughout the 10th day of each condition, participants performed four times a working memory task (N-Back, comprising 3- and 0-back), and task-related blood-oxygen-level-dependent (BOLD) activity was measured in the last session with functional magnetic resonance imaging. Compared to placebo, participants showed a higher error rate and a longer reaction time in 3- against 0-back trials in the caffeine condition; also, in the withdrawal condition we observed a higher error rate compared to placebo. However, task-related BOLD activity, i.e., an increased attention network and decreased default mode network activity in 3- versus 0-back, did not show significant differences among three conditions. Interestingly, irrespective of 3- or 0-back, BOLD activity was reduced in the right hippocampus in the caffeine condition compared to placebo. Adding to the earlier evidence showing increasing cerebral metabolic demands for WM function after acute caffeine intake, our data suggest that such demands might be impeded over daily intake and therefore result in a worse performance. Finally, the reduced hippocampal activity may reflect caffeine-associated hippocampal grey matter plasticity reported in the previous analysis. The findings of this study reveal an adapted neurocognitive response to daily caffeine exposure and highlight the importance of classifying impacts of caffeine on clinical and healthy populations.
Collapse
Affiliation(s)
- Yu-Shiuan Lin
- Centre for Chronobiology, University Psychiatric Clinics Basel, Wilhelm-Klein Strasse 27, 4002, Basel, Switzerland. .,Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland. .,Neuropsychiatry and Brain Imaging, Psychiatric Hospital of the University of Basel, Basel, Switzerland.
| | - Janine Weibel
- Centre for Chronobiology, University Psychiatric Clinics Basel, Wilhelm-Klein Strasse 27, 4002, Basel, Switzerland.,Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
| | - Hans-Peter Landolt
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Sleep and Health Zurich, University Center of Competence, University of Zurich, Zurich, Switzerland
| | - Francesco Santini
- Division of Radiological Physics, Department of Radiology, University Hospital Basel, Basel, Switzerland.,Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Helen Slawik
- Centre for Chronobiology, University Psychiatric Clinics Basel, Wilhelm-Klein Strasse 27, 4002, Basel, Switzerland.,Clinical Sleep Laboratory, Psychiatric Hospital of the University of Basel, Basel, Switzerland
| | - Stefan Borgwardt
- Neuropsychiatry and Brain Imaging, Psychiatric Hospital of the University of Basel, Basel, Switzerland
| | - Christian Cajochen
- Centre for Chronobiology, University Psychiatric Clinics Basel, Wilhelm-Klein Strasse 27, 4002, Basel, Switzerland. .,Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland.
| | - Carolin Franziska Reichert
- Centre for Chronobiology, University Psychiatric Clinics Basel, Wilhelm-Klein Strasse 27, 4002, Basel, Switzerland.,Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
| |
Collapse
|
16
|
Tang M, Sun Q, Zhang Y, Li H, Wang D, Wang Y, Wang Z. Circadian rhythm in restless legs syndrome. Front Neurol 2023; 14:1105463. [PMID: 36908590 PMCID: PMC9995399 DOI: 10.3389/fneur.2023.1105463] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
Restless legs syndrome (RLS) is a sensorimotor disorder with a obvious circadian rhythm, as its symptoms often occur or worsen only in the evening or at night. The mechanisms behind the rhythms of RLS have not yet been fully elucidated. This review explores possible causes for the circadian fluctuations of the symptomatology, including the levels of iron, dopamine, melatonin, melanocortin, and thyroid-stimulating hormone in the brain, as well as conditions such as peripheral hypoxia and microvascular function disorders. The metabolic disturbances of the substances above can create a pathological imbalance, which is further aggravated by physiological fluctuations of circadian rhythms, and results in the worsening of RLS symptoms at night. The review concludes with the suggestions for RLS treatment and research directions in the future.
Collapse
Affiliation(s)
- Mingyang Tang
- Sleep Center, Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Qingqing Sun
- Sleep Center, Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yanan Zhang
- Sleep Center, Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Huimin Li
- Sleep Center, Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Dong Wang
- Sleep Center, Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Ying Wang
- Sleep Center, Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Zan Wang
- Sleep Center, Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
17
|
The Role of the Adenosine System on Emotional and Cognitive Disturbances Induced by Ethanol Binge Drinking in the Immature Brain and the Beneficial Effects of Caffeine. Pharmaceuticals (Basel) 2022; 15:ph15111323. [DOI: 10.3390/ph15111323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
Binge drinking intake is the most common pattern of ethanol consumption by adolescents, which elicits emotional disturbances, mainly anxiety and depressive symptoms, as well as cognitive alterations. Ethanol exposure may act on the adenosine neuromodulation system by increasing adenosine levels, consequently increasing the activation of adenosine receptors in the brain. The adenosine modulation system is involved in the control of mood and memory behavior. However, there is a gap in the knowledge about the exact mechanisms related to ethanol exposure’s hazardous effects on the immature brain (i.e., during adolescence) and the role of the adenosine system thereupon. The present review attempts to provide a comprehensive picture of the role of the adenosinergic system on emotional and cognitive disturbances induced by ethanol during adolescence, exploring the potential benefits of caffeine administration in view of its action as a non-selective antagonist of adenosine receptors.
Collapse
|
18
|
Dias ALA, de Oliveira Golzio AMF, de Lima Santos BH, da Silva Stiebbe Salvadori MG, Dos Santos SG, da Silva MS, de Almeida RN, Barbosa FF. Post-learning caffeine administration improves 'what-when' and 'what-where' components of episodic-like memory in rats. Behav Brain Res 2022; 433:113982. [PMID: 35779707 DOI: 10.1016/j.bbr.2022.113982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/30/2022] [Accepted: 06/27/2022] [Indexed: 11/02/2022]
Abstract
Episodic-like memory (ELM) consists in the capacity of nonhuman animals to remember 'where' and 'when' a specific episode occurred ('what'). Previous studies have showed that Wistar rats can form an ELM, but not after a 24 h retention delay. On the other hand, it has been demonstrated that caffeine can improve episodic memory consolidation in humans. Therefore, we verified whether acute post-sample caffeine administration could improve ELM consolidation in Wistar rats, as well if it could be related to neurochemical changes in the prefrontal cortex and hippocampus - regions related to episodic-like memory processing. 46 Male Wistar Rats, approximately 3 months-old, were divided into four groups as follows: untreated (n = 11), saline (n = 11), caffeine 10 mg ∕kg i.p (n = 12); caffeine 15 mg∕kgi.p (n = 12) and tested in WWWhen/ELM task. The animals treated with caffeine in different dosages (10 mg/kg and 15 mg/kg) discriminated temporally and spatially the objects, respectively. These groups also showed a dopamine renewal rate in the hippocampus, suggesting that there was an increase in the turnover compared with the groups with no caffeine administration. We can conclude that caffeine leads to an improvement in the consolidation of the temporal ('what-when') and spatial ('what-where') aspects of episodic-like memory.
Collapse
Affiliation(s)
| | | | | | - Mirian Graciela da Silva Stiebbe Salvadori
- Departamento de Psicologia, Programa de Pós-Graduação em Neurociência Cognitiva e Comportamento, Centro de Ciências Humanas, Letras e Artes, Universidade Federal da Paraíba, Brazil
| | - Sócrates Golzio Dos Santos
- Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal da Paraíba, Brazil
| | - Marcelo Sobral da Silva
- Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal da Paraíba, Brazil
| | - Reinaldo Nóbrega de Almeida
- Programa de Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, Brazil
| | - Flavio Freitas Barbosa
- Departamento de Psicologia, Programa de Pós-Graduação em Neurociência Cognitiva e Comportamento, Centro de Ciências Humanas, Letras e Artes, Universidade Federal da Paraíba, Brazil.
| |
Collapse
|
19
|
Casadó-Anguera V, Casadó V. Unmasking allosteric binding sites: Novel targets for GPCR drug discovery. Expert Opin Drug Discov 2022; 17:897-923. [PMID: 35649692 DOI: 10.1080/17460441.2022.2085684] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Unexpected non-apparent and hidden allosteric binding sites are non-classical and non-apparent allosteric centers in 3-D X-ray protein structures until orthosteric or allosteric ligands bind to them. The orthosteric center of one protomer that modulates binding centers of the other protomers within an oligomer is also an unexpected allosteric site. Furthermore, another partner protein can also produce these effects, acting as an unexpected allosteric modulator. AREAS COVERED This review summarizes both classical and non-classical allosterism. The authors focus on G protein-coupled receptor (GPCR) oligomers as a paradigm of allosteric molecules. Moreover, they show several examples of unexpected allosteric sites such as hidden allosteric sites in a protomer that appear after the interaction with other molecules and the allosterism exerted between orthosteric sites within GPCR oligomer, emphasizing on the allosteric modulations that can occur between binding sites. EXPERT OPINION The study of these new non-classical allosteric sites will expand the diversity of allosteric control on the function of orthosteric sites within proteins, whether GPCRs or other receptors, enzymes or transporters. Moreover, the design of new drugs targeting these hidden allosteric sites or already known orthosteric sites acting as allosteric sites in protein homo- or hetero-oligomers will increase the therapeutic potential of allosterism.
Collapse
Affiliation(s)
- Verònica Casadó-Anguera
- Laboratory of Molecular Neuropharmacology, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, and Institute of Biomedicine of the Universitat de Barcelona, Barcelona, Spain.,Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Vicent Casadó
- Laboratory of Molecular Neuropharmacology, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, and Institute of Biomedicine of the Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
20
|
Reichert CF, Deboer T, Landolt HP. Adenosine, caffeine, and sleep-wake regulation: state of the science and perspectives. J Sleep Res 2022; 31:e13597. [PMID: 35575450 PMCID: PMC9541543 DOI: 10.1111/jsr.13597] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 01/11/2023]
Abstract
For hundreds of years, mankind has been influencing its sleep and waking state through the adenosinergic system. For ~100 years now, systematic research has been performed, first started by testing the effects of different dosages of caffeine on sleep and waking behaviour. About 70 years ago, adenosine itself entered the picture as a possible ligand of the receptors where caffeine hooks on as an antagonist to reduce sleepiness. Since the scientific demonstration that this is indeed the case, progress has been fast. Today, adenosine is widely accepted as an endogenous sleep‐regulatory substance. In this review, we discuss the current state of the science in model organisms and humans on the working mechanisms of adenosine and caffeine on sleep. We critically investigate the evidence for a direct involvement in sleep homeostatic mechanisms and whether the effects of caffeine on sleep differ between acute intake and chronic consumption. In addition, we review the more recent evidence that adenosine levels may also influence the functioning of the circadian clock and address the question of whether sleep homeostasis and the circadian clock may interact through adenosinergic signalling. In the final section, we discuss the perspectives of possible clinical applications of the accumulated knowledge over the last century that may improve sleep‐related disorders. We conclude our review by highlighting some open questions that need to be answered, to better understand how adenosine and caffeine exactly regulate and influence sleep.
Collapse
Affiliation(s)
- Carolin Franziska Reichert
- Centre for Chronobiology, University Psychiatric Clinics Basel, Basel, Switzerland.,Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland.,Center for Affective, Stress, and Sleep Disorders, University Psychiatric Clinics Basel, Basel, Switzerland
| | - Tom Deboer
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Hans-Peter Landolt
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland.,Sleep & Health Zürich, University Center of Competence, University of Zürich, Zürich, Switzerland
| |
Collapse
|
21
|
Fernández-Dueñas V, Bonaventura J, Aso E, Luján R, Ferré S, Ciruela F. Overcoming the Challenges of Detecting GPCR Oligomerization in the Brain. Curr Neuropharmacol 2022; 20:1035-1045. [PMID: 34736381 PMCID: PMC9886828 DOI: 10.2174/1570159x19666211104145727] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/14/2021] [Accepted: 10/14/2021] [Indexed: 11/22/2022] Open
Abstract
G protein-coupled receptors (GPCRs) constitute the largest group of membrane receptor proteins controlling brain activity. Accordingly, GPCRs are the main target of commercial drugs for most neurological and neuropsychiatric disorders. One of the mechanisms by which GPCRs regulate neuronal function is by homo- and heteromerization, with the establishment of direct protein-protein interactions between the same and different GPCRs. The occurrence of GPCR homo- and heteromers in artificial systems is generally well accepted, but more specific methods are necessary to address GPCR oligomerization in the brain. Here, we revise some of the techniques that have mostly contributed to reveal GPCR oligomers in native tissue, which include immunogold electron microscopy, proximity ligation assay (PLA), resonance energy transfer (RET) between fluorescent ligands and the Amplified Luminescent Proximity Homogeneous Assay (ALPHA). Of note, we use the archetypical GPCR oligomer, the adenosine A2A receptor (A2AR)-dopamine D2 receptor (D2R) heteromer as an example to illustrate the implementation of these techniques, which can allow visualizing GPCR oligomers in the human brain under normal and pathological conditions. Indeed, GPCR oligomerization may be involved in the pathophysiology of neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Víctor Fernández-Dueñas
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L’Hospitalet de Llobregat, Spain;,Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, 08907 L’Hospitalet de Llobregat, Spain;,Address correspondence to these authors at the Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L’Hospitalet de Llobregat, Spain; E-mails: ,
| | - Jordi Bonaventura
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L’Hospitalet de Llobregat, Spain;,Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, 08907 L’Hospitalet de Llobregat, Spain
| | - Ester Aso
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L’Hospitalet de Llobregat, Spain;,Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, 08907 L’Hospitalet de Llobregat, Spain
| | - Rafael Luján
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
| | - Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L’Hospitalet de Llobregat, Spain;,Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, 08907 L’Hospitalet de Llobregat, Spain;,Address correspondence to these authors at the Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L’Hospitalet de Llobregat, Spain; E-mails: ,
| |
Collapse
|
22
|
Mori A, Chen JF, Uchida S, Durlach C, King SM, Jenner P. The Pharmacological Potential of Adenosine A 2A Receptor Antagonists for Treating Parkinson's Disease. Molecules 2022; 27:2366. [PMID: 35408767 PMCID: PMC9000505 DOI: 10.3390/molecules27072366] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 02/07/2023] Open
Abstract
The adenosine A2A receptor subtype is recognized as a non-dopaminergic pharmacological target for the treatment of neurodegenerative disorders, notably Parkinson's disease (PD). The selective A2A receptor antagonist istradefylline is approved in the US and Japan as an adjunctive treatment to levodopa/decarboxylase inhibitors in adults with PD experiencing OFF episodes or a wearing-off phenomenon; however, the full potential of this drug class remains to be explored. In this article, we review the pharmacology of adenosine A2A receptor antagonists from the perspective of the treatment of both motor and non-motor symptoms of PD and their potential for disease modification.
Collapse
Affiliation(s)
- Akihisa Mori
- Kyowa Kirin Co., Ltd., Tokyo 100-0004, Japan; (A.M.); (S.U.)
| | - Jiang-Fan Chen
- Molecular Neuropharmacology Laboratory, Wenzhou Medical University, Wenzhou 325015, China;
| | - Shinichi Uchida
- Kyowa Kirin Co., Ltd., Tokyo 100-0004, Japan; (A.M.); (S.U.)
| | | | | | - Peter Jenner
- Institute of Pharmaceutical Science, Kings College London, London SE1 9NH, UK
| |
Collapse
|
23
|
Ostrom KF, LaVigne JE, Brust TF, Seifert R, Dessauer CW, Watts VJ, Ostrom RS. Physiological roles of mammalian transmembrane adenylyl cyclase isoforms. Physiol Rev 2022; 102:815-857. [PMID: 34698552 PMCID: PMC8759965 DOI: 10.1152/physrev.00013.2021] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/20/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
Adenylyl cyclases (ACs) catalyze the conversion of ATP to the ubiquitous second messenger cAMP. Mammals possess nine isoforms of transmembrane ACs, dubbed AC1-9, that serve as major effector enzymes of G protein-coupled receptors (GPCRs). The transmembrane ACs display varying expression patterns across tissues, giving the potential for them to have a wide array of physiological roles. Cells express multiple AC isoforms, implying that ACs have redundant functions. Furthermore, all transmembrane ACs are activated by Gαs, so it was long assumed that all ACs are activated by Gαs-coupled GPCRs. AC isoforms partition to different microdomains of the plasma membrane and form prearranged signaling complexes with specific GPCRs that contribute to cAMP signaling compartments. This compartmentation allows for a diversity of cellular and physiological responses by enabling unique signaling events to be triggered by different pools of cAMP. Isoform-specific pharmacological activators or inhibitors are lacking for most ACs, making knockdown and overexpression the primary tools for examining the physiological roles of a given isoform. Much progress has been made in understanding the physiological effects mediated through individual transmembrane ACs. GPCR-AC-cAMP signaling pathways play significant roles in regulating functions of every cell and tissue, so understanding each AC isoform's role holds potential for uncovering new approaches for treating a vast array of pathophysiological conditions.
Collapse
Affiliation(s)
| | - Justin E LaVigne
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
| | - Tarsis F Brust
- Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, West Palm Beach, Florida
| | - Roland Seifert
- Institute of Pharmacology, Hannover Medical School, Hannover, Germany
| | - Carmen W Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas
| | - Val J Watts
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
- Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana
| | - Rennolds S Ostrom
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California
| |
Collapse
|
24
|
Glial Purinergic Signaling-Mediated Oxidative Stress (GPOS) in Neuropsychiatric Disorders. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1075440. [PMID: 35281471 PMCID: PMC8916856 DOI: 10.1155/2022/1075440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 01/21/2022] [Accepted: 02/09/2022] [Indexed: 11/17/2022]
Abstract
Oxidative stress (OS) has been implicated in the progression of multiple neuropsychiatric disorders, including schizophrenia (SZ), major depressive disorder (MDD), bipolar disorder, and autism. However, whether glial purinergic signaling interaction with oxidative/antioxidative system displays an important role in neuropsychiatric disorders is still unclear. In this review, we firstly summarize the oxidative/antioxidative pathways shared in different glial cells and highlight the cell type-specific difference in response to OS. Then, we collect the evidence showing the regulation of purinergic signaling in OS with an emphasis on adenosine and its receptors, P2Y1 receptor in the P2Y family and P2X7receptor in the P2X family. Available data shows that the activation of P1 receptors and P2X accelerates the OS; reversely, the activation of the P2Y family (P2Y1) causes protective effect against OS. Finally, we discuss current findings demonstrating the contribution of the purinergic signaling system to neuropsychiatric disorders and point out the potential role of OS in this process to propose a “glial purinergic-oxidative stress” (“GPOS”) hypothesis for future development of therapeutic strategies against a variety of neuropsychiatric disorders.
Collapse
|
25
|
Ferré S, Ciruela F, Dessauer CW, González-Maeso J, Hébert TE, Jockers R, Logothetis DE, Pardo L. G protein-coupled receptor-effector macromolecular membrane assemblies (GEMMAs). Pharmacol Ther 2022; 231:107977. [PMID: 34480967 PMCID: PMC9375844 DOI: 10.1016/j.pharmthera.2021.107977] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 08/11/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022]
Abstract
G protein-coupled receptors (GPCRs) are the largest group of receptors involved in cellular signaling across the plasma membrane and a major class of drug targets. The canonical model for GPCR signaling involves three components - the GPCR, a heterotrimeric G protein and a proximal plasma membrane effector - that have been generally thought to be freely mobile molecules able to interact by 'collision coupling'. Here, we synthesize evidence that supports the existence of GPCR-effector macromolecular membrane assemblies (GEMMAs) comprised of specific GPCRs, G proteins, plasma membrane effector molecules and other associated transmembrane proteins that are pre-assembled prior to receptor activation by agonists, which then leads to subsequent rearrangement of the GEMMA components. The GEMMA concept offers an alternative and complementary model to the canonical collision-coupling model, allowing more efficient interactions between specific signaling components, as well as the integration of the concept of GPCR oligomerization as well as GPCR interactions with orphan receptors, truncated GPCRs and other membrane-localized GPCR-associated proteins. Collision-coupling and pre-assembled mechanisms are not exclusive and likely both operate in the cell, providing a spectrum of signaling modalities which explains the differential properties of a multitude of GPCRs in their different cellular environments. Here, we explore the unique pharmacological characteristics of individual GEMMAs, which could provide new opportunities to therapeutically modulate GPCR signaling.
Collapse
Affiliation(s)
- Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Addiction, Intramural Research Program, NIH, DHHS, Baltimore, MD, USA.
| | - Francisco Ciruela
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, IDIBELL, University of Barcelona, L’Hospitalet de Llobregat, Spain
| | - Carmen W. Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Javier González-Maeso
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Terence E. Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec
| | - Ralf Jockers
- University of Paris, Institute Cochin, INSERM, CNRS, Paris, France
| | - Diomedes E. Logothetis
- Laboratory of Electrophysiology, Departments of Pharmaceutical Sciences, Chemistry and Chemical Biology and Center for Drug Discovery, School of Pharmacy at the Bouvé College of Health Sciences and College of Science, Northeastern University, Boston, Massachusetts, USA
| | - Leonardo Pardo
- Laboratory of Computational Medicine, Biostatistics Unit, Faculty of Medicine, Autonomous University of Barcelona, Bellaterra, Spain
| |
Collapse
|
26
|
Rodrigues MS, Ferreira SG, Quiroz C, Earley CJ, García-Borreguero D, Cunha RA, Ciruela F, Köfalvi A, Ferré S. Brain Iron Deficiency Changes the Stoichiometry of Adenosine Receptor Subtypes in Cortico-Striatal Terminals: Implications for Restless Legs Syndrome. Molecules 2022; 27:1489. [PMID: 35268590 PMCID: PMC8911604 DOI: 10.3390/molecules27051489] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/19/2022] [Accepted: 02/21/2022] [Indexed: 01/01/2023] Open
Abstract
Brain iron deficiency (BID) constitutes a primary pathophysiological mechanism in restless legs syndrome (RLS). BID in rodents has been widely used as an animal model of RLS, since it recapitulates key neurochemical changes reported in RLS patients and shows an RLS-like behavioral phenotype. Previous studies with the BID-rodent model of RLS demonstrated increased sensitivity of cortical pyramidal cells to release glutamate from their striatal nerve terminals driving striatal circuits, a correlative finding of the cortical motor hyperexcitability of RLS patients. It was also found that BID in rodents leads to changes in the adenosinergic system, a downregulation of the inhibitory adenosine A1 receptors (A1Rs) and upregulation of the excitatory adenosine A2A receptors (A2ARs). It was then hypothesized, but not proven, that the BID-induced increased sensitivity of cortico-striatal glutamatergic terminals could be induced by a change in A1R/A2AR stoichiometry in favor of A2ARs. Here, we used a newly developed FACS-based synaptometric analysis to compare the relative abundance on A1Rs and A2ARs in cortico-striatal and thalamo-striatal glutamatergic terminals (labeled with vesicular glutamate transporters VGLUT1 and VGLUT2, respectively) of control and BID rats. It could be demonstrated that BID (determined by measuring transferrin receptor density in the brain) is associated with a selective decrease in the A1R/A2AR ratio in VGLUT1 positive-striatal terminals.
Collapse
Affiliation(s)
- Matilde S. Rodrigues
- CNC-Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; (M.S.R.); (S.G.F.); (R.A.C.); (A.K.)
| | - Samira G. Ferreira
- CNC-Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; (M.S.R.); (S.G.F.); (R.A.C.); (A.K.)
| | - César Quiroz
- Integrative Neurobiology Section, National Institute on Drug Abuse, Baltimore, MD 21224, USA;
| | | | | | - Rodrigo A. Cunha
- CNC-Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; (M.S.R.); (S.G.F.); (R.A.C.); (A.K.)
- Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L’Hospitalet de Llobregat, Spain;
- Neuropharmacology and Pain Group, Neuroscience Program, Institut d’Investigació Biomèdica de Belvitge, Idibell, 08907 L’Hospitalet de Llobregat, Spain
| | - Attila Köfalvi
- CNC-Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; (M.S.R.); (S.G.F.); (R.A.C.); (A.K.)
| | - Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Baltimore, MD 21224, USA;
| |
Collapse
|
27
|
Mao LM, Demehri S, Wang JQ. Upregulation of Src Family Tyrosine Kinases in the Rat Striatum by Adenosine A 2A Receptors. J Mol Neurosci 2022; 72:802-811. [PMID: 35041190 PMCID: PMC8986616 DOI: 10.1007/s12031-021-01961-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 12/20/2021] [Indexed: 11/25/2022]
Abstract
Adenosine A2A receptors are Golf-coupled receptors and are predominantly expressed in the striatum of mammalian brains. As a mostly postsynaptic receptor, A2A receptors are implicated in the regulation of a variety of intracellular signaling pathways in striatopallidal output neurons and are linked to the pathogenesis of various neuropsychiatric and neurological disorders. This study investigated the possible role of A2A receptors in the modulation of the Src family kinase (SFK) in the adult rat striatum. In acutely prepared striatal slices, adding the A2A receptor agonist PSB-0777 induced a significant increase in phosphorylation of SFKs at a conserved autophosphorylation site (Y416) in the caudate putamen (CPu). This increase was also seen in the nucleus accumbens (NAc). Another A2A agonist CGS-21680 showed the similar ability to elevate SFK Y416 phosphorylation in the striatum. Treatment with the A2A receptor antagonist KW-6002 blocked the effect of PSB-0777 on SFK Y416 phosphorylation. In addition, PSB-0777 enhanced kinase activity of two key SFK members (Src and Fyn) immunoprecipitated from the striatum. These data demonstrate a positive linkage from A2A receptors to the SFK signaling pathway in striatal neurons. Activation of A2A receptors leads to the upregulation of phosphorylation of SFKs (Src and Fyn) at an activation-associated autophosphorylation site and kinase activity of these SFK members.
Collapse
Affiliation(s)
- Li-Min Mao
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, 64108, USA
| | - Shannon Demehri
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, 64108, USA
| | - John Q Wang
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, 64108, USA.
- Department of Anesthesiology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, 64108, USA.
| |
Collapse
|
28
|
Ferré S, Belcher AM, Bonaventura J, Quiroz C, Sánchez-Soto M, Casadó-Anguera V, Cai NS, Moreno E, Boateng CA, Keck TM, Florán B, Earley CJ, Ciruela F, Casadó V, Rubinstein M, Volkow ND. Functional and pharmacological role of the dopamine D 4 receptor and its polymorphic variants. Front Endocrinol (Lausanne) 2022; 13:1014678. [PMID: 36267569 PMCID: PMC9578002 DOI: 10.3389/fendo.2022.1014678] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
The functional and pharmacological significance of the dopamine D4 receptor (D4R) has remained the least well understood of all the dopamine receptor subtypes. Even more enigmatic has been the role of the very prevalent human DRD4 gene polymorphisms in the region that encodes the third intracellular loop of the receptor. The most common polymorphisms encode a D4R with 4 or 7 repeats of a proline-rich sequence of 16 amino acids (D4.4R and D4.7R). DRD4 polymorphisms have been associated with individual differences linked to impulse control-related neuropsychiatric disorders, with the most consistent associations established between the gene encoding D4.7R and attention-deficit hyperactivity disorder (ADHD) and substance use disorders. The function of D4R and its polymorphic variants is being revealed by addressing the role of receptor heteromerization and the relatively avidity of norepinephrine for D4R. We review the evidence conveying a significant and differential role of D4.4R and D4.7R in the dopaminergic and noradrenergic modulation of the frontal cortico-striatal pyramidal neuron, with implications for the moderation of constructs of impulsivity as personality traits. This differential role depends on their ability to confer different properties to adrenergic α2A receptor (α2AR)-D4R heteromers and dopamine D2 receptor (D2R)-D4R heteromers, preferentially localized in the perisomatic region of the frontal cortical pyramidal neuron and its striatal terminals, respectively. We also review the evidence to support the D4R as a therapeutic target for ADHD and other impulse-control disorders, as well as for restless legs syndrome.
Collapse
Affiliation(s)
- Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes on Drug Abuse, Baltimore, MD, United States
- *Correspondence: Sergi Ferré,
| | - Annabelle M. Belcher
- Division of Addiction Research and Treatment, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jordi Bonaventura
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes on Drug Abuse, Baltimore, MD, United States
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, Spain
| | - César Quiroz
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes on Drug Abuse, Baltimore, MD, United States
| | - Marta Sánchez-Soto
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes on Drug Abuse, Baltimore, MD, United States
| | - Verònica Casadó-Anguera
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
| | - Ning-Sheng Cai
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes on Drug Abuse, Baltimore, MD, United States
| | - Estefanía Moreno
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
| | - Comfort A. Boateng
- Department of Basic Pharmaceutical Sciences, Fred Wilson School of Pharmacy, High Point, NC, United States
| | - Thomas M. Keck
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, NJ, United States
| | - Benjamín Florán
- Departament of Physiology, Biophysics and Neurosciences, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Christopher J. Earley
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, Spain
| | - Vicent Casadó
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
| | - Marcelo Rubinstein
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas and, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nora D. Volkow
- National Institute on Drug Abuse, National Institutes of Health, Rockville, MD, United States
| |
Collapse
|
29
|
Akhmadulina A, Chimagomedova A, Levin O, Poluektov M. Clinical phenomenology and pathophysiology of daytime and nighttime imperative movements. Zh Nevrol Psikhiatr Im S S Korsakova 2022; 122:67-72. [DOI: 10.17116/jnevro202212211267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
30
|
Abstract
Restless legs syndrome (RLS) is a common sensorimotor disorder characterized by an urge to move that appears during rest or is exacerbated by rest, that occurs in the evening or night and that disappears during movement or is improved by movement. Symptoms vary considerably in age at onset, frequency and severity, with severe forms affecting sleep, quality of life and mood. Patients with RLS often display periodic leg movements during sleep or resting wakefulness. RLS is considered to be a complex condition in which predisposing genetic factors, environmental factors and comorbidities contribute to the expression of the disorder. RLS occurs alone or with comorbidities, for example, iron deficiency and kidney disease, but also with cardiovascular diseases, diabetes mellitus and neurological, rheumatological and respiratory disorders. The pathophysiology is still unclear, with the involvement of brain iron deficiency, dysfunction in the dopaminergic and nociceptive systems and altered adenosine and glutamatergic pathways as hypotheses being investigated. RLS is poorly recognized by physicians and it is accordingly often incorrectly diagnosed and managed. Treatment guidelines recommend initiation of therapy with low doses of dopamine agonists or α2δ ligands in severe forms. Although dopaminergic treatment is initially highly effective, its long-term use can result in a serious worsening of symptoms known as augmentation. Other treatments include opioids and iron preparations.
Collapse
|
31
|
Selective Manipulation of G-Protein γ 7 Subunit in Mice Provides New Insights into Striatal Control of Motor Behavior. J Neurosci 2021; 41:9065-9081. [PMID: 34544837 DOI: 10.1523/jneurosci.1211-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/26/2021] [Accepted: 09/11/2021] [Indexed: 01/15/2023] Open
Abstract
Stimulatory coupling of dopamine D1 (D1R) and adenosine A2A receptors (A2AR) to adenylyl cyclase within the striatum is mediated through a specific Gαolfβ2γ7 heterotrimer to ultimately modulate motor behaviors. To dissect the individual roles of the Gαolfβ2γ7 heterotrimer in different populations of medium spiny neurons (MSNs), we produced and characterized conditional mouse models, in which the Gng7 gene was deleted in either the D1R- or A2AR/D2R-expressing MSNs. We show that conditional loss of γ7 disrupts the cell type-specific assembly of the Gαolfβ2γ7 heterotrimer, thereby identifying its circumscribed roles acting downstream of either the D1Rs or A2ARs in coordinating motor behaviors, including in vivo responses to psychostimulants. We reveal that Gαolfβ2γ7/cAMP signal in D1R-MSNs does not impact spontaneous and amphetamine-induced locomotor behaviors in male and female mice, while its loss in A2AR/D2R-MSNs results in a hyperlocomotor phenotype and enhanced locomotor response to amphetamine. Additionally, Gαolfβ2γ7/cAMP signal in either D1R- or A2AR/D2R-expressing MSNs is not required for the activation of PKA signaling by amphetamine. Finally, we show that Gαolfβ2γ7 signaling acting downstream of D1Rs is selectively implicated in the acute locomotor-enhancing effects of morphine. Collectively, these results support the general notion that receptors use specific Gαβγ proteins to direct the fidelity of downstream signaling pathways and to elicit a diverse repertoire of cellular functions. Specifically, these findings highlight the critical role for the γ7 protein in determining the cellular level, and hence, the function of the Gαolfβ2γ7 heterotrimer in several disease states associated with dysfunctional striatal signaling.SIGNIFICANCE STATEMENT Dysfunction or imbalance of cAMP signaling in the striatum has been linked to several neurologic and neuropsychiatric disorders, including Parkinson's disease, dystonia, schizophrenia, and drug addiction. By genetically targeting the γ7 subunit in distinct striatal neuronal subpopulations in mice, we demonstrate that the formation and function of the Gαolfβ2γ7 heterotrimer, which represents the rate-limiting step for cAMP production in the striatum, is selectively disrupted. Furthermore, we reveal cell type-specific roles for Gαolfβ2γ7-mediated cAMP production in the control of spontaneous locomotion as well as behavioral and molecular responses to psychostimulants. Our findings identify the γ7 protein as a novel therapeutic target for disease states associated with dysfunctional striatal cAMP signaling.
Collapse
|
32
|
Klotz L, Enz R. MGluR7 is a presynaptic metabotropic glutamate receptor at ribbon synapses of inner hair cells. FASEB J 2021; 35:e21855. [PMID: 34644430 DOI: 10.1096/fj.202100672r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/15/2021] [Accepted: 07/29/2021] [Indexed: 01/17/2023]
Abstract
Glutamate is the most pivotal excitatory neurotransmitter in the central nervous system. Metabotropic glutamate receptors (mGluRs) dimerize and can couple to inhibitory intracellular signal cascades, thereby protecting glutamatergic neurons from excessive excitation and cell death. MGluR7 is correlated with age-related hearing deficits and noise-induced hearing loss; however its exact localization in the cochlea is unknown. Here, we analyzed the expression and localization of mGluR7a and mGluR7b in mouse cochlear wholemounts in detail, using confocal microscopy and 3D reconstructions. We observed a presynaptic localization of mGluR7a at inner hair cells (IHCs), close to the synaptic ribbon. To detect mGluR7b, newly generated antibodies were characterized and showed co-localization with mGluR7a at IHC ribbon synapses. Compared to the number of synaptic ribbons, the numbers of mGluR7a and mGluR7b puncta were reduced at higher frequencies (48 to 64 kHz) and in older animals (6 and 12 months). Previously, we reported a presynaptic localization of mGluR4 and mGluR8b at this synapse type. This enables the possibility for the formation of homo- and/or heterodimeric receptors composed of mGluR4, mGluR7a, mGluR7b and mGluR8b at IHC ribbon synapses. These receptor complexes might represent new molecular targets suited for pharmacological concepts to protect the cochlea against noxious stimuli and excitotoxicity.
Collapse
Affiliation(s)
- Lisa Klotz
- Institute for Biochemistry, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Ralf Enz
- Institute for Biochemistry, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
33
|
He Y, Li Y, Pu Z, Chen M, Gao Y, Chen L, Ruan Y, Pan X, Zhou Y, Ge Y, Zhou J, Zheng W, Huang Z, Li Z, Chen JF. Striatopallidal Pathway Distinctly Modulates Goal-Directed Valuation and Acquisition of Instrumental Behavior via Striatopallidal Output Projections. Cereb Cortex 2021; 30:1366-1381. [PMID: 31690946 DOI: 10.1093/cercor/bhz172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 07/03/2019] [Accepted: 07/03/2019] [Indexed: 12/19/2022] Open
Abstract
The striatopallidal pathway is specialized for control of motor and motivational behaviors, but its causal role in striatal control of instrumental learning remains undefined (partly due to the confounding motor effects). Here, we leveraged the transient and "time-locked" optogenetic manipulations with the reward delivery to minimize motor confounding effect, to better define the striatopallidal control of instrumental behaviors. Optogenetic (Arch) silencing of the striatopallidal pathway in the dorsomedial striatum (DMS) and dorsolateral striatum (DLS) promoted goal-directed and habitual behaviors, respectively, without affecting acquisition of instrumental behaviors, indicating striatopallidal pathway suppression of instrumental behaviors under physiological condition. Conversely, striatopallidal pathway activation mainly affected the acquisition of instrumental behaviors with the acquisition suppression achieved by either optogenetic (ChR2) or chemicogenetic (hM3q) activation, by strong (10 mW, but not weak 1 mW) optogenetic activation, by the time-locked (but not random) optogenetic activation with the reward and by the DMS (but not DLS) striatopallidal pathway. Lastly, striatopallidal pathway modulated instrumental behaviors through striatopallidal output projections into the external globus pallidus (GPe) since optogenetic activation of the striatopallidal pathway in the DMS and of the striatopallidal output projections in the GPe similarly suppressed goal-directed behavior. Thus, the striatopallidal pathway confers distinctive and inhibitory controls of animal's sensitivity to goal-directed valuation and acquisition of instrumental behaviors under normal and over-activation conditions, through the output projections into GPe.
Collapse
Affiliation(s)
- Yan He
- School of Optometry and Ophthalmology and Eye Hospital, The Institute of Molecular Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yan Li
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Zhilan Pu
- School of Optometry and Ophthalmology and Eye Hospital, The Institute of Molecular Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Mozi Chen
- School of Optometry and Ophthalmology and Eye Hospital, The Institute of Molecular Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Ying Gao
- School of Optometry and Ophthalmology and Eye Hospital, The Institute of Molecular Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Li Chen
- Department of Pharmacology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Yang Ruan
- School of Optometry and Ophthalmology and Eye Hospital, The Institute of Molecular Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Xinran Pan
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yuling Zhou
- School of Optometry and Ophthalmology and Eye Hospital, The Institute of Molecular Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yuanyuan Ge
- School of Optometry and Ophthalmology and Eye Hospital, The Institute of Molecular Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Jianhong Zhou
- School of Optometry and Ophthalmology and Eye Hospital, The Institute of Molecular Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Wu Zheng
- School of Optometry and Ophthalmology and Eye Hospital, The Institute of Molecular Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Zhili Huang
- Department of Pharmacology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Zhihui Li
- School of Optometry and Ophthalmology and Eye Hospital, The Institute of Molecular Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Jiang-Fan Chen
- School of Optometry and Ophthalmology and Eye Hospital, The Institute of Molecular Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| |
Collapse
|
34
|
Ferré S, Guitart X, Quiroz C, Rea W, García-Malo C, Garcia-Borreguero D, Allen RP, Earley CJ. Akathisia and Restless Legs Syndrome: Solving the Dopaminergic Paradox. Sleep Med Clin 2021; 16:249-267. [PMID: 33985651 DOI: 10.1016/j.jsmc.2021.02.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Akathisia is an urgent need to move that is associated with treatment with dopamine receptor blocking agents (DRBAs) and with restless legs syndrome (RLS). The pathogenetic mechanism of akathisia has not been resolved. This article proposes that it involves an increased presynaptic dopaminergic transmission in the ventral striatum and concomitant strong activation of postsynaptic dopamine D1 receptors, which form complexes (heteromers) with dopamine D3 and adenosine A1 receptors. It also proposes that in DRBA-induced akathisia, increased dopamine release depends on inactivation of autoreceptors, whereas in RLS it depends on a brain iron deficiency-induced down-regulation of striatal presynaptic A1 receptors.
Collapse
Affiliation(s)
- Sergi Ferré
- Integrative Neurobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Triad Building, 333 Cassell Drive, Baltimore, MD 21224, USA.
| | - Xavier Guitart
- Integrative Neurobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Triad Building, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - César Quiroz
- Integrative Neurobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Triad Building, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - William Rea
- Integrative Neurobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Triad Building, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Celia García-Malo
- Sleep Research Institute, Paseo de la Habana 151, Madrid 28036, Spain
| | | | - Richard P Allen
- Department of Neurology, Johns Hopkins University, Johns Hopkins Bayview Medical Center, 5501 Hopkins Bayview Circle, Baltimore, MD 21224, USA
| | - Christopher J Earley
- Department of Neurology, Johns Hopkins University, Johns Hopkins Bayview Medical Center, 5501 Hopkins Bayview Circle, Baltimore, MD 21224, USA
| |
Collapse
|
35
|
Misganaw D. Heteromerization of dopaminergic receptors in the brain: Pharmacological implications. Pharmacol Res 2021; 170:105600. [PMID: 33836279 DOI: 10.1016/j.phrs.2021.105600] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/17/2021] [Accepted: 04/02/2021] [Indexed: 12/15/2022]
Abstract
Dopamine exerts its physiological effects through two subtypes of receptors, i.e. the receptors of the D1 family (D1R and D5R) and the D2 family (D2R, D3R, and D4R), which differ in their pattern of distribution, affinity, and signaling. The D1-like subfamily (D1R and D5R) are coupled to Gαs/olf proteins to activate adenylyl cyclase whereas the D2-like receptors are coupled to Gαi/o subunits and suppress the activity of adenylyl cyclase. Dopamine receptors are capable of forming homodimers, heterodimers, and higher-order oligomeric complexes, resulting in a change in the individual protomers' recognition, signaling, and pharmacology. Heteromerization has the potential to modify the canonical pharmacological features of individual monomeric units such as ligand affinity, activation, signaling, and cellular trafficking through allosteric interactions, reviving the field and introducing a new pharmacological target. Since heteromers are expressed and formed in a tissue-specific manner, they could provide the framework to design selective and effective drug candidates, such as brain-penetrant heterobivalent drugs and interfering peptides, with limited side effects. Therefore, heteromerization could be a promising area of pharmacology research, as it could contribute to the development of novel pharmacological interventions for dopamine dysregulated brain disorders such as addiction, schizophrenia, cognition, Parkinson's disease, and other motor-related disorders. This review is articulated based on the three criteria established by the International Union of Basic and Clinical Pharmacology for GPCR heterodimers (IUPHAR): evidence of co-localization and physical interactions in native or primary tissue, presence of a new physiological and functional property than the individual protomers, and loss of interaction and functional fingerprints upon heterodimer disruption.
Collapse
Affiliation(s)
- Desye Misganaw
- Pharmacology and Toxicology Unit, Department of Pharmacy, College of Medicine and Health Science, Wollo University, P.O. Box 1145, Dessie, Ethiopia.
| |
Collapse
|
36
|
Holstein SE, Barkell GA, Young MR. Caffeine increases alcohol self-administration, an effect that is independent of dopamine D 2 receptor function. Alcohol 2021; 91:61-73. [PMID: 33429015 DOI: 10.1016/j.alcohol.2020.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 12/14/2020] [Accepted: 12/21/2020] [Indexed: 01/09/2023]
Abstract
The rising popularity of alcohol mixed with energy drinks (AmEDs) has become a significant public health concern, with AmED users reporting higher levels of alcohol intake than non-AmED users. One mechanism proposed to explain this heightened level of alcohol intake in AmED users is that the high levels of caffeine found in energy drinks may increase the positive reinforcing properties of alcohol, an effect that may be dependent on interactions between adenosine receptor signaling pathways and the dopamine D2 receptor. Therefore, the purpose of the current study was to confirm whether caffeine does increase the positive reinforcing effects of alcohol using both fixed ratio (FR) and progressive ratio (PR) designs, and to investigate a potential role of the dopamine D2 receptor to caffeine-induced increases in alcohol self-administration. Male Long-Evans rats were trained to self-administer a sweetened alcohol solution (10% v/v alcohol + 2% w/v sucrose) on an FR2 schedule of reinforcement, and the effects of caffeine (0, 5, 10, and 20 mg/kg, i. p. [intraperitoneally]) on the maintenance of alcohol self-administration and alcohol break point were examined. Parallel experiments in rats trained to self-administer sucrose (0.8% w/v) were conducted to determine whether caffeine's reinforcement-enhancing effects extended to a non-drug reinforcer. Caffeine pretreatment (5-10 mg/kg) significantly increased sweetened alcohol self-administration and motivation for a sweetened alcohol reinforcer. However, similar increases in self-administration of a non-drug reinforcer were not observed. Contrary to our hypothesis, the D2 receptor antagonist eticlopride did not block a caffeine-induced increase in sweetened alcohol self-administration, nor did it alter caffeine-induced increases in motivation for a sweetened alcohol reinforcer. Taken together, these results support the hypothesis that caffeine increases the positive reinforcing effects of alcohol, which may explain caffeine-induced increases in alcohol intake. However, the reinforcement-enhancing effects of caffeine appear to be independent of D2 receptor function.
Collapse
Affiliation(s)
- Sarah E Holstein
- Department of Psychology, Lycoming College, One College Place, Williamsport, PA, 17701, United States.
| | - Gillian A Barkell
- Department of Psychology, Lycoming College, One College Place, Williamsport, PA, 17701, United States
| | - Megan R Young
- Department of Psychology, Lycoming College, One College Place, Williamsport, PA, 17701, United States
| |
Collapse
|
37
|
Valle-León M, Callado LF, Aso E, Cajiao-Manrique MM, Sahlholm K, López-Cano M, Soler C, Altafaj X, Watanabe M, Ferré S, Fernández-Dueñas V, Menchón JM, Ciruela F. Decreased striatal adenosine A 2A-dopamine D 2 receptor heteromerization in schizophrenia. Neuropsychopharmacology 2021; 46:665-672. [PMID: 33010795 PMCID: PMC8027896 DOI: 10.1038/s41386-020-00872-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 08/04/2020] [Accepted: 08/26/2020] [Indexed: 02/04/2023]
Abstract
According to the adenosine hypothesis of schizophrenia, the classically associated hyperdopaminergic state may be secondary to a loss of function of the adenosinergic system. Such a hypoadenosinergic state might either be due to a reduction of the extracellular levels of adenosine or alterations in the density of adenosine A2A receptors (A2ARs) or their degree of functional heteromerization with dopamine D2 receptors (D2R). In the present study, we provide preclinical and clinical evidences for this latter mechanism. Two animal models for the study of schizophrenia endophenotypes, namely the phencyclidine (PCP) mouse model and the A2AR knockout mice, were used to establish correlations between behavioural and molecular studies. In addition, a new AlphaLISA-based method was implemented to detect native A2AR-D2R heteromers in mouse and human brain. First, we observed a reduction of prepulse inhibition in A2AR knockout mice, similar to that observed in the PCP animal model of sensory gating impairment of schizophrenia, as well as a significant upregulation of striatal D2R without changes in A2AR expression in PCP-treated animals. In addition, PCP-treated animals showed a significant reduction of striatal A2AR-D2R heteromers, as demonstrated by the AlphaLISA-based method. A significant and pronounced reduction of A2AR-D2R heteromers was next demonstrated in postmortem caudate nucleus from schizophrenic subjects, even though both D2R and A2AR were upregulated. Finally, in PCP-treated animals, sub-chronic administration of haloperidol or clozapine counteracted the reduction of striatal A2AR-D2R heteromers. The degree of A2AR-D2R heteromer formation in schizophrenia might constitute a hallmark of the illness, which indeed should be further studied to establish possible correlations with chronic antipsychotic treatments.
Collapse
Affiliation(s)
- Marta Valle-León
- grid.5841.80000 0004 1937 0247Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, UB, L’Hospitalet de Llobregat, Barcelona, Spain ,grid.418284.30000 0004 0427 2257Neuropharmacology and Pain Group, Neuroscience Program, Institut d’Investigació Biomèdica de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Luis F. Callado
- grid.11480.3c0000000121671098Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain ,grid.413448.e0000 0000 9314 1427Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Instituto Salud Carlos III, Madrid, Spain ,Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Ester Aso
- grid.5841.80000 0004 1937 0247Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, UB, L’Hospitalet de Llobregat, Barcelona, Spain ,grid.418284.30000 0004 0427 2257Neuropharmacology and Pain Group, Neuroscience Program, Institut d’Investigació Biomèdica de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - María M. Cajiao-Manrique
- grid.5841.80000 0004 1937 0247Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, UB, L’Hospitalet de Llobregat, Barcelona, Spain ,grid.418284.30000 0004 0427 2257Neuropharmacology and Pain Group, Neuroscience Program, Institut d’Investigació Biomèdica de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain ,grid.412041.20000 0001 2106 639XBordeaux International Neuroscience Master, University of Bordeaux, Bordeaux, France
| | - Kristoffer Sahlholm
- grid.5841.80000 0004 1937 0247Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, UB, L’Hospitalet de Llobregat, Barcelona, Spain ,grid.418284.30000 0004 0427 2257Neuropharmacology and Pain Group, Neuroscience Program, Institut d’Investigació Biomèdica de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain ,grid.4714.60000 0004 1937 0626Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden ,grid.12650.300000 0001 1034 3451Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Marc López-Cano
- grid.5841.80000 0004 1937 0247Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, UB, L’Hospitalet de Llobregat, Barcelona, Spain ,grid.418284.30000 0004 0427 2257Neuropharmacology and Pain Group, Neuroscience Program, Institut d’Investigació Biomèdica de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Concepció Soler
- grid.418284.30000 0004 0427 2257Neuropharmacology and Pain Group, Neuroscience Program, Institut d’Investigació Biomèdica de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain ,grid.5841.80000 0004 1937 0247Immunology Unit, Faculty of Medicine and Health Sciences, Department of Pathology and Experimental Therapeutics, University of Barcelona, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Xavier Altafaj
- grid.5841.80000 0004 1937 0247Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, UB, L’Hospitalet de Llobregat, Barcelona, Spain ,grid.418284.30000 0004 0427 2257Neuropharmacology and Pain Group, Neuroscience Program, Institut d’Investigació Biomèdica de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Masahiko Watanabe
- grid.39158.360000 0001 2173 7691Department of Anatomy, Hokkaido University School of Medicine, Sapporo, 060-0818 Japan
| | - Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA.
| | - Víctor Fernández-Dueñas
- grid.5841.80000 0004 1937 0247Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, UB, L’Hospitalet de Llobregat, Barcelona, Spain ,grid.418284.30000 0004 0427 2257Neuropharmacology and Pain Group, Neuroscience Program, Institut d’Investigació Biomèdica de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - José M. Menchón
- grid.413448.e0000 0000 9314 1427Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Instituto Salud Carlos III, Madrid, Spain ,grid.411129.e0000 0000 8836 0780Department of Psychiatry, University Hospital of Bellvitge, L’Hospitalet de Llobregat, Barcelona, Spain ,grid.418284.30000 0004 0427 2257Psychiatry and Mental Health Group, Neuroscience Program, Institut d’Investigació Biomèdica de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain ,grid.5841.80000 0004 1937 0247Department of Clinical Sciences, School of Medicine, University of Barcelona, UB, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, UB, L'Hospitalet de Llobregat, Barcelona, Spain. .,Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
38
|
Qian M, Ricarte A, Wouters E, Dalton JAR, Risseeuw MDP, Giraldo J, Van Calenbergh S. Discovery of a true bivalent dopamine D 2 receptor agonist. Eur J Med Chem 2021; 212:113151. [PMID: 33450620 DOI: 10.1016/j.ejmech.2020.113151] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/06/2020] [Accepted: 12/28/2020] [Indexed: 10/22/2022]
Abstract
Employing two different alkyne-modified dopamine agonists to construct bivalent compounds via click chemistry resulted in the identification of a bivalent ligand (11c) for dopamine D2 receptor homodimer, which, compared to its parent monomeric alkyne, showed a 16-fold higher binding affinity for the dopamine D2 receptor and a 5-fold higher potency in a cAMP assay in HEK 293T cells stably expressing D2R. Molecular modeling revealed that 11c can indeed bridge the orthosteric binding sites of a D2R homodimer in a relaxed conformation via the TM5-TM6 interface and allows to largely rationalize the results of the receptor assays.
Collapse
Affiliation(s)
- Mingcheng Qian
- Department of Medicinal Chemistry, School of Pharmacy, Changzhou University, Changzhou, 213164, Jiangsu, China; Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000, Ghent, Belgium
| | - Adrián Ricarte
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain; Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain; Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigaciói InnovacióParc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Elise Wouters
- Laboratory of Toxicology, Ghent University, Ottergemsesteenweg 460, B-9000, Ghent, Belgium
| | - James A R Dalton
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain; Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain; Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigaciói InnovacióParc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Martijn D P Risseeuw
- Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000, Ghent, Belgium
| | - Jesús Giraldo
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain; Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain; Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigaciói InnovacióParc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Serge Van Calenbergh
- Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000, Ghent, Belgium.
| |
Collapse
|
39
|
Crans RAJ, Ciruela F. Dopaminergic-cholinergic imbalance in movement disorders: a role for the novel striatal dopamine D 2- muscarinic acetylcholine M 1 receptor heteromer. Neural Regen Res 2021; 16:1406-1408. [PMID: 33318429 PMCID: PMC8284294 DOI: 10.4103/1673-5374.300988] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
- René A. J Crans
- Laboratory of Toxicology, Department of Bioanalysis, Ghent University, Ghent, Belgium; Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina i Ciències de la Salut, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Francisco Ciruela
- Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina i Ciències de la Salut, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat; Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
40
|
Mori A. How do adenosine A 2A receptors regulate motor function? Parkinsonism Relat Disord 2020; 80 Suppl 1:S13-S20. [PMID: 33349575 DOI: 10.1016/j.parkreldis.2020.09.025] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 01/17/2023]
Abstract
Adenosine A2A receptor antagonism is a new therapeutic strategy in the symptomatic treatment of Parkinson's disease (PD). This review addresses how adenosine A2A receptors are involved with the control of motor function via the basal ganglia-thalamocortical circuit, and considers the anatomical localization and physiological function of the receptor, along with its ultrastructural localization in critical areas/neurons of the circuit. Based on this understanding of the functional significance of the adenosine A2A receptor in the basal ganglia, the mode of action of A2A receptor antagonists is explored in terms of the dynamic functioning of the basal ganglia and the activity of the internal circuits of the striatum in PD. Finally, the pathophysiological differences between the normal and PD states are examined to emphasize the importance of the adenosine A2A receptor.
Collapse
|
41
|
Gonçalves MCB, Glaser T, Oliveira SLBD, Ulrich H. Adenosinergic-Dopaminergic Signaling in Mood Disorders: A Mini-Review. J Caffeine Adenosine Res 2020. [DOI: 10.1089/caff.2020.0009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
| | - Talita Glaser
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | | | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| |
Collapse
|
42
|
Martel JC, Gatti McArthur S. Dopamine Receptor Subtypes, Physiology and Pharmacology: New Ligands and Concepts in Schizophrenia. Front Pharmacol 2020; 11:1003. [PMID: 32765257 PMCID: PMC7379027 DOI: 10.3389/fphar.2020.01003] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022] Open
Abstract
Dopamine receptors are widely distributed within the brain where they play critical modulator roles on motor functions, motivation and drive, as well as cognition. The identification of five genes coding for different dopamine receptor subtypes, pharmacologically grouped as D1- (D1 and D5) or D2-like (D2S, D2L, D3, and D4) has allowed the demonstration of differential receptor function in specific neurocircuits. Recent observation on dopamine receptor signaling point at dopamine-glutamate-NMDA neurobiology as the most relevant in schizophrenia and for the development of new therapies. Progress in the chemistry of D1- and D2-like receptor ligands (agonists, antagonists, and partial agonists) has provided more selective compounds possibly able to target the dopamine receptors homo and heterodimers and address different schizophrenia symptoms. Moreover, an extensive evaluation of the functional effect of these agents on dopamine receptor coupling and intracellular signaling highlights important differences that could also result in highly differentiated clinical pharmacology. The review summarizes the recent advances in the field, addressing the relevance of emerging new targets in schizophrenia in particular in relation to the dopamine - glutamate NMDA systems interactions.
Collapse
|
43
|
Salery M, Trifilieff P, Caboche J, Vanhoutte P. From Signaling Molecules to Circuits and Behaviors: Cell-Type-Specific Adaptations to Psychostimulant Exposure in the Striatum. Biol Psychiatry 2020; 87:944-953. [PMID: 31928716 DOI: 10.1016/j.biopsych.2019.11.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/30/2019] [Accepted: 11/01/2019] [Indexed: 12/17/2022]
Abstract
Addiction is characterized by a compulsive pattern of drug seeking and consumption and a high risk of relapse after withdrawal that are thought to result from persistent adaptations within brain reward circuits. Drugs of abuse increase dopamine (DA) concentration in these brain areas, including the striatum, which shapes an abnormal memory trace of drug consumption that virtually highjacks reward processing. Long-term neuronal adaptations of gamma-aminobutyric acidergic striatal projection neurons (SPNs) evoked by drugs of abuse are critical for the development of addiction. These neurons form two mostly segregated populations, depending on the DA receptor they express and their output projections, constituting the so-called direct (D1 receptor) and indirect (D2 receptor) SPN pathways. Both SPN subtypes receive converging glutamate inputs from limbic and cortical regions, encoding contextual and emotional information, together with DA, which mediates reward prediction and incentive values. DA differentially modulates the efficacy of glutamate synapses onto direct and indirect SPN pathways by recruiting distinct striatal signaling pathways, epigenetic and genetic responses likely involved in the transition from casual drug use to addiction. Herein we focus on recent studies that have assessed psychostimulant-induced alterations in a cell-type-specific manner, from remodeling of input projections to the characterization of specific molecular events in each SPN subtype and their impact on long-lasting behavioral adaptations. We discuss recent evidence revealing the complex and concerted action of both SPN populations on drug-induced behavioral responses, as these studies can contribute to the design of future strategies to alleviate specific behavioral components of addiction.
Collapse
Affiliation(s)
- Marine Salery
- Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Pierre Trifilieff
- NutriNeuro, Unité Mixte de Recherche (UMR) 1286, Institut National de la Recherche Agronomique, Bordeaux Institut Polytechnique, University of Bordeaux, Bordeaux, France
| | - Jocelyne Caboche
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Sorbonne Université, Faculty of Sciences, Paris, France; Centre National de la Recherche Scientifique, UMR8246, Paris, France; Institut National de la Santé et de la Recherche Médicale, U1130, Paris France.
| | - Peter Vanhoutte
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Sorbonne Université, Faculty of Sciences, Paris, France; Centre National de la Recherche Scientifique, UMR8246, Paris, France; Institut National de la Santé et de la Recherche Médicale, U1130, Paris France
| |
Collapse
|
44
|
Crans RAJ, Wouters E, Valle-León M, Taura J, Massari CM, Fernández-Dueñas V, Stove CP, Ciruela F. Striatal Dopamine D 2-Muscarinic Acetylcholine M 1 Receptor-Receptor Interaction in a Model of Movement Disorders. Front Pharmacol 2020; 11:194. [PMID: 32231561 PMCID: PMC7083216 DOI: 10.3389/fphar.2020.00194] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 02/11/2020] [Indexed: 12/17/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder characterized by motor control deficits, which is associated with the loss of striatal dopaminergic neurons from the substantia nigra. In parallel to dopaminergic denervation, there is an increase of acetylcholine within the striatum, resulting in a striatal dopaminergic–cholinergic neurotransmission imbalance. Currently, available PD pharmacotherapy (e.g., prodopaminergic drugs) does not reinstate the altered dopaminergic–cholinergic balance. In addition, it can eventually elicit cholinergic-related adverse effects. Here, we investigated the interplay between dopaminergic and cholinergic systems by assessing the physical and functional interaction of dopamine D2 and muscarinic acetylcholine M1 receptors (D2R and M1R, respectively), both expressed at striatopallidal medium spiny neurons. First, we provided evidence for the existence of D2R–M1R complexes via biochemical (i.e., co-immunoprecipitation) and biophysical (i.e., BRET1 and NanoBiT®) assays, performed in transiently transfected HEK293T cells. Subsequently, a D2R–M1R co-distribution in the mouse striatum was observed through double-immunofluorescence staining and AlphaLISA® immunoassay. Finally, we evaluated the functional interplay between both receptors via behavioral studies, by implementing the classical acute reserpine pharmacological animal model of experimental parkinsonism. Reserpinized mice were administered with a D2R-selective agonist (sumanirole) and/or an M1R-selective antagonist (VU0255035), and alterations in PD-related behavioral tasks (i.e., locomotor activity) were evaluated. Importantly, VU0255035 (10 mg/kg) potentiated the antiparkinsonian-like effects (i.e., increased locomotor activity and decreased catalepsy) of an ineffective sumanirole dose (3 mg/kg). Altogether, our data suggest the existence of putative striatal D2R/M1R heteromers, which might be a relevant target to manage PD motor impairments with fewer adverse effects.
Collapse
Affiliation(s)
- René A J Crans
- Laboratory of Toxicology, Department of Bioanalysis, Ghent University, Ghent, Belgium.,Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL-Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Elise Wouters
- Laboratory of Toxicology, Department of Bioanalysis, Ghent University, Ghent, Belgium
| | - Marta Valle-León
- Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL-Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Jaume Taura
- Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL-Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Caio M Massari
- Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL-Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Programa de Poìs-graduação em Bioquiìmica, Centro de Ciencias Bioloìgicas, Universidade Federal de Santa Catarina, Florianoìpolis, Brazil
| | - Víctor Fernández-Dueñas
- Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL-Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Christophe P Stove
- Laboratory of Toxicology, Department of Bioanalysis, Ghent University, Ghent, Belgium
| | - Francisco Ciruela
- Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL-Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
45
|
Exercise-Induced Adaptations to the Mouse Striatal Adenosine System. Neural Plast 2020; 2020:5859098. [PMID: 32399024 PMCID: PMC7204111 DOI: 10.1155/2020/5859098] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/18/2019] [Accepted: 12/27/2019] [Indexed: 12/13/2022] Open
Abstract
Adenosine acts as a key regulator of striatum activity, in part, through the antagonistic modulation of dopamine activity. Exercise can increase adenosine activity in the brain, which may impair dopaminergic functions in the striatum. Therefore, long-term repeated bouts of exercise may subsequently generate plasticity in striatal adenosine systems in a manner that promotes dopaminergic activity. This study investigated the effects of long-term voluntary wheel running on adenosine 1 (A1R), adenosine 2A (A2AR), dopamine 1 (D1R), and dopamine 2 (D2R) receptor protein expression in adult mouse dorsal and ventral striatum structures using immunohistochemistry. In addition, equilibrative nucleoside transporter 1 (ENT1) protein expression was examined after wheel running, as ENT1 regulates the bidirectional flux of adenosine between intra- and extracellular space. The results suggest that eight weeks of running wheel access spared age-related increases of A1R and A2AR protein concentrations across the dorsal and ventral striatal structures. Wheel running mildly reduced ENT1 protein levels in ventral striatum subregions. Moreover, wheel running mildly increased D2R protein density within striatal subregions in the dorsal medial striatum, nucleus accumbens core, and the nucleus accumbens shell. However, D1R protein expression in the striatum was unchanged by wheel running. These data suggest that exercise promotes adaptations to striatal adenosine systems. Exercise-reduced A1R and A2AR and exercise-increased D2R protein levels may contribute to improved dopaminergic signaling in the striatum. These findings may have implications for cognitive and behavioral processes, as well as motor and psychiatric diseases that involve the striatum.
Collapse
|
46
|
Ferré S, Ciruela F, Casadó V, Pardo L. Oligomerization of G protein-coupled receptors: Still doubted? PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 169:297-321. [DOI: 10.1016/bs.pmbts.2019.11.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
47
|
Yuan S, Li H, Xie J, Sun X. Quantitative Trait Module-Based Genetic Analysis of Alzheimer's Disease. Int J Mol Sci 2019; 20:E5912. [PMID: 31775305 PMCID: PMC6928939 DOI: 10.3390/ijms20235912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/21/2019] [Accepted: 11/22/2019] [Indexed: 01/02/2023] Open
Abstract
The pathological features of Alzheimer's Disease (AD) first appear in the medial temporal lobe and then in other brain structures with the development of the disease. In this work, we investigated the association between genetic loci and subcortical structure volumes of AD on 393 samples in the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort. Brain subcortical structures were clustered into modules using Pearson's correlation coefficient of volumes across all samples. Module volumes were used as quantitative traits to identify not only the main effect loci but also the interactive effect loci for each module. Thirty-five subcortical structures were clustered into five modules, each corresponding to a particular brain structure/area, including the limbic system (module I), the corpus callosum (module II), thalamus-cerebellum-brainstem-pallidum (module III), the basal ganglia neostriatum (module IV), and the ventricular system (module V). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment results indicate that the gene annotations of the five modules were distinct, with few overlaps between different modules. We identified several main effect loci and interactive effect loci for each module. All these loci are related to the function of module structures and basic biological processes such as material transport and signal transduction.
Collapse
Affiliation(s)
| | | | | | - Xiao Sun
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China; (S.Y.)
| |
Collapse
|
48
|
Hong SI, Peyton L, Chern Y, Choi DS. Novel Adenosine Analog, N6-(4-Hydroxybenzyl)-Adenosine, Dampens Alcohol Drinking and Seeking Behaviors. J Pharmacol Exp Ther 2019; 371:260-267. [PMID: 31409667 PMCID: PMC6812055 DOI: 10.1124/jpet.119.261529] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 08/09/2019] [Indexed: 12/11/2022] Open
Abstract
Adenosine signaling is associated with ethanol-related behaviors. We previously found that adenosine A2A receptor (A2AR) activation dampens ethanol drinking behaviors in equilibrative nucleoside transporter 1 (ENT1) knockout mice, and A2AR inhibition augments reward-seeking behavior in wild-type mice. The novel adenosine analog N6-(4-hydroxybenzyl)-adenosine (NHBA), which is isolated from the rhizomes of Gastrodia elata, activates A2AR and inhibits ENT1. Here, we examined the effects of NHBA on ethanol drinking in the two-bottle choice test and operant ethanol seeking behaviors. We selected mice exhibiting high ethanol drinking behavior in the two-bottle choice test. NHBA (0.1 mg/kg, i.p.) reduced ethanol drinking behavior in a limited-access 3-hour drinking session in high-consumption ethanol drinking mice, and NHBA (0.1 mg/kg, i.p.) did not alter locomotor activity in the open-field test. Operant conditioning with 10% ethanol and 10% sucrose (10E10S) reward increased zone entries and time spent in the ethanol zone, while NHBA (0.1 mg/kg, i.p.) dampened ethanol zone preference in the Y-maze. Furthermore, NHBA (0.1 mg/kg, i.p.) devalued 10E10S and 10% ethanol (10E) reward after operant conditioning with 10E10S and 10E. Taken together, NHBA through A2AR activation and ENT1 modulation may dampen ethanol drinking and seeking behaviors, suggesting that NHBA is a potential therapeutic agent for treating alcohol use disorder. SIGNIFICANCE STATEMENT: Our work highlights that A2AR activation and ENT1 inhibition by a novel adenosine analog isolated from Gastrodia elata, N6-(4-hydroxybenzyl)-adenosine, decreases ethanol drinking and seeking behaviors. We suggest that NHBA is a potential therapeutic agent to treat alcohol use disorder.
Collapse
Affiliation(s)
- Sa-Ik Hong
- Department of Molecular Pharmacology and Experimental Therapeutics (S.-I.H., L.P., D.-S.C.), Neuroscience Program (D.-S.C.), and Department of Psychiatry and Psychology (D.-S.C.), Mayo Clinic College of Medicine, Rochester, Minnesota; and Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (Y.C.)
| | - Lee Peyton
- Department of Molecular Pharmacology and Experimental Therapeutics (S.-I.H., L.P., D.-S.C.), Neuroscience Program (D.-S.C.), and Department of Psychiatry and Psychology (D.-S.C.), Mayo Clinic College of Medicine, Rochester, Minnesota; and Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (Y.C.)
| | - Yijuang Chern
- Department of Molecular Pharmacology and Experimental Therapeutics (S.-I.H., L.P., D.-S.C.), Neuroscience Program (D.-S.C.), and Department of Psychiatry and Psychology (D.-S.C.), Mayo Clinic College of Medicine, Rochester, Minnesota; and Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (Y.C.)
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics (S.-I.H., L.P., D.-S.C.), Neuroscience Program (D.-S.C.), and Department of Psychiatry and Psychology (D.-S.C.), Mayo Clinic College of Medicine, Rochester, Minnesota; and Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (Y.C.)
| |
Collapse
|
49
|
Mallozzi C, Pepponi R, Visentin S, Chiodi V, Lombroso PJ, Bader M, Popoli P, Domenici MR. The activity of the Striatal-enriched protein tyrosine phosphatase in neuronal cells is modulated by adenosine A 2A receptor. J Neurochem 2019; 152:284-298. [PMID: 31520531 DOI: 10.1111/jnc.14866] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/30/2019] [Accepted: 09/01/2019] [Indexed: 12/15/2022]
Abstract
We recently demonstrated that a tonic activation of adenosine A2A receptors (A2A Rs) is required for cocaine-induced synaptic depression and increase in the activity of STriatal-Enriched protein tyrosine Phosphatase (STEP). In this study, we elaborated on the relationship between A2A R and STEP using genetic, pharmacological, and cellular tools. We found that the activities of protein tyrosine phosphatases (PTPs), and in particular of STEP, are significantly increased in the striatum and hippocampus of a transgenic rat strain over-expressing the neuronal A2A R (NSEA2A ) with respect to wild-type (WT) rats. Moreover the selective A2A R agonist 4-[2-[[6-Amino-9-(N-ethyl-β-d-ribofuranuronamidosyl)-9H-purin-2-yl]amino]ethyl]benzenepropanoic acid hydrochloride up-regulates PTPs and STEP activities in WT but not in NSEA2A rats, while the selective A2A R antagonist 4-(-2-[7-amino-2-{2-furyl}{1,2,4}triazolo{2,3-a} {1,3,5}triazin-5-yl-amino]ethyl)phenol restores the tyrosine phosphatase activities in NSEA2A , having no effects in WT rats. In addition, while cocaine induced the activation of PTP and STEP in WT rats, it failed to increase phosphatase activity in NSEA2A rats. A2A Rs modulate STEP activity also in the SH-SY5Y neuroblastoma cell line, where a calcium-dependent calcineurin/PP1 pathway was found to play a major role. In summary, the present study identified a novel interaction between A2A R and STEP that could have important clinical implications, since STEP has emerged as key regulator of signaling pathways involved in neurodegenerative and neuropsychiatric diseases and A2A Rs are considered a promising target for the development of therapeutic strategies for different diseases of the central nervous system. Read the Editorial Highlight for this article on page 270.
Collapse
Affiliation(s)
- Cinzia Mallozzi
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | - Rita Pepponi
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Sergio Visentin
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Valentina Chiodi
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Paul J Lombroso
- Child Study Center, Yale School of Medicine, New Haven, CT, USA
| | - Michael Bader
- Max-Delbrűck-Center for Molecular Medicine, Berlin, Germany
| | - Patrizia Popoli
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Maria Rosaria Domenici
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
50
|
Ferré S, Ciruela F. Functional and Neuroprotective Role of Striatal Adenosine A 2A Receptor Heterotetramers. J Caffeine Adenosine Res 2019; 9:89-97. [PMID: 31559390 PMCID: PMC6761580 DOI: 10.1089/caff.2019.0008] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
In the striatum, adenosine A2A receptors (A2AR) are mainly expressed within the soma and dendrites of the striatopallidal neuron. A predominant proportion of these striatal postsynaptic A2AR form part of the macromolecular complexes that include A2AR-dopamine D2 receptor (D2R) heteromers, Golf and Gi/o proteins, and the effector adenylyl cyclase (AC), subtype AC5. The A2AR-D2R heteromers have a tetrameric structure, constituted by A2AR and D2R homomers. By means of reciprocal antagonistic allosteric interactions and antagonistic interactions at the effector level between adenosine and dopamine, the A2AR-D2R heterotetramer-AC5 complex acts an integrative molecular device, which determines a switch between the adenosine-facilitated activation and the dopamine-facilitated inhibition of the striatopallidal neuron. Striatal adenosine also plays an important presynaptic modulatory role, driving the function of corticostriatal terminals. This control is mediated by adenosine A1 receptors (A1R) and A2AR, which establish intermolecular interactions forming A1R-A2AR heterotetramers. Here, we review the functional role of both presynaptic and postsynaptic striatal A2AR heterotetramers as well as their possible neuroprotective role. We hypothesize that alterations in the homomer/heteromer stoichiometry (i.e., increase or decrease in the proportion of A2AR forming homomers or heteromers) are pathogenetically involved in neurological disorders, specifically in Parkinson's disease and restless legs syndrome.
Collapse
Affiliation(s)
- Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, Maryland
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine, IDIBELL, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| |
Collapse
|