1
|
Pauchon B, Beauchamps V, Gomez-Mérino D, Erblang M, Drogou C, Beers PV, Guillard M, Quiquempoix M, Léger D, Chennaoui M, Sauvet F. Caffeine Intake Alters Recovery Sleep after Sleep Deprivation. Nutrients 2024; 16:3442. [PMID: 39458438 PMCID: PMC11510014 DOI: 10.3390/nu16203442] [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: 09/23/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Caffeine is a well-known psychostimulant reputed to alleviate the deleterious effects of sleep deprivation. Nevertheless, caffeine can alter sleep duration and quality, particularly during recovery sleep. We evaluated the effects of acute caffeine intake on the duration and quality of recovery sleep following total sleep deprivation (TSD), taking into account daily caffeine consumption. METHODS Forty-one participants performed a double-blind, crossover TSD protocol (38 h of continuous wakefulness) with acute caffeine or placebo. Caffeine (2.5 mg/kg) or placebo was administered twice during continuous wakefulness (last treatment 6.5 h before bedtime for the recovery night). Polysomnographic measurements were recorded using a connected headband. RESULTS TSD was associated with a rebound in total sleep time (TST) on the recovery night (+110.2 ± 23.2 min, p < 0.001). Caffeine intake decreased this recovery TST (-30.2 ± 8.2 min p = 0.02) and the N3 sleep stage duration (-35.6 ± 23.2 min, p < 0.01). Caffeine intake altered recovery sleep continuity (increased number of long awakenings), stability (higher stage transition frequency), and organization (less time spent in complete sleep cycle) and decreased the delta power spectral density during NREM sleep. On the recovery night, habitual daily caffeine consumption was negatively correlated with TST in caffeine and placebo conditions and positively correlated with wake after sleep onset (WASO) duration and with the frequency of long (>2 min) awakenings in the caffeine condition only. CONCLUSIONS Acute caffeine intake during TSD affects nighttime recovery sleep, with an interaction with daily consumption. These results may influence advice on caffeine intake for night-shift workers. (NCT03859882).
Collapse
Affiliation(s)
- Benoit Pauchon
- Ecole du Val de Grace, 75006 Paris, France;
- Hôpital d’Instruction des Armées (HIA) Sainte Anne, 83100 Toulon, France
| | - Vincent Beauchamps
- Institut de Recherche Biomédicale des Armées (IRBA), 91190 Brétigny sur Orge, France; (V.B.)
- URP 7330 VIFASOM, Université Paris Cité, Hôtel Dieu, 75004 Paris, France
| | - Danielle Gomez-Mérino
- Institut de Recherche Biomédicale des Armées (IRBA), 91190 Brétigny sur Orge, France; (V.B.)
- URP 7330 VIFASOM, Université Paris Cité, Hôtel Dieu, 75004 Paris, France
| | | | - Catherine Drogou
- Institut de Recherche Biomédicale des Armées (IRBA), 91190 Brétigny sur Orge, France; (V.B.)
- URP 7330 VIFASOM, Université Paris Cité, Hôtel Dieu, 75004 Paris, France
| | - Pascal Van Beers
- Institut de Recherche Biomédicale des Armées (IRBA), 91190 Brétigny sur Orge, France; (V.B.)
- URP 7330 VIFASOM, Université Paris Cité, Hôtel Dieu, 75004 Paris, France
| | - Mathias Guillard
- Institut de Recherche Biomédicale des Armées (IRBA), 91190 Brétigny sur Orge, France; (V.B.)
- URP 7330 VIFASOM, Université Paris Cité, Hôtel Dieu, 75004 Paris, France
| | - Michaël Quiquempoix
- Institut de Recherche Biomédicale des Armées (IRBA), 91190 Brétigny sur Orge, France; (V.B.)
- URP 7330 VIFASOM, Université Paris Cité, Hôtel Dieu, 75004 Paris, France
| | - Damien Léger
- URP 7330 VIFASOM, Université Paris Cité, Hôtel Dieu, 75004 Paris, France
- APHP, Hôtel-Dieu, Centre du Sommeil et de la Vigilance, DMU THOROS, 75004 Paris, France
| | - Mounir Chennaoui
- Institut de Recherche Biomédicale des Armées (IRBA), 91190 Brétigny sur Orge, France; (V.B.)
- URP 7330 VIFASOM, Université Paris Cité, Hôtel Dieu, 75004 Paris, France
| | - Fabien Sauvet
- Ecole du Val de Grace, 75006 Paris, France;
- Institut de Recherche Biomédicale des Armées (IRBA), 91190 Brétigny sur Orge, France; (V.B.)
- URP 7330 VIFASOM, Université Paris Cité, Hôtel Dieu, 75004 Paris, France
| |
Collapse
|
2
|
Schädlich IS, Winzer R, Stabernack J, Tolosa E, Magnus T, Rissiek B. The role of the ATP-adenosine axis in ischemic stroke. Semin Immunopathol 2023:10.1007/s00281-023-00987-3. [PMID: 36917241 DOI: 10.1007/s00281-023-00987-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/25/2023] [Indexed: 03/16/2023]
Abstract
In ischemic stroke, the primary neuronal injury caused by the disruption of energy supply is further exacerbated by secondary sterile inflammation. The inflammatory cascade is largely initiated by the purine adenosine triphosphate (ATP) which is extensively released to the interstitial space during brain ischemia and functions as an extracellular danger signaling molecule. By engaging P2 receptors, extracellular ATP activates microglia leading to cytokine and chemokine production and subsequent immune cell recruitment from the periphery which further amplifies post-stroke inflammation. The ectonucleotidases CD39 and CD73 shape and balance the inflammatory environment by stepwise degrading extracellular ATP to adenosine which itself has neuroprotective and anti-inflammatory signaling properties. The neuroprotective effects of adenosine are mainly mediated through A1 receptors and inhibition of glutamatergic excitotoxicity, while the anti-inflammatory capacities of adenosine have been primarily attributed to A2A receptor activation on infiltrating immune cells in the subacute phase after stroke. In this review, we summarize the current state of knowledge on the ATP-adenosine axis in ischemic stroke, discuss contradictory results, and point out potential pitfalls towards translating therapeutic approaches from rodent stroke models to human patients.
Collapse
Affiliation(s)
- Ines Sophie Schädlich
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Riekje Winzer
- Institute of Immunology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Joschi Stabernack
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Eva Tolosa
- Institute of Immunology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| | - Björn Rissiek
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| |
Collapse
|
3
|
Quiquempoix M, Drogou C, Erblang M, Van Beers P, Guillard M, Tardo-Dino PE, Rabat A, Léger D, Chennaoui M, Gomez-Merino D, Sauvet F. Relationship between Habitual Caffeine Consumption, Attentional Performance, and Individual Alpha Frequency during Total Sleep Deprivation. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:4971. [PMID: 36981883 PMCID: PMC10049386 DOI: 10.3390/ijerph20064971] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 06/18/2023]
Abstract
(1) Background: Caffeine is a psychostimulant that is well known to mitigate the deleterious effects of sleep debt. Our aim was to assess the effects of acute caffeine intake on cognitive vulnerability and brain activity during total sleep deprivation (TSD), taking into account habitual caffeine consumption. (2) Methods: Thirty-seven subjects were evaluated in a double-blind, crossover, total sleep deprivation protocol with caffeine or placebo treatment. Vigilant attention was evaluated every six hours during TSD using the psychomotor vigilance test (PVT) with EEG recordings. The influence of habitual caffeine consumption was analyzed by categorizing subjects into low, moderate, and high consumers. (3) Results: The PVT reaction time (RT) increased during TSD and was lower in the caffeine condition vs. the placebo condition. The RT was shorter in the low-caffeine consumers compared to moderate and high consumers, regardless of conditions and treatments. The TSD-related increase in EEG power was attenuated by acute caffeine intake independently of habitual caffeine consumption, and the individual alpha frequency (IAF) was lower in the high-consumption group. The IAF was negatively correlated with daytime sleepiness. Moreover, a correlation analysis showed that the higher the daily caffeine consumption, the higher the RT and the lower the IAF. (4) Conclusions: A high level of habitual caffeine consumption decreases attentional performance and alpha frequencies, decreasing tolerance to sleep deprivation.
Collapse
Affiliation(s)
- Michael Quiquempoix
- Institut de Recherche Biomédicale des Armées (IRBA), 91223 Brétigny sur Orge, France (F.S.)
- URP 7330 VIFASOM, Université Paris Cité, Hôtel-Dieu, 75004 Paris, France
| | - Catherine Drogou
- Institut de Recherche Biomédicale des Armées (IRBA), 91223 Brétigny sur Orge, France (F.S.)
- URP 7330 VIFASOM, Université Paris Cité, Hôtel-Dieu, 75004 Paris, France
| | - Mégane Erblang
- Institut de Recherche Biomédicale des Armées (IRBA), 91223 Brétigny sur Orge, France (F.S.)
- URP 7330 VIFASOM, Université Paris Cité, Hôtel-Dieu, 75004 Paris, France
- Laboratoire de Biologie de l’Exercice pour la Performance et la Santé (UMR LBEPS), Université d’Evry, 91025 Evry-Courcouronnes, France
| | - Pascal Van Beers
- Institut de Recherche Biomédicale des Armées (IRBA), 91223 Brétigny sur Orge, France (F.S.)
- URP 7330 VIFASOM, Université Paris Cité, Hôtel-Dieu, 75004 Paris, France
| | - Mathias Guillard
- Institut de Recherche Biomédicale des Armées (IRBA), 91223 Brétigny sur Orge, France (F.S.)
- URP 7330 VIFASOM, Université Paris Cité, Hôtel-Dieu, 75004 Paris, France
| | - Pierre-Emmanuel Tardo-Dino
- Institut de Recherche Biomédicale des Armées (IRBA), 91223 Brétigny sur Orge, France (F.S.)
- URP 7330 VIFASOM, Université Paris Cité, Hôtel-Dieu, 75004 Paris, France
- Laboratoire de Biologie de l’Exercice pour la Performance et la Santé (UMR LBEPS), Université d’Evry, 91025 Evry-Courcouronnes, France
| | - Arnaud Rabat
- Institut de Recherche Biomédicale des Armées (IRBA), 91223 Brétigny sur Orge, France (F.S.)
- URP 7330 VIFASOM, Université Paris Cité, Hôtel-Dieu, 75004 Paris, France
| | - Damien Léger
- URP 7330 VIFASOM, Université Paris Cité, Hôtel-Dieu, 75004 Paris, France
- APHP, Hôtel-Dieu, Centre du Sommeil et de la Vigilance, 75004 Paris, France
| | - Mounir Chennaoui
- Institut de Recherche Biomédicale des Armées (IRBA), 91223 Brétigny sur Orge, France (F.S.)
- URP 7330 VIFASOM, Université Paris Cité, Hôtel-Dieu, 75004 Paris, France
| | - Danielle Gomez-Merino
- Institut de Recherche Biomédicale des Armées (IRBA), 91223 Brétigny sur Orge, France (F.S.)
- URP 7330 VIFASOM, Université Paris Cité, Hôtel-Dieu, 75004 Paris, France
| | - Fabien Sauvet
- Institut de Recherche Biomédicale des Armées (IRBA), 91223 Brétigny sur Orge, France (F.S.)
- URP 7330 VIFASOM, Université Paris Cité, Hôtel-Dieu, 75004 Paris, France
| |
Collapse
|
4
|
Segu A, Kannan NN. The duration of caffeine treatment plays an essential role in its effect on sleep and circadian rhythm. SLEEP ADVANCES : A JOURNAL OF THE SLEEP RESEARCH SOCIETY 2023; 4:zpad014. [PMID: 37193284 PMCID: PMC10108652 DOI: 10.1093/sleepadvances/zpad014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/28/2023] [Indexed: 05/18/2023]
Abstract
Sleep is regulated by the homeostatic system and the circadian clock. Caffeine intake promotes wakefulness in Drosophila. In humans, caffeine is consumed on a daily basis and hence it is important to understand the effect of prolonged caffeine intake on both circadian and homeostatic regulation of sleep. Furthermore, sleep changes with age and the impact of caffeine on age-dependent sleep fragmentation are yet to be understood. Hence in the present study, we examined the effect of short exposure to caffeine on homeostatic sleep and age-dependent sleep fragmentation in Drosophila. We further assessed the effect of prolonged exposure to caffeine on homeostatic sleep and circadian clock. The results of our study showed that short exposure to caffeine reduces sleep and food intake in mature flies. It also enhances sleep fragmentation with increasing age. However, we have not assessed the effect of caffeine on food intake in older flies. On the other hand, prolonged caffeine exposure did not exert any significant effect on the duration of sleep and food intake in mature flies. Nevertheless, prolonged caffeine ingestion decreased the morning and evening anticipatory activity in these flies indicating that it affects the circadian rhythm. These flies also exhibited phase delay in the clock gene timeless transcript oscillation and exhibited either behavioral arrhythmicity or a longer free-running period under constant darkness. In summary, the results of our studies showed that short exposure to caffeine increases the sleep fragmentation with age whereas prolonged caffeine exposure disrupts the circadian clock.
Collapse
Affiliation(s)
- Aishwarya Segu
- Chronobiology Laboratory, School of Biology, Indian Institute of Science Education and Research (IISER), Thiruvananthapuram, India
| | - Nisha N Kannan
- Chronobiology Laboratory, School of Biology, Indian Institute of Science Education and Research (IISER), Thiruvananthapuram, India
| |
Collapse
|
5
|
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
|
6
|
Rei N, Valente CA, Vaz SH, Farinha-Ferreira M, Ribeiro JA, Sebastião AM. Changes in adenosine receptors and neurotrophic factors in the SOD1G93A mouse model of amyotrophic lateral sclerosis: Modulation by chronic caffeine. PLoS One 2022; 17:e0272104. [PMID: 36516126 PMCID: PMC9749988 DOI: 10.1371/journal.pone.0272104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 11/13/2022] [Indexed: 12/15/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by the progressive degeneration of corticospinal tract motor neurons. Previous studies showed that adenosine-mediated neuromodulation is disturbed in ALS and that vascular endothelial growth factor (VEGF) has a neuroprotective function in ALS mouse models. We evaluated how adenosine (A1R and A2AR) and VEGF (VEGFA, VEGFB, VEGFR-1 and VEGFR-2) system markers are altered in the cortex and spinal cord of pre-symptomatic and symptomatic SOD1G93A mice. We then assessed if/how chronic treatment of SOD1G93A mice with a widely consumed adenosine receptor antagonist, caffeine, modulates VEGF system and/or the levels of Brain-derived Neurotrophic Factor (BDNF), known to be under control of A2AR. We found out decreases in A1R and increases in A2AR levels even before disease onset. Concerning the VEGF system, we detected increases of VEGFB and VEGFR-2 levels in the spinal cord at pre-symptomatic stage, which reverses at the symptomatic stage, and decreases of VEGFA levels in the cortex, in very late disease states. Chronic treatment with caffeine rescued cortical A1R levels in SOD1G93A mice, bringing them to control levels, while rendering VEGF signaling nearly unaffected. In contrast, BDNF levels were significantly affected in SOD1G93A mice treated with caffeine, being decreased in the cortex and increased in spinal the cord. Altogether, these findings suggest an early dysfunction of the adenosinergic system in ALS and highlights the possibility that the negative influence of caffeine previously reported in ALS animal models results from interference with BDNF rather than with the VEGF signaling molecules.
Collapse
Affiliation(s)
- Nádia Rei
- Faculdade de Medicina, Instituto de Farmacologia e Neurociências, Universidade de Lisboa, Lisboa, Portugal
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal
| | - Cláudia A. Valente
- Faculdade de Medicina, Instituto de Farmacologia e Neurociências, Universidade de Lisboa, Lisboa, Portugal
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal
| | - Sandra H. Vaz
- Faculdade de Medicina, Instituto de Farmacologia e Neurociências, Universidade de Lisboa, Lisboa, Portugal
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal
| | - Miguel Farinha-Ferreira
- Faculdade de Medicina, Instituto de Farmacologia e Neurociências, Universidade de Lisboa, Lisboa, Portugal
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal
| | - Joaquim A. Ribeiro
- Faculdade de Medicina, Instituto de Farmacologia e Neurociências, Universidade de Lisboa, Lisboa, Portugal
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal
| | - Ana M. Sebastião
- Faculdade de Medicina, Instituto de Farmacologia e Neurociências, Universidade de Lisboa, Lisboa, Portugal
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal
- * E-mail:
| |
Collapse
|
7
|
Paiva I, Cellai L, Meriaux C, Poncelet L, Nebie O, Saliou JM, Lacoste AS, Papegaey A, Drobecq H, Le Gras S, Schneider M, Malik EM, Müller CE, Faivre E, Carvalho K, Gomez-Murcia V, Vieau D, Thiroux B, Eddarkaoui S, Lebouvier T, Schueller E, Tzeplaeff L, Grgurina I, Seguin J, Stauber J, Lopes LV, Buee L, Buée-Scherrer V, Cunha RA, Ait-Belkacem R, Sergeant N, Annicotte JS, Boutillier AL, Blum D. Caffeine intake exerts dual genome-wide effects on hippocampal metabolism and learning-dependent transcription. J Clin Invest 2022; 132:149371. [PMID: 35536645 PMCID: PMC9197525 DOI: 10.1172/jci149371] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/05/2022] [Indexed: 12/01/2022] Open
Abstract
Caffeine is the most widely consumed psychoactive substance in the world. Strikingly, the molecular pathways engaged by its regular consumption remain unclear. We herein addressed the mechanisms associated with habitual (chronic) caffeine consumption in the mouse hippocampus using untargeted orthogonal omics techniques. Our results revealed that chronic caffeine exerts concerted pleiotropic effects in the hippocampus at the epigenomic, proteomic, and metabolomic levels. Caffeine lowered metabolism-related processes (e.g., at the level of metabolomics and gene expression) in bulk tissue, while it induced neuron-specific epigenetic changes at synaptic transmission/plasticity-related genes and increased experience-driven transcriptional activity. Altogether, these findings suggest that regular caffeine intake improves the signal-to-noise ratio during information encoding, in part through fine-tuning of metabolic genes, while boosting the salience of information processing during learning in neuronal circuits.
Collapse
Affiliation(s)
- Isabel Paiva
- Laboratoire de Neuroscience Cognitives et Adaptatives, University of Strasbourg, CNRS, UMR7364, Strasbourg, France
| | | | - Céline Meriaux
- Alzheimer and Tauopathies, Inserm UMR-S1172, Lille, France
| | | | - Ouada Nebie
- Alzheimer and Tauopathies, Inserm UMR-S1172, Lille, France
| | | | | | | | - Hervé Drobecq
- CIIL - Centre d'Infection et d'Immunité de Lille (CIIL), Inserm 1019, Lille, France
| | - Stéphanie Le Gras
- GenomEast Platform, University Strasbourg, CNRS UMR 7104, Inserm U1258, Lille, France
| | - Marion Schneider
- PharmaCenter Bonn, Pharmaceutical Institute, University of Bonn, Bonn, Germany
| | - Enas M Malik
- PharmaCenter Bonn, Pharmaceutical Institute, University of Bonn, Bonn, Germany
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, University of Bonn, Bonn, Germany
| | - Emilie Faivre
- Alzheimer and Tauopathies, Inserm UMR-S1172, Lille, France
| | - Kevin Carvalho
- Alzheimer and Tauopathies, Inserm UMR-S1172, Lille, France
| | | | - Didier Vieau
- Alzheimer and Tauopathies, Inserm UMR-S1172, Lille, France
| | - Bryan Thiroux
- Alzheimer and Tauopathies, Inserm UMR-S1172, Lille, France
| | | | | | - Estelle Schueller
- Laboratoire de Neuroscience Cognitives et Adaptatives, Université de Strasbourg, Strasbourg, France
| | - Laura Tzeplaeff
- Laboratoire de Neuroscience Cognitives et Adaptatives, University of Strasbourg, Strasbourg, France
| | - Iris Grgurina
- Laboratoire de Neuroscience Cognitives et Adaptatives, Université de Strasbourg, Strasbourg, France
| | - Jonathan Seguin
- Laboratoire de Neuroscience Cognitives et Adaptatives, Université de Strasbourg, Strasbourg, France
| | | | - Luisa V Lopes
- Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal
| | - Luc Buee
- Alzheimer and Tauopathies, Inserm UMR-S1172, Lille, France
| | | | - Rodrigo A Cunha
- Center for Neuroscience of Coimbra, University of Coimbra, Coimbra, Portugal
| | | | | | | | - Anne-Laurence Boutillier
- Laboratoire de Neuroscience Cognitives et Adaptatives, Université de Strasbourg, Strasbourg, France
| | - David Blum
- INSERM U837, University Lille-Nord de France, UDSL, Lille, France
| |
Collapse
|
8
|
IJzerman AP, Jacobson KA, Müller CE, Cronstein BN, Cunha RA. International Union of Basic and Clinical Pharmacology. CXII: Adenosine Receptors: A Further Update. Pharmacol Rev 2022; 74:340-372. [PMID: 35302044 PMCID: PMC8973513 DOI: 10.1124/pharmrev.121.000445] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Our previous International Union of Basic and Clinical Pharmacology report on the nomenclature and classification of adenosine receptors (2011) contained a number of emerging developments with respect to this G protein-coupled receptor subfamily, including protein structure, protein oligomerization, protein diversity, and allosteric modulation by small molecules. Since then, a wealth of new data and results has been added, allowing us to explore novel concepts such as target binding kinetics and biased signaling of adenosine receptors, to examine a multitude of receptor structures and novel ligands, to gauge new pharmacology, and to evaluate clinical trials with adenosine receptor ligands. This review should therefore be considered a further update of our previous reports from 2001 and 2011. SIGNIFICANCE STATEMENT: Adenosine receptors (ARs) are of continuing interest for future treatment of chronic and acute disease conditions, including inflammatory diseases, neurodegenerative afflictions, and cancer. The design of AR agonists ("biased" or not) and antagonists is largely structure based now, thanks to the tremendous progress in AR structural biology. The A2A- and A2BAR appear to modulate the immune response in tumor biology. Many clinical trials for this indication are ongoing, whereas an A2AAR antagonist (istradefylline) has been approved as an anti-Parkinson agent.
Collapse
Affiliation(s)
- Adriaan P IJzerman
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (A.P.IJ.); National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Molecular Recognition Section, Bethesda, Maryland (K.A.J.); Universität Bonn, Bonn, Germany (C.E.M.); New York University School of Medicine, New York, New York (B.N.C.); and Center for Neurosciences and Cell Biology and Faculty of Medicine, University of Coimbra, Coimbra, Portugal (R.A.C.)
| | - Kenneth A Jacobson
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (A.P.IJ.); National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Molecular Recognition Section, Bethesda, Maryland (K.A.J.); Universität Bonn, Bonn, Germany (C.E.M.); New York University School of Medicine, New York, New York (B.N.C.); and Center for Neurosciences and Cell Biology and Faculty of Medicine, University of Coimbra, Coimbra, Portugal (R.A.C.)
| | - Christa E Müller
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (A.P.IJ.); National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Molecular Recognition Section, Bethesda, Maryland (K.A.J.); Universität Bonn, Bonn, Germany (C.E.M.); New York University School of Medicine, New York, New York (B.N.C.); and Center for Neurosciences and Cell Biology and Faculty of Medicine, University of Coimbra, Coimbra, Portugal (R.A.C.)
| | - Bruce N Cronstein
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (A.P.IJ.); National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Molecular Recognition Section, Bethesda, Maryland (K.A.J.); Universität Bonn, Bonn, Germany (C.E.M.); New York University School of Medicine, New York, New York (B.N.C.); and Center for Neurosciences and Cell Biology and Faculty of Medicine, University of Coimbra, Coimbra, Portugal (R.A.C.)
| | - Rodrigo A Cunha
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (A.P.IJ.); National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Molecular Recognition Section, Bethesda, Maryland (K.A.J.); Universität Bonn, Bonn, Germany (C.E.M.); New York University School of Medicine, New York, New York (B.N.C.); and Center for Neurosciences and Cell Biology and Faculty of Medicine, University of Coimbra, Coimbra, Portugal (R.A.C.)
| |
Collapse
|
9
|
Halpin-Veszeleiova K, Hatfield SM. Therapeutic Targeting of Hypoxia-A2-Adenosinergic Pathway in COVID-19 Patients. Physiology (Bethesda) 2022; 37:46-52. [PMID: 34486395 PMCID: PMC8742736 DOI: 10.1152/physiol.00010.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The hypoxia-hypoxia-inducible factor (HIF)-1α-A2-adenosinergic pathway protects tissues from inflammatory damage during antipathogen immune responses. The elimination of this physiological tissue-protecting mechanism by supplemental oxygenation may contribute to the high mortality of oxygen-ventilated COVID-19 patients by exacerbating inflammatory lung damage. Restoration of this pathway with hypoxia-adenosinergic drugs may improve outcomes in these patients.
Collapse
Affiliation(s)
- Katarina Halpin-Veszeleiova
- New England Inflammation and Tissue Protection Institute, Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts
| | - Stephen M Hatfield
- New England Inflammation and Tissue Protection Institute, Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts
| |
Collapse
|
10
|
Bourgeois-Vionnet J, Ryvlin P, Elsensohn MH, Michel V, Valton L, Derambure P, Frazzini V, Hirsch E, Maillard L, Bartolomei F, Biberon J, Petit J, Biraben A, Crespel A, Thomas P, Lemesle-Martin M, Convers P, Leclercq M, Boulogne S, Roy P, Rheims S. Coffee consumption and seizure frequency in patients with drug-resistant focal epilepsy. Epilepsy Behav 2022; 126:108486. [PMID: 34929474 DOI: 10.1016/j.yebeh.2021.108486] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/28/2021] [Accepted: 11/29/2021] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To assess the relation between coffee consumption and seizure frequency in patients with drug-resistant focal epilepsy. METHODS Cross-sectional analysis of data collected in the SAVE study, which included patients with drug-resistant focal epilepsy during long-term EEG monitoring. Patients in whom both coffee consumption and data about seizure frequency, including focal to bilateral tonic-clonic seizures (FBTCS), were available were selected. Coffee consumption was collected using a standardized self-report questionnaire and classified into four groups: none, rare (from less than 1 cup/week to up 3 cups/week), moderate (from 4 cups/week to 3 cups/day), and high (more than 4 cups/day). RESULTS Six hundred and nineteen patients were included. There was no relation between coffee consumption and total seizure frequency (p = 0.902). In contrast, the number of FBTCS reported over the past year was significantly associated with usual coffee consumption (p = 0.029). Specifically, number of FBCTS in patients who reported moderate coffee consumption was lower than in others. In comparison with patients with moderate coffee consumption, the odds ratio (95%CI) for reporting at least 1 FBTCS per year was 1.6 (1.03-2.49) in patients who never take coffee, 1.62 (1.02-2.57) in those with rare consumption and 2.05 (1.24-3.4) in those with high consumption. Multiple ordinal logistic regression showed a trend toward an association between coffee consumption and number of FBTCS (p = 0.08). CONCLUSIONS AND RELEVANCE Our data suggest that effect of coffee consumption on seizures might depend on dose with potential benefits on FBTCS frequency at moderate doses. These results will have to be confirmed by prospective studies.
Collapse
Affiliation(s)
- Julie Bourgeois-Vionnet
- Department of Functional Neurology and Epileptology, Hospices Civils de Lyon and University of Lyon, Lyon, France
| | - Philippe Ryvlin
- Department of Clinical Neurosciences, Centre Hospitalo-Universitaire Vaudois, Lausanne, Switzerland
| | - Mad-Hélénie Elsensohn
- Lyon University, Lyon, France; Équipe Biostatistique Santé, Laboratoire de Biométrie et Biologie Évolutive, CNRS UMR 5558, Villeurbanne, France; Service de Biostatistique-Bioinformatique, Pôle Santé Publique, Hospices Civils de Lyon, Lyon, France
| | | | - Luc Valton
- Clinical Neurophysiology, Department of Neurology, University Hospital of Toulouse, CerCO CNRS UMR 5549, University of Toulouse, France
| | - Philippe Derambure
- Department of Clinical Neurophysiology, Lille University Medical Center, EA 1046, University of Lille 2, France
| | - Valerio Frazzini
- Epileptology Unit, Assistance Publique-Hôpitaux de Paris, Groupe hospitalier Pitié-Salpêtrière and Brain and Spine Institute (ICM; INSERM UMRS1127, CNRS UMR7225, UPMC University Paris 06), Paris, France
| | - Edouard Hirsch
- Department of Neurology, University Hospital of Strasbourg, Strasbourg, France
| | - Louis Maillard
- Neurology Department, University Hospital of Nancy, Nancy, France
| | - Fabrice Bartolomei
- Clinical Neurophysiology and Epileptology Department, Timone Hospital, Marseille, France
| | - Julien Biberon
- Department of Clinical Neurophysiology, INSERM U930, University Hospital of Tours, Tours, France
| | - Jerôme Petit
- La Teppe Epilepsy Center, Tain l'Hermitage, France
| | - Arnaud Biraben
- Department of Neurology, University Hospital of Rennes, Rennes, France
| | | | - Pierre Thomas
- Neurology Department University Hospitals of Nice, Nice, France
| | | | - Philippe Convers
- Department of Neurology, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Mathilde Leclercq
- Department of Functional Neurology and Epileptology, Hospices Civils de Lyon and University of Lyon, Lyon, France
| | - Sébastien Boulogne
- Department of Functional Neurology and Epileptology, Hospices Civils de Lyon and University of Lyon, Lyon, France; Lyon's Neuroscience Research Center, INSERM U1028 / CNRS UMR 5292 and Lyon 1 University, Lyon, France
| | - Pascal Roy
- Lyon University, Lyon, France; Équipe Biostatistique Santé, Laboratoire de Biométrie et Biologie Évolutive, CNRS UMR 5558, Villeurbanne, France; Service de Biostatistique-Bioinformatique, Pôle Santé Publique, Hospices Civils de Lyon, Lyon, France
| | - Sylvain Rheims
- Department of Functional Neurology and Epileptology, Hospices Civils de Lyon and University of Lyon, Lyon, France; Lyon's Neuroscience Research Center, INSERM U1028 / CNRS UMR 5292 and Lyon 1 University, Lyon, France; Epilepsy Institute, Lyon, France.
| | | |
Collapse
|
11
|
Rossetto IMU, Cagnon VHA, Kido LA, Lizarte Neto FS, Tirapelli LF, Tirapelli DPDC, de Almeida Chuffa LG, Martinez FE, Martinez M. Caffeine consumption attenuates ethanol-induced inflammation through the regulation of adenosinergic receptors in the UChB rats cerebellum. Toxicol Res (Camb) 2021; 10:835-849. [PMID: 34484675 DOI: 10.1093/toxres/tfab067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/27/2021] [Accepted: 06/25/2021] [Indexed: 12/20/2022] Open
Abstract
Caffeine consumption is able to interfere in cellular processes related to inflammatory mechanisms by acting through the adenosinergic system. This study aimed to recognize alterations related to adenosinergic system and inflammatory process in the cerebellum of University of Chile Bibulous (UChB) rats after the consumption of ethanol and caffeine. UChB and Wistar rats, males at 5 months old, were divided into the groups (n = 15/group): (i) Control (Wistar rats receiving water); (ii) Ethanol group (UChB rats receiving ethanol solution at 10%) and (iii) Ethanol+caffeine group (UChB rats receiving ethanol solution at 10% added of 3 g/L of caffeine). The cerebellar tissue was collected and processed for immunohistochemistry, Reverse transcription polymerase chain reaction (RT-PCR) and western blotting techniques for the adenosinergic receptors A1 and A2a and inflammatory markers, including Nuclear factor kappa B (NFkB), TLR4, TLR2, MyD88, TNF-α, COX-2, iNOS and microglial marker Iba-1. Results showed ethanol and caffeine consumption differentially altering the immunolocalization of adenosinergic receptors and inflammatory markers in the cerebellar tissue. The A2a receptor was overexpressed in the Ethanol group and was evident in the glial cells. The Ethanol group had increased protein levels for NFκB and TLR4, expressively in Bergmann glia and Purkinje cells. Caffeine reduced the expression of these markers to levels similar to those found in the Control group. The A1 gene was upregulated the Ethanol group, but not its protein levels, suggesting post-transcriptional interference. In conclusion, caffeine seems to attenuate ethanol-induced inflammation in the cerebellum of UChB rats through the A1 and A2a modulation, playing a neuroprotective role in the chronic context of ethanol consumption.
Collapse
Affiliation(s)
- Isabela Maria Urra Rossetto
- Department of Structural and Functional Biology, University of Campinas (UNICAMP), 255 Monteiro Lobato St, Campinas, SP 13083-862, Brazil
| | - Valéria Helena Alves Cagnon
- Department of Structural and Functional Biology, University of Campinas (UNICAMP), 255 Monteiro Lobato St, Campinas, SP 13083-862, Brazil
| | - Larissa Akemi Kido
- Department of Food and Nutrition, University of Campinas (UNICAMP), 80 Monteiro Lobato St, Campinas, SP 13083-862, Brazil
| | - Fermino Sanches Lizarte Neto
- Department of Surgery and Anatomy, University of São Paulo (USP), 3900 Bandeirantes Ave, Ribeirão Preto, SP 14049-900, Brazil
| | - Luís Fernando Tirapelli
- Department of Surgery and Anatomy, University of São Paulo (USP), 3900 Bandeirantes Ave, Ribeirão Preto, SP 14049-900, Brazil
| | | | - Luiz Gustavo de Almeida Chuffa
- Department of Structural and Functional Biology, State University of São Paulo (UNESP), 250 Prof. Dr. Antônio Celso Wagner Zanin St, Botucatu, SP 18618-689, Brazil
| | - Francisco Eduardo Martinez
- Department of Structural and Functional Biology, State University of São Paulo (UNESP), 250 Prof. Dr. Antônio Celso Wagner Zanin St, Botucatu, SP 18618-689, Brazil
| | - Marcelo Martinez
- Department of Morphology and Pathology, Federal University of São Carlos (UFSCar), 13571 Biblioteca Comunitária Ave, São Carlos, SP 13565-905, Brazil
| |
Collapse
|
12
|
Caffeine - treat or trigger? Disparate behavioral and long-term dopaminergic changes in control and schizophrenia-like Wisket rats. Physiol Behav 2021; 236:113410. [PMID: 33819453 DOI: 10.1016/j.physbeh.2021.113410] [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: 01/15/2021] [Revised: 03/12/2021] [Accepted: 03/26/2021] [Indexed: 11/20/2022]
Abstract
The influence of caffeine on behavioral functions in both healthy and schizophrenic subjects is controversial. Here we aimed to reveal the effects of repeated caffeine pre- and post-training treatments on motor and exploratory activities and cognitive functions in a reward-based test (Ambitus) along with a brain region-specific dopamine D2 receptor profile in control and schizophrenia-like WISKET model rats. In the control animals, pre-treatment caused temporary enhancement in motor activity, while permanent improvement in learning function was detected in the WISKET animals. Post-treatment produced significant impairments in both groups. Caffeine caused short-lasting hyperactivity followed by a rebound in the inactive phase determined in undisturbed circumstance. Caffeine treatment substantially enhanced the dopamine D2 receptor mediated G-protein activation in the prefrontal cortex and olfactory bulb of both groups, while it increased in the dorsal striatum and cerebral cortex only in the WISKET animals. Caffeine enhanced the maximal binding capacity in the hippocampus and cerebral cortex of WISKET animals, but it decreased in the prefrontal cortex of the control animals. Regarding the dopamine D2 receptor mRNA expression, caffeine treatment caused significant enhancement in the prefrontal cortex of WISKET animals, while it increased the hippocampal dopamine D2 receptor protein amount in both groups. This study highlights the disparate effects of caffeine pre- versus post-training treatments on behavioral parameters in both control and schizophrenia-like animals and the prolonged changes in the dopaminergic system. It is supposed that the delayed depressive effects of caffeine might be compensated by frequent coffee intake, as observed in schizophrenic patients.
Collapse
|
13
|
Beamer E, Kuchukulla M, Boison D, Engel T. ATP and adenosine-Two players in the control of seizures and epilepsy development. Prog Neurobiol 2021; 204:102105. [PMID: 34144123 DOI: 10.1016/j.pneurobio.2021.102105] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/07/2021] [Accepted: 06/09/2021] [Indexed: 02/08/2023]
Abstract
Despite continuous advances in understanding the underlying pathogenesis of hyperexcitable networks and lowered seizure thresholds, the treatment of epilepsy remains a clinical challenge. Over one third of patients remain resistant to current pharmacological interventions. Moreover, even when effective in suppressing seizures, current medications are merely symptomatic without significantly altering the course of the disease. Much effort is therefore invested in identifying new treatments with novel mechanisms of action, effective in drug-refractory epilepsy patients, and with the potential to modify disease progression. Compelling evidence has demonstrated that the purines, ATP and adenosine, are key mediators of the epileptogenic process. Extracellular ATP concentrations increase dramatically under pathological conditions, where it functions as a ligand at a host of purinergic receptors. ATP, however, also forms a substrate pool for the production of adenosine, via the action of an array of extracellular ATP degrading enzymes. ATP and adenosine have assumed largely opposite roles in coupling neuronal excitability to energy homeostasis in the brain. This review integrates and critically discusses novel findings regarding how ATP and adenosine control seizures and the development of epilepsy. This includes purine receptor P1 and P2-dependent mechanisms, release and reuptake mechanisms, extracellular and intracellular purine metabolism, and emerging receptor-independent effects of purines. Finally, possible purine-based therapeutic strategies for seizure suppression and disease modification are discussed.
Collapse
Affiliation(s)
- Edward Beamer
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin D02 YN77, Ireland; Centre for Bioscience, Manchester Metropolitan University, John Dalton Building, All Saints Campus, Manchester M15 6BH, UK
| | - Manvitha Kuchukulla
- Department of Neurosurgery, Robert Wood Johnson & New Jersey Medical Schools, Rutgers University, Piscataway, NJ 08854, USA
| | - Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson & New Jersey Medical Schools, Rutgers University, Piscataway, NJ 08854, USA.
| | - Tobias Engel
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin D02 YN77, Ireland; FutureNeuro, Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin D02 YN77, Ireland.
| |
Collapse
|
14
|
Pereira-Figueiredo D, Nascimento AA, Cunha-Rodrigues MC, Brito R, Calaza KC. Caffeine and Its Neuroprotective Role in Ischemic Events: A Mechanism Dependent on Adenosine Receptors. Cell Mol Neurobiol 2021; 42:1693-1725. [PMID: 33730305 DOI: 10.1007/s10571-021-01077-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/05/2021] [Indexed: 02/07/2023]
Abstract
Ischemia is characterized by a transient, insufficient, or permanent interruption of blood flow to a tissue, which leads to an inadequate glucose and oxygen supply. The nervous tissue is highly active, and it closely depends on glucose and oxygen to satisfy its metabolic demand. Therefore, ischemic conditions promote cell death and lead to a secondary wave of cell damage that progressively spreads to the neighborhood areas, called penumbra. Brain ischemia is one of the main causes of deaths and summed with retinal ischemia comprises one of the principal reasons of disability. Although several studies have been performed to investigate the mechanisms of damage to find protective/preventive interventions, an effective treatment does not exist yet. Adenosine is a well-described neuromodulator in the central nervous system (CNS), and acts through four subtypes of G-protein-coupled receptors. Adenosine receptors, especially A1 and A2A receptors, are the main targets of caffeine in daily consumption doses. Accordingly, caffeine has been greatly studied in the context of CNS pathologies. In fact, adenosine system, as well as caffeine, is involved in neuroprotection effects in different pathological situations. Therefore, the present review focuses on the role of adenosine/caffeine in CNS, brain and retina, ischemic events.
Collapse
Affiliation(s)
- D Pereira-Figueiredo
- Neurobiology of the Retina Laboratory, Biomedical Sciences Program, Biomedical Institute, Fluminense Federal University, Niterói, RJ, Brazil
| | - A A Nascimento
- Neurobiology of the Retina Laboratory, Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil
| | - M C Cunha-Rodrigues
- Neurobiology of the Retina Laboratory, Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil
| | - R Brito
- Laboratory of Neuronal Physiology and Pathology, Cellular and Molecular Biology Department, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil
| | - K C Calaza
- Neurobiology of the Retina Laboratory, Biomedical Sciences Program, Biomedical Institute, Fluminense Federal University, Niterói, RJ, Brazil. .,Neurobiology of the Retina Laboratory, Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil. .,Neurobiology Department, Biology Institute of Fluminense Federal University, Niteroi, RJ, Brazil.
| |
Collapse
|
15
|
Spinozzi E, Baldassarri C, Acquaticci L, Del Bello F, Grifantini M, Cappellacci L, Riccardo P. Adenosine receptors as promising targets for the management of ocular diseases. Med Chem Res 2021; 30:353-370. [PMID: 33519168 PMCID: PMC7829661 DOI: 10.1007/s00044-021-02704-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022]
Abstract
The ocular drug discovery arena has undergone a significant improvement in the last few years culminating in the FDA approvals of 8 new drugs. However, despite a large number of drugs, generics, and combination products available, it remains an urgent need to find breakthrough strategies and therapies for tackling ocular diseases. Targeting the adenosinergic system may represent an innovative strategy for discovering new ocular therapeutics. This review focused on the recent advance in the field and described the numerous nucleoside and non-nucleoside modulators of the four adenosine receptors (ARs) used as potential tools or clinical drug candidates.
Collapse
Affiliation(s)
- Eleonora Spinozzi
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Cecilia Baldassarri
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Laura Acquaticci
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Fabio Del Bello
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Mario Grifantini
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Loredana Cappellacci
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Petrelli Riccardo
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| |
Collapse
|
16
|
Caruana DA, Dudek SM. Adenosine A 1 Receptor-Mediated Synaptic Depression in the Developing Hippocampal Area CA2. Front Synaptic Neurosci 2020; 12:21. [PMID: 32612520 PMCID: PMC7307308 DOI: 10.3389/fnsyn.2020.00021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/04/2020] [Indexed: 12/22/2022] Open
Abstract
Immunolabeling for adenosine A1 receptors (A1Rs) is high in hippocampal area CA2 in adult rats, and the potentiating effects of caffeine or other A1R-selective antagonists on synaptic responses are particularly robust at Schaffer collateral synapses in CA2. Interestingly, the pronounced staining for A1Rs in CA2 is not apparent until rats are 4 weeks old, suggesting that developmental changes other than receptor distribution underlie the sensitivity of CA2 synapses to A1R antagonists in young animals. To evaluate the role of A1R-mediated postsynaptic signals at these synapses, we tested whether A1R agonists regulate synaptic transmission at Schaffer collateral inputs to CA2 and CA1. We found that the selective A1R agonist CCPA caused a lasting depression of synaptic responses in both CA2 and CA1 neurons in slices obtained from juvenile rats (P14), but that the effect was observed only in CA2 in slices prepared from adult animals (~P70). Interestingly, blocking phosphodiesterase activity with rolipram inhibited the CCPA-induced depression in CA1, but not in CA2, indicative of robust phosphodiesterase activity in CA1 neurons. Likewise, synaptic responses in CA2 and CA1 differed in their sensitivity to the adenylyl cyclase activator, forskolin, in that it increased synaptic transmission in CA2, but had little effect in CA1. These findings suggest that the A1R-mediated synaptic depression tracks the postnatal development of immunolabeling for A1Rs and that the enhanced sensitivity to antagonists in CA2 at young ages is likely due to robust adenylyl cyclase activity and weak phosphodiesterase activity rather than to enrichment of A1Rs.
Collapse
Affiliation(s)
- Douglas A. Caruana
- School of Life and Health Sciences, Aston University, Birmingham, United Kingdom
- Neurobiology Laboratory, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health, Research Triangle Park, NC, United States
| | - Serena M. Dudek
- Neurobiology Laboratory, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health, Research Triangle Park, NC, United States
| |
Collapse
|
17
|
Lusardi TA, Lytle NK, Gebril HM, Boison D. Effects of Preinjury and Postinjury Exposure to Caffeine in a Rat Model of Traumatic Brain Injury. J Caffeine Adenosine Res 2020; 10:12-24. [PMID: 32181443 PMCID: PMC7071069 DOI: 10.1089/caff.2019.0012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: Lethal apnea is a significant cause of acute mortality following a severe traumatic brain injury (TBI). TBI is associated with a surge of adenosine, which also suppresses respiratory function in the brainstem. Methods and Materials: This study examined the acute and chronic effects of caffeine, an adenosine receptor antagonist, on acute mortality and morbidity after fluid percussion injury. Results: We demonstrate that, regardless of preinjury caffeine exposure, an acute bolus of caffeine given immediately following the injury dosedependently prevented lethal apnea and has no detrimental effects on motor performance following sublethal injuries. Finally, we demonstrate that chronic caffeine treatment after injury, but not caffeine withdrawal, impairs recovery of motor function. Conclusions: Preexposure of the injured brain to caffeine does not have a major impact on acute and delayed outcome parameters; more importantly, a single acute dose of caffeine after the injury can prevent lethal apnea regardless of chronic caffeine preexposure.
Collapse
Affiliation(s)
- Theresa A. Lusardi
- School of Medicine Computational Biology Program, Oregon Health and Science University, Portland, Oregon
- Robert Stone Dow Neurobiology Laboratories, LRI, Portland, Oregon
| | - Nikki K. Lytle
- Robert Stone Dow Neurobiology Laboratories, LRI, Portland, Oregon
- Salk Institute for Biological Studies, La Jolla, California
| | - Hoda M. Gebril
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey
| | - Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey
| |
Collapse
|
18
|
Pereira-Figueiredo D, Brito R, Araújo DSM, Nascimento AA, Lyra ESB, Cheibub AMSS, Pereira Netto AD, Ventura ALM, Paes-de-Carvalho R, Calaza KC. Caffeine exposure ameliorates acute ischemic cell death in avian developing retina. Purinergic Signal 2020; 16:41-59. [PMID: 32078115 PMCID: PMC7166236 DOI: 10.1007/s11302-020-09687-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 01/15/2020] [Indexed: 12/20/2022] Open
Abstract
In infants, the main cause of blindness is retinopathy of prematurity that stems in a hypoxic-ischemic condition. Caffeine is a psychoactive compound that at low to moderate concentrations, selectively inhibits adenosine A1 and A2A receptors. Caffeine exerts beneficial effects in central nervous system of adult animal models and humans, whereas it seems to have malefic effect on the developing tissue. We observed that 48-h exposure (during synaptogenesis) to a moderate dose of caffeine (30 mg/kg of egg) activated pro-survival signaling pathways, including ERK, CREB, and Akt phosphorylation, alongside BDNF production, and reduced retinal cell death promoted by oxygen glucose deprivation in the chick retina. Blockade of TrkB receptors and inhibition of CREB prevented caffeine protection effect. Similar signaling pathways were described in previously reported data concerning chemical preconditioning mechanism triggered by NMDA receptors activation, with low concentrations of agonist. In agreement to these data, caffeine increased NMDA receptor activity. Caffeine decreased the levels of the chloride co-transporter KCC2 and delayed the developmental shift on GABAA receptor response from depolarizing to hyperpolarizing. These results suggest that the caffeine-induced delaying in depolarizing effect of GABA could be facilitating NMDA receptor activity. DPCPX, an A1 adenosine receptor antagonist, but not A2A receptor inhibitor, mimicked the effect of caffeine, suggesting that the effect of caffeine occurs through A1 receptor blockade. In summary, an in vivo caffeine exposure could increase the resistance of the retina to ischemia-induced cell death, by triggering survival pathways involving CREB phosphorylation and BDNF production/TrkB activation.
Collapse
Affiliation(s)
- D. Pereira-Figueiredo
- Neurobiology of the Retina Laboratory, Fluminense Federal University, Niterói, RJ Brazil
- Laboratory of Neurochemistry, Department of Neurobiology and Program of Neurosciences, Fluminense Federal University, Niterói, RJ Brazil
| | - R. Brito
- Cellular Signaling and Metabolic Modulation Laboratory, Cellular and Molecular Biology Department, Institute of Biology, Fluminense Federal University, Niterói, RJ Brazil
| | - D. S. M. Araújo
- Neurobiology of the Retina Laboratory, Fluminense Federal University, Niterói, RJ Brazil
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - A. A. Nascimento
- Neurobiology of the Retina Laboratory, Fluminense Federal University, Niterói, RJ Brazil
- Laboratory of Neurochemistry, Department of Neurobiology and Program of Neurosciences, Fluminense Federal University, Niterói, RJ Brazil
| | - E. S. B. Lyra
- Fundamental and Applied Analytical Chemistry Laboratory (LAQAFA), Department of Analytical Chemistry, Chemistry Institute, Fluminense Federal University, Niterói, RJ Brazil
| | - A. M. S. S. Cheibub
- Fundamental and Applied Analytical Chemistry Laboratory (LAQAFA), Department of Analytical Chemistry, Chemistry Institute, Fluminense Federal University, Niterói, RJ Brazil
| | - A. D. Pereira Netto
- Fundamental and Applied Analytical Chemistry Laboratory (LAQAFA), Department of Analytical Chemistry, Chemistry Institute, Fluminense Federal University, Niterói, RJ Brazil
| | - A. L. M. Ventura
- Laboratory of Neurochemistry, Department of Neurobiology and Program of Neurosciences, Fluminense Federal University, Niterói, RJ Brazil
- Neurobiology Department, Biology Institute of Fluminense Federal University, Niteroi, Rio de Janeiro Brazil
| | - R. Paes-de-Carvalho
- Laboratory of Cellular Neurobiology, Fluminense Federal University, Niterói, RJ Brazil
- Laboratory of Neurochemistry, Department of Neurobiology and Program of Neurosciences, Fluminense Federal University, Niterói, RJ Brazil
- Neurobiology Department, Biology Institute of Fluminense Federal University, Niteroi, Rio de Janeiro Brazil
| | - K. C. Calaza
- Neurobiology of the Retina Laboratory, Fluminense Federal University, Niterói, RJ Brazil
- Laboratory of Neurochemistry, Department of Neurobiology and Program of Neurosciences, Fluminense Federal University, Niterói, RJ Brazil
- Neurobiology Department, Biology Institute of Fluminense Federal University, Niteroi, Rio de Janeiro Brazil
| |
Collapse
|
19
|
Petković B, Kesić S, Pešić V. Critical View on the Usage of Ribavirin in Already Existing Psychostimulant-Use Disorder. Curr Pharm Des 2020; 26:466-484. [PMID: 31939725 PMCID: PMC8383468 DOI: 10.2174/1381612826666200115094642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 12/21/2019] [Indexed: 12/12/2022]
Abstract
Substance-use disorder represents a frequently hidden non-communicable chronic disease. Patients with intravenous drug addiction are at high risk of direct exposure to a variety of viral infections and are considered to be the largest subpopulation infected with the hepatitis C virus. Ribavirin is a synthetic nucleoside analog that has been used as an integral component of hepatitis C therapy. However, ribavirin medication is quite often associated with pronounced psychiatric adverse effects. It is not well understood to what extent ribavirin per se contributes to changes in drug-related neurobehavioral disturbances, especially in the case of psychostimulant drugs, such as amphetamine. It is now well-known that repeated amphetamine usage produces psychosis in humans and behavioral sensitization in animals. On the other hand, ribavirin has an affinity for adenosine A1 receptors that antagonistically modulate the activity of dopamine D1 receptors, which play a critical role in the development of behavioral sensitization. This review will focus on the current knowledge of neurochemical/ neurobiological changes that exist in the psychostimulant drug-addicted brain itself and the antipsychotic-like efficiency of adenosine agonists. Particular attention will be paid to the potential side effects of ribavirin therapy, and the opportunities and challenges related to its application in already existing psychostimulant-use disorder.
Collapse
Affiliation(s)
- Branka Petković
- Address correspondence to this author at the Department of Neurophysiology, Institute for Biological Research “Siniša Stanković” - National Institute of Republic of Serbia, University of Belgrade, Despota Stefana Blvd. 142, 11060, Belgrade, Serbia; Tel: +381-11-20-78-300; Fax: +381-11-27-61-433; E-mail:
| | | | | |
Collapse
|
20
|
Chern Y, Rei N, Ribeiro JA, Sebastião AM. Adenosine and Its Receptors as Potential Drug Targets in Amyotrophic Lateral Sclerosis. J Caffeine Adenosine Res 2019. [DOI: 10.1089/caff.2019.0016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Yijuang Chern
- Division of Neuroscience, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Nádia Rei
- Faculdade de Medicina, Instituto de Farmacologia e Neurociências, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal
| | - Joaquim A. Ribeiro
- Faculdade de Medicina, Instituto de Farmacologia e Neurociências, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal
| | - Ana M. Sebastião
- Faculdade de Medicina, Instituto de Farmacologia e Neurociências, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
21
|
Wang Y, Venton BJ. Caffeine Modulates Spontaneous Adenosine and Oxygen Changes during Ischemia and Reperfusion. ACS Chem Neurosci 2019; 10:1941-1949. [PMID: 30252436 PMCID: PMC7003050 DOI: 10.1021/acschemneuro.8b00251] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Adenosine is an endogenous neuroprotectant that modulates vasodilation in the central nervous system. Oxygen changes occur when there is an increase in local cerebral blood flow and thus are a measure of vasodilation. Transient oxygen events following rapid adenosine events have been recently discovered, but the relationship between adenosine and blood flow change during ischemia/reperfusion (I/R) has not been characterized. Caffeine is a nonselective adenosine receptor antagonist that can modulate the effects of adenosine in the brain, but how it affects adenosine and oxygen levels during I/R is also unknown. In this study, extracellular changes in adenosine and oxygen were simultaneously monitored using fast-scan cyclic voltammetry during bilateral common carotid artery occlusion (BCCAO) and the effects of a specific A2A antagonist, SCH 442416, or general antagonist, caffeine, were studied. Measurements were made in the caudate-putamen for 1 h of normoxia, followed by 30 min of BCCAO and 30 min of reperfusion. The frequency and number of both adenosine and oxygen transient events significantly increased during I/R. The specific A2A antagonist, SCH 442416 (3 mg/kg, i.p.), eliminated the increase in adenosine and oxygen events caused by I/R. The general adenosine receptor antagonist, caffeine (100 mg/kg, i.p.), decreased the frequency of adenosine and oxygen transient events during I/R. These results demonstrate that, during BCCAO, there are more rapid release events of the neuromodulator adenosine and correlated local oxygen changes, and these rapid, local effects are dampened by caffeine and other A2A antagonists.
Collapse
Affiliation(s)
- Ying Wang
- Department of Chemistry , University of Virginia , Charlottesville , Virginia 22904 , United States
| | - B Jill Venton
- Department of Chemistry , University of Virginia , Charlottesville , Virginia 22904 , United States
| |
Collapse
|
22
|
Kim Y, Kim H, Lee J, Lee JK, Min SJ, Seong J, Rhim H, Tae J, Lee HJ, Choo H. Discovery of β-Arrestin Biased Ligands of 5-HT 7R. J Med Chem 2018; 61:7218-7233. [PMID: 30028132 DOI: 10.1021/acs.jmedchem.8b00642] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Though many studies have been published about therapeutic potentials of selective 5-HT7R ligands, there have been few biased ligands of 5-HT7R. The development of potent and selective biased ligands of 5-HT7R would be of great help in understanding the relationship between pharmacological effects and G protein/β-arrestin signaling pathways of 5-HT7R. In order to identify 5-HT7R ligands with biased agonism, we designed and synthesized a series of tetrahydroazepine derivatives 1 and 2 with arylpyrazolo moiety or arylisoxazolo moiety. Through several biological evaluations such as binding affinity, selectivity profile, and functions in G protein and β-arrestin signaling pathways, 3-(4-chlorophenyl)-1,4,5,6,7,8-hexahydropyrazolo[3,4- d]azepine 1g was discovered as the β-arrestin biased ligand of 5-HT7R. In an electroencephalogram (EEG) test, 1g increased total non-rapid eye movement (NREM) sleep time and decreased total rapid eye movement (REM) sleep time.
Collapse
Affiliation(s)
- Youngjae Kim
- Center for Neuro-Medicine , Brain Research Institute, Korea Institute of Science and Technology , Seoul 02792 , Republic of Korea.,Department of Chemistry , Yonsei University , Seoul 03722 , Republic of Korea
| | - Hyunguk Kim
- School of Electrical Engineering , Korea Advanced Institute of Science and Technology , Daejeon 34141 , Republic of Korea
| | - Jieon Lee
- Center for Neuro-Medicine , Brain Research Institute, Korea Institute of Science and Technology , Seoul 02792 , Republic of Korea.,Division of Bio-Medical Science & Technology, KIST School , Korea University of Science and Technology , Seoul 02792 , Republic of Korea
| | - Jae Kyun Lee
- Center for Neuro-Medicine , Brain Research Institute, Korea Institute of Science and Technology , Seoul 02792 , Republic of Korea
| | - Sun-Joon Min
- Department of Chemical & Molecular Engineering/Applied Chemistry , Hanyang University , Ansan , Gyeonggi-do 15588 , Republic of Korea
| | - Jihye Seong
- Center for Neuro-Medicine , Brain Research Institute, Korea Institute of Science and Technology , Seoul 02792 , Republic of Korea.,Division of Bio-Medical Science & Technology, KIST School , Korea University of Science and Technology , Seoul 02792 , Republic of Korea.,Convergence Research Center for Diagnosis Treatment Care of Dementia , Korea Institute of Science and Technology , Seoul 02792 , Republic of Korea
| | - Hyewhon Rhim
- Division of Bio-Medical Science & Technology, KIST School , Korea University of Science and Technology , Seoul 02792 , Republic of Korea.,Center for Neuroscience , Brain Research Institute, Korea Institute of Science and Technology , Seoul 02792 , Republic of Korea
| | - Jinsung Tae
- Department of Chemistry , Yonsei University , Seoul 03722 , Republic of Korea
| | - Hyunjoo Jenny Lee
- School of Electrical Engineering , Korea Advanced Institute of Science and Technology , Daejeon 34141 , Republic of Korea
| | - Hyunah Choo
- Center for Neuro-Medicine , Brain Research Institute, Korea Institute of Science and Technology , Seoul 02792 , Republic of Korea.,Division of Bio-Medical Science & Technology, KIST School , Korea University of Science and Technology , Seoul 02792 , Republic of Korea
| |
Collapse
|
23
|
Laughlin BW, Bailey IR, Rice SA, Barati Z, Bogren LK, Drew KL. Precise Control of Target Temperature Using N 6-Cyclohexyladenosine and Real-Time Control of Surface Temperature. Ther Hypothermia Temp Manag 2018; 8:108-116. [PMID: 29480748 DOI: 10.1089/ther.2017.0020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Targeted temperature management is standard of care for cardiac arrest and is in clinical trials for stroke. N6-cyclohexyladenosine (CHA), an A1 adenosine receptor (A1AR) agonist, inhibits thermogenesis and induces onset of hibernation in hibernating species. Despite promising thermolytic efficacy of CHA, prior work has failed to achieve and maintain a prescribed target core body temperature (Tb) between 32°C and 34°C for 24 hours. We instrumented Sprague-Dawley rats (n = 19) with indwelling arterial and venous cannulae and a transmitter for monitoring Tb and ECG, then administered CHA via continuous IV infusion or intraperitoneal (IP) injection. In the first experiment (n = 11), we modulated ambient temperature and increased the dose of CHA in an attempt to manage Tb. In the second experiment (n = 8), we administered CHA (0.25 mg/[kg·h]) via continuous IV infusion and modulated cage surface temperature to control Tb. We rewarmed animals by increasing surface temperature at 1°C h-1 and discontinued CHA after Tb reached 36.5°C. Tb, brain temperature (Tbrain), heart rate, blood gas, and electrolytes were also monitored. Results show that titrating dose to adjust for individual variation in response to CHA led to tolerance and failed to manage a prescribed Tb. Starting with a dose (0.25 mg/[kg·h]) and modulating surface temperature to prevent overcooling proved to be an effective means to achieve and maintain Tb between 32°C and 34°C for 24 hours. Increasing surface temperature to 37°C during CHA administration brought Tb back to normothermic levels. All animals treated in this way rewarmed without incident. During the initiation of cooling, we observed bradycardia within 30 minutes of the start of IV infusion, transient hyperglycemia, and a mild hypercapnia; the latter normalized via metabolic compensation. In conclusion, we describe an intravenous delivery protocol for CHA at 0.25 mg/(kg·h) that, when coupled with conductive cooling, achieves and maintains a prescribed and consistent target Tb between 32°C and 34°C for 24 hours.
Collapse
Affiliation(s)
- Bernard W Laughlin
- 1 Institute of Arctic Biology, University of Alaska Fairbanks College of Natural Science and Mathematics , Fairbanks, Alaska.,2 Department of Chemistry and Biochemistry, University of Alaska Fairbanks College of Natural Science and Mathematics , Fairbanks, Alaska
| | - Isaac R Bailey
- 1 Institute of Arctic Biology, University of Alaska Fairbanks College of Natural Science and Mathematics , Fairbanks, Alaska.,2 Department of Chemistry and Biochemistry, University of Alaska Fairbanks College of Natural Science and Mathematics , Fairbanks, Alaska
| | - Sarah A Rice
- 1 Institute of Arctic Biology, University of Alaska Fairbanks College of Natural Science and Mathematics , Fairbanks, Alaska.,2 Department of Chemistry and Biochemistry, University of Alaska Fairbanks College of Natural Science and Mathematics , Fairbanks, Alaska
| | - Zeinab Barati
- 1 Institute of Arctic Biology, University of Alaska Fairbanks College of Natural Science and Mathematics , Fairbanks, Alaska
| | - Lori K Bogren
- 1 Institute of Arctic Biology, University of Alaska Fairbanks College of Natural Science and Mathematics , Fairbanks, Alaska
| | - Kelly L Drew
- 1 Institute of Arctic Biology, University of Alaska Fairbanks College of Natural Science and Mathematics , Fairbanks, Alaska.,2 Department of Chemistry and Biochemistry, University of Alaska Fairbanks College of Natural Science and Mathematics , Fairbanks, Alaska
| |
Collapse
|
24
|
Bucolo C, Platania CBM, Drago F, Bonfiglio V, Reibaldi M, Avitabile T, Uva M. Novel Therapeutics in Glaucoma Management. Curr Neuropharmacol 2018; 16:978-992. [PMID: 28925883 PMCID: PMC6120119 DOI: 10.2174/1570159x15666170915142727] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 06/26/2017] [Accepted: 09/03/2017] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Glaucoma is a progressive optic neuropathy characterized by retinal ganglion cell death and alterations of visual field. Elevated intraocular pressure (IOP) is considered the main risk factor of glaucoma, even though other factors cannot be ruled out, such as epigenetic mechanisms. OBJECTIVE An overview of the ultimate promising experimental drugs to manage glaucoma has been provided. RESULTS In particular, we have focused on purinergic ligands, KATP channel activators, gases (nitric oxide, carbon monoxide and hydrogen sulfide), non-glucocorticoid steroidal compounds, neurotrophic factors, PI3K/Akt activators, citicoline, histone deacetylase inhibitors, cannabinoids, dopamine and serotonin receptors ligands, small interference RNA, and Rho kinase inhibitors. CONCLUSIONS The review has been also endowed of a brief chapter on last reports about potential neuroprotective benefits of anti-glaucoma drugs already present in the market.
Collapse
Affiliation(s)
- Claudio Bucolo
- Address correspondence to this author at the Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; Tel: +39 095 4781196;
| | | | | | | | | | | | | |
Collapse
|
25
|
Melchior JR, Jones SR. Chronic ethanol exposure increases inhibition of optically targeted phasic dopamine release in the nucleus accumbens core and medial shell ex vivo. Mol Cell Neurosci 2017; 85:93-104. [PMID: 28942046 PMCID: PMC5698100 DOI: 10.1016/j.mcn.2017.09.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/08/2017] [Accepted: 09/18/2017] [Indexed: 01/07/2023] Open
Abstract
Dopamine signaling encodes reward learning and motivated behavior through modulation of synaptic signaling in the nucleus accumbens, and aberrations in these processes are thought to underlie obsessive behaviors associated with alcohol abuse. The nucleus accumbens is divided into core and shell sub-regions with overlapping but also divergent contributions to behavior. Here we optogenetically targeted dopamine projections to the accumbens allowing us to isolate stimulation of dopamine terminals ex vivo. We applied 5 pulse (phasic) light stimulations to probe intrinsic differences in dopamine release parameters across regions. Also, we exposed animals to 4weeks of chronic intermittent ethanol vapor and measured phasic release. We found that initial release probability, uptake rate and autoreceptor inhibition were greater in the accumbens core compared to the shell, yet the shell showed greater phasic release ratios. Following chronic ethanol, uptake rates were increased in the core but not the shell, suggesting region-specific neuronal adaptations. Conversely, kappa opioid receptor function was upregulated in both regions to a similar extent, suggesting a local mechanism of kappa opioid receptor regulation that is generalized across the nucleus accumbens. These data suggest that dopamine axons in the nucleus accumbens core and shell display differences in intrinsic release parameters, and that ethanol-induced adaptations to dopamine neuron terminal fields may not be homogeneous. Also, chronic ethanol exposure induces an upregulation in kappa opioid receptor function, providing a mechanism for potential over-inhibition of accumbens dopamine signaling which may negatively impact downstream synaptic function and ultimately bias choice towards previously reinforced alcohol use behaviors.
Collapse
Affiliation(s)
- James R Melchior
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Sara R Jones
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States.
| |
Collapse
|
26
|
The evolution of heart failure with reduced ejection fraction pharmacotherapy: What do we have and where are we going? Pharmacol Ther 2017; 178:67-82. [DOI: 10.1016/j.pharmthera.2017.03.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
27
|
Ning YL, Yang N, Chen X, Zhao ZA, Zhang XZ, Chen XY, Li P, Zhao Y, Zhou YG. Chronic caffeine exposure attenuates blast-induced memory deficit in mice. Chin J Traumatol 2017; 18:204-11. [PMID: 26764541 DOI: 10.1016/j.cjtee.2015.10.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVE To investigate the effects of three different ways of chronic caffeine administration on blast- induced memory dysfunction and to explore the underlying mechanisms. METHODS Adult male C57BL/6 mice were used and randomly divided into five groups: control: without blast exposure, con-water: administrated with water continuously before and after blast-induced traumatic brain injury (bTBI), con-caffeine: administrated with caffeine continuously for 1 month before and after bTBI, pre-caffeine: chronically administrated with caffeine for 1 month before bTBI and withdrawal after bTBI, post-caffeine: chronically administrated with caffeine after bTBI. After being subjected to moderate intensity of blast injury, mice were recorded for learning and memory performance using Morris water maze (MWM) paradigms at 1, 4, and 8 weeks post-blast injury. Neurological deficit scoring, glutamate concentration, proinflammatory cytokines production, and neuropathological changes at 24 h, 1, 4, and 8 weeks post-bTBI were examined to evaluate the brain injury in early and prolonged stages. Adenosine A1 receptor expression was detected using qPCR. RESULTS All of the three ways of chronic caffeine exposure ameliorated blast-induced memory deficit, which is correlated with the neuroprotective effects against excitotoxicity, inflammation, astrogliosis and neuronal loss at different stages of injury. Continuous caffeine treatment played positive roles in both early and prolonged stages of bTBI; pre-bTBI and post-bTBI treatment of caffeine tended to exert neuroprotective effects at early and prolonged stages of bTBI respectively. Up-regulation of adenosine A1 receptor expression might contribute to the favorable effects of chronic caffeine consumption. CONCLUSION Since caffeinated beverages are widely consumed in both civilian and military personnel and are convenient to get, the results may provide a promising prophylactic strategy for blast-induced neurotrauma and the consequent cognitive impairment.
Collapse
Affiliation(s)
- Ya-Lei Ning
- Molecular Biology Center, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital,Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Pharmacological targeting of adenosine receptor signaling. Mol Aspects Med 2017; 55:4-8. [PMID: 28088486 DOI: 10.1016/j.mam.2016.12.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 12/22/2016] [Accepted: 12/23/2016] [Indexed: 12/20/2022]
Abstract
Adenosine receptor signaling plays important roles in normal physiology, but is also known to modulate the development or progression of several different diseases. The design of new, efficient, and safe pharmacological approaches to target the adenosine system may have considerable therapeutic potential, but is also associated with many challenges. This review summarizes the main challenges of adenosine receptor targeted treatment including tolerance, disease stage, cell type-specific effects, caffeine intake, adenosine level assessment and receptor distribution in vivo. Moreover, we discuss several potential ways to overcome these obstacles (i.e., the use of partial agonists, indirect receptor targeting, allosteric enhancers, prodrugs, non-receptor-mediated effects, neoreceptors, conditional knockouts). It is important to address these concerns during development of new and successful therapeutic approaches targeting the adenosine system.
Collapse
|
29
|
Caravan I, Sevastre Berghian A, Moldovan R, Decea N, Orasan R, Filip GA. Modulatory effects of caffeine on oxidative stress and anxiety-like behavior in ovariectomized rats. Can J Physiol Pharmacol 2016; 94:961-72. [DOI: 10.1139/cjpp-2015-0502] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Menopause is accompanied by enhanced oxidative stress and behavioral changes, effects attenuated by antioxidants. The aim of this study was to evaluate the effects of caffeine on behavior and oxidative stress in an experimental model of menopause. Female rats were divided into the following groups: sham-operated (CON), sham-operated and caffeine-treated (CAF), ovariectomized (OVX), ovariectomized and caffeine-treated (OVX+CAF). Caffeine (6 mg/kg) and vehicle were administered for 21 days (subchronic) and 42 days (chronic), using 2 experimental subsets. Behavioral tests and oxidative stress parameters in the blood, whole brain, and hippocampus were assessed. The subchronic administration of caffeine decreased the lipid peroxidation and improved the antioxidant defense in the blood and brain. The GSH/GGSG ratio in the brain was improved by chronic administration, with reduced activities of antioxidant enzymes and enhanced nitric oxide and malondialdehyde levels. In particular, the lipid peroxidation in the hippocampus decreased in both experiments. The rats became hyperactive after 21 days of treatment, but no effect was observed after chronic administration. In both experimental subsets, caffeine had anxiolytic effects as tested in elevated plus maze. The administration of low doses of caffeine, for a short period of time, may be a new therapeutic approach to modulating the oxidative stress and anxiety in menopause.
Collapse
Affiliation(s)
- Ionut Caravan
- Department of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 1 Clinicilor Street, 400006 Cluj-Napoca, Romania
- Department of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 1 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Alexandra Sevastre Berghian
- Department of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 1 Clinicilor Street, 400006 Cluj-Napoca, Romania
- Department of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 1 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Remus Moldovan
- Department of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 1 Clinicilor Street, 400006 Cluj-Napoca, Romania
- Department of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 1 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Nicoleta Decea
- Department of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 1 Clinicilor Street, 400006 Cluj-Napoca, Romania
- Department of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 1 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Remus Orasan
- Department of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 1 Clinicilor Street, 400006 Cluj-Napoca, Romania
- Department of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 1 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Gabriela Adriana Filip
- Department of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 1 Clinicilor Street, 400006 Cluj-Napoca, Romania
- Department of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 1 Clinicilor Street, 400006 Cluj-Napoca, Romania
| |
Collapse
|
30
|
Muqaku B, Tahir A, Klepeisz P, Bileck A, Kreutz D, Mayer RL, Meier SM, Gerner M, Schmetterer K, Gerner C. Coffee consumption modulates inflammatory processes in an individual fashion. Mol Nutr Food Res 2016; 60:2529-2541. [DOI: 10.1002/mnfr.201600328] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 06/22/2016] [Accepted: 06/26/2016] [Indexed: 12/19/2022]
Affiliation(s)
- Besnik Muqaku
- Department of Analytical Chemistry, Faculty of Chemistry; University of Vienna, Vienna; Austria
| | - Ammar Tahir
- Department of Analytical Chemistry, Faculty of Chemistry; University of Vienna, Vienna; Austria
| | - Philip Klepeisz
- Institute of Cancer Research, Department of Medicine I; Medical University of Vienna, Vienna; Austria
| | - Andrea Bileck
- Department of Analytical Chemistry, Faculty of Chemistry; University of Vienna, Vienna; Austria
| | - Dominique Kreutz
- Department of Analytical Chemistry, Faculty of Chemistry; University of Vienna, Vienna; Austria
| | - Rupert L. Mayer
- Department of Analytical Chemistry, Faculty of Chemistry; University of Vienna, Vienna; Austria
| | - Samuel M. Meier
- Department of Analytical Chemistry, Faculty of Chemistry; University of Vienna, Vienna; Austria
| | - Marlene Gerner
- Department of Laboratory Medicine; Medical University of Vienna; Vienna Austria
| | - Klaus Schmetterer
- Department of Laboratory Medicine; Medical University of Vienna; Vienna Austria
| | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry; University of Vienna, Vienna; Austria
| |
Collapse
|
31
|
Cunha RA. How does adenosine control neuronal dysfunction and neurodegeneration? J Neurochem 2016; 139:1019-1055. [PMID: 27365148 DOI: 10.1111/jnc.13724] [Citation(s) in RCA: 335] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 05/23/2016] [Accepted: 06/23/2016] [Indexed: 12/11/2022]
Abstract
The adenosine modulation system mostly operates through inhibitory A1 (A1 R) and facilitatory A2A receptors (A2A R) in the brain. The activity-dependent release of adenosine acts as a brake of excitatory transmission through A1 R, which are enriched in glutamatergic terminals. Adenosine sharpens salience of information encoding in neuronal circuits: high-frequency stimulation triggers ATP release in the 'activated' synapse, which is locally converted by ecto-nucleotidases into adenosine to selectively activate A2A R; A2A R switch off A1 R and CB1 receptors, bolster glutamate release and NMDA receptors to assist increasing synaptic plasticity in the 'activated' synapse; the parallel engagement of the astrocytic syncytium releases adenosine further inhibiting neighboring synapses, thus sharpening the encoded plastic change. Brain insults trigger a large outflow of adenosine and ATP, as a danger signal. A1 R are a hurdle for damage initiation, but they desensitize upon prolonged activation. However, if the insult is near-threshold and/or of short-duration, A1 R trigger preconditioning, which may limit the spread of damage. Brain insults also up-regulate A2A R, probably to bolster adaptive changes, but this heightens brain damage since A2A R blockade affords neuroprotection in models of epilepsy, depression, Alzheimer's, or Parkinson's disease. This initially involves a control of synaptotoxicity by neuronal A2A R, whereas astrocytic and microglia A2A R might control the spread of damage. The A2A R signaling mechanisms are largely unknown since A2A R are pleiotropic, coupling to different G proteins and non-canonical pathways to control the viability of glutamatergic synapses, neuroinflammation, mitochondria function, and cytoskeleton dynamics. Thus, simultaneously bolstering A1 R preconditioning and preventing excessive A2A R function might afford maximal neuroprotection. The main physiological role of the adenosine modulation system is to sharp the salience of information encoding through a combined action of adenosine A2A receptors (A2A R) in the synapse undergoing an alteration of synaptic efficiency with an increased inhibitory action of A1 R in all surrounding synapses. Brain insults trigger an up-regulation of A2A R in an attempt to bolster adaptive plasticity together with adenosine release and A1 R desensitization; this favors synaptotocity (increased A2A R) and decreases the hurdle to undergo degeneration (decreased A1 R). Maximal neuroprotection is expected to result from a combined A2A R blockade and increased A1 R activation. This article is part of a mini review series: "Synaptic Function and Dysfunction in Brain Diseases".
Collapse
Affiliation(s)
- 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
|
32
|
Structure-Bioactivity Relationships of Methylxanthines: Trying to Make Sense of All the Promises and the Drawbacks. Molecules 2016; 21:molecules21080974. [PMID: 27472311 PMCID: PMC6273298 DOI: 10.3390/molecules21080974] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 07/02/2016] [Accepted: 07/19/2016] [Indexed: 12/05/2022] Open
Abstract
Methylxanthines are a group of phytochemicals derived from the purine base xanthine and obtained from plant secondary metabolism. They are unobtrusively included in daily diet in common products as coffee, tea, energetic drinks, or chocolate. Caffeine is by far the most studied methylxanthine either in animal or epidemiologic studies. Theophylline and theobromine are other relevant methylxanthines also commonly available in the aforementioned sources. There are many disseminated myths about methylxanthines but there is increased scientific knowledge to discuss all the controversy and promise shown by these intriguing phytochemicals. In fact, many beneficial physiologic outcomes have been suggested for methylxanthines in areas as important and diverse as neurodegenerative and respiratory diseases, diabetes or cancer. However, there have always been toxicity concerns with methylxanthine (over)consumption and pharmacologic applications. Herein, we explore the structure-bioactivity relationships to bring light those enumerated effects. The potential shown by methylxanthines in such a wide range of conditions should substantiate many other scientific endeavors that may highlight their adequacy as adjuvant therapy agents and may contribute to the advent of functional foods. Newly designed targeted molecules based on methylxanthine structure may originate more specific and effective outcomes.
Collapse
|
33
|
Abstract
Adenosine, as the brain’s endogenous anticonvulsant, is considered to be responsible for seizure arrest and postictal refractoriness. On the other hand, deficiencies within the adenosine-based neuromodulatory system may contribute to epileptogenesis. Based on these natural mechanisms and on findings that adenosine and its analogs can suppress pharmacoresistant seizures, a new field of adenosine-based therapies has emerged, including the use of adenosine receptor agonists and adenosine transport inhibitors, or the inhibition of adenosine kinase, which is thought to be the key enzyme for the regulation of intra- and extracellular adenosine levels. However, most of these pharmacological approaches are limited by strong systemic side effects ranging from a decrease of heart rate, blood pressure, and body temperature to sedation. Recently, new strategies have been developed aimed at the local reconstitution of the inhibitory adenosinergic tone by intracerebral implantation of cells engineered to release adenosine. Adenosine-releasing cells or devices implanted into or near a seizure focus offer new hopes for a side effect-free therapy for pharmacoresistant epilepsy.
Collapse
Affiliation(s)
- Detlev Boison
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
34
|
Ferré S. Mechanisms of the psychostimulant effects of caffeine: implications for substance use disorders. Psychopharmacology (Berl) 2016; 233:1963-79. [PMID: 26786412 PMCID: PMC4846529 DOI: 10.1007/s00213-016-4212-2] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 01/09/2016] [Indexed: 11/28/2022]
Abstract
BACKGROUND The psychostimulant properties of caffeine are reviewed and compared with those of prototypical psychostimulants able to cause substance use disorders (SUD). Caffeine produces psychomotor-activating, reinforcing, and arousing effects, which depend on its ability to disinhibit the brake that endogenous adenosine imposes on the ascending dopamine and arousal systems. OBJECTIVES A model that considers the striatal adenosine A2A-dopamine D2 receptor heteromer as a key modulator of dopamine-dependent striatal functions (reward-oriented behavior and learning of stimulus-reward and reward-response associations) is introduced, which should explain most of the psychomotor and reinforcing effects of caffeine. HIGHLIGHTS The model can explain the caffeine-induced rotational behavior in rats with unilateral striatal dopamine denervation and the ability of caffeine to reverse the adipsic-aphagic syndrome in dopamine-deficient rodents. The model can also explain the weaker reinforcing effects and low abuse liability of caffeine, compared with prototypical psychostimulants. Finally, the model can explain the actual major societal dangers of caffeine: the ability of caffeine to potentiate the addictive and toxic effects of drugs of abuse, with the particularly alarming associations of caffeine (as adulterant) with cocaine, amphetamine derivatives, synthetic cathinones, and energy drinks with alcohol, and the higher sensitivity of children and adolescents to the psychostimulant effects of caffeine and its potential to increase vulnerability to SUD. CONCLUSIONS The striatal A2A-D2 receptor heteromer constitutes an unequivocal main pharmacological target of caffeine and provides the main mechanisms by which caffeine potentiates the acute and long-term effects of prototypical psychostimulants.
Collapse
Affiliation(s)
- Sergi Ferré
- Integrative Neurobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Triad Technology Building, 333 Cassell Drive, Baltimore, MD, 21224, USA.
| |
Collapse
|
35
|
Klaft ZJ, Hollnagel JO, Salar S, Calişkan G, Schulz SB, Schneider UC, Horn P, Koch A, Holtkamp M, Gabriel S, Gerevich Z, Heinemann U. Adenosine A1 receptor-mediated suppression of carbamazepine-resistant seizure-like events in human neocortical slices. Epilepsia 2016; 57:746-56. [PMID: 27087530 DOI: 10.1111/epi.13360] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The need for alternative pharmacologic strategies in treatment of epilepsies is pressing for about 30% of patients with epilepsy who do not experience satisfactory seizure control with present treatments. In temporal lobe epilepsy (TLE) even up to 80% of patients are pharmacoresistant, and surgical resection of the ictogenic tissue is only possible for a minority of TLE patients. In this study we investigate purinergic modulation of drug-resistant seizure-like events (SLEs) in human temporal cortex slices. METHODS Layer V/VI field potentials from a total of 77 neocortical slices from 17 pharmacoresistant patients were recorded to monitor SLEs induced by application of 8 mM [K(+) ] and 50 μm bicuculline. RESULTS Activating A1 receptors with a specific agonist completely suppressed SLEs in 73% of human temporal cortex slices. In the remaining slices, incidence of SLEs was markedly reduced. Because a subportion of slices can be pharmacosensitive, we tested effects of an A1 agonist, in slices insensitive to a high dose of carbamazepine (50 μm). Also in these cases the A1 agonist was equally efficient. Moreover, ATP and adenosine blocked or modulated SLEs, an effect mediated not by P2 receptors but rather by adenosine A1 receptors. SIGNIFICANCE Selective activation of A1 receptors mediates a strong anticonvulsant action in human neocortical slices from pharmacoresistant patients. We propose that our human slice model of seizure-like activity is a feasible option for future studies investigating new antiepileptic drug (AED) candidates.
Collapse
Affiliation(s)
- Zin-Juan Klaft
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jan-Oliver Hollnagel
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Seda Salar
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Gürsel Calişkan
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Steffen B Schulz
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ulf C Schneider
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Peter Horn
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Arend Koch
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Martin Holtkamp
- Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Epilepsy-Center Berlin-Brandenburg, Krankenhaus Königin Elisabeth Herzberge, Berlin, Germany
| | - Siegrun Gabriel
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Zoltan Gerevich
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Uwe Heinemann
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Neuroscience Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
36
|
Caffeine promotes wakefulness via dopamine signaling in Drosophila. Sci Rep 2016; 6:20938. [PMID: 26868675 PMCID: PMC4751479 DOI: 10.1038/srep20938] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 12/10/2015] [Indexed: 12/12/2022] Open
Abstract
Caffeine is the most widely-consumed psychoactive drug in the world, but our understanding of how caffeine affects our brains is relatively incomplete. Most studies focus on effects of caffeine on adenosine receptors, but there is evidence for other, more complex mechanisms. In the fruit fly Drosophila melanogaster, which shows a robust diurnal pattern of sleep/wake activity, caffeine reduces nighttime sleep behavior independently of the one known adenosine receptor. Here, we show that dopamine is required for the wake-promoting effect of caffeine in the fly, and that caffeine likely acts presynaptically to increase dopamine signaling. We identify a cluster of neurons, the paired anterior medial (PAM) cluster of dopaminergic neurons, as the ones relevant for the caffeine response. PAM neurons show increased activity following caffeine administration, and promote wake when activated. Also, inhibition of these neurons abrogates sleep suppression by caffeine. While previous studies have focused on adenosine-receptor mediated mechanisms for caffeine action, we have identified a role for dopaminergic neurons in the arousal-promoting effect of caffeine.
Collapse
|
37
|
Donegan RK, Lieberman RL. Discovery of Molecular Therapeutics for Glaucoma: Challenges, Successes, and Promising Directions. J Med Chem 2016; 59:788-809. [PMID: 26356532 PMCID: PMC5547565 DOI: 10.1021/acs.jmedchem.5b00828] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Glaucoma, a heterogeneous ocular disorder affecting ∼60 million people worldwide, is characterized by painless neurodegeneration of retinal ganglion cells (RGCs), resulting in irreversible vision loss. Available therapies, which decrease the common causal risk factor of elevated intraocular pressure, delay, but cannot prevent, RGC death and blindness. Notably, it is changes in the anterior segment of the eye, particularly in the drainage of aqueous humor fluid, which are believed to bring about changes in pressure. Thus, it is primarily this region whose properties are manipulated in current and emerging therapies for glaucoma. Here, we focus on the challenges associated with developing treatments, review the available experimental methods to evaluate the therapeutic potential of new drugs, describe the development and evaluation of emerging Rho-kinase inhibitors and adenosine receptor ligands that offer the potential to improve aqueous humor outflow and protect RGCs simultaneously, and present new targets and approaches on the horizon.
Collapse
Affiliation(s)
- Rebecca K Donegan
- School of Chemistry and Biochemistry, Georgia Institute of Technology , 901 Atlantic Drive NW, Atlanta, Georgia 30332-0400, United States
| | - Raquel L Lieberman
- School of Chemistry and Biochemistry, Georgia Institute of Technology , 901 Atlantic Drive NW, Atlanta, Georgia 30332-0400, United States
| |
Collapse
|
38
|
Abstract
G protein-coupled receptors (GPCRs) remain a major domain of pharmaceutical discovery. The identification of GPCR lead compounds and their optimization are now structure-based, thanks to advances in X-ray crystallography, molecular modeling, protein engineering and biophysical techniques. In silico screening provides useful hit molecules. New pharmacological approaches to tuning the pleotropic action of GPCRs include: allosteric modulators, biased ligands, GPCR heterodimer-targeted compounds, manipulation of polypharmacology, receptor antibodies and tailoring of drug molecules to fit GPCR pharmacogenomics. Measurements of kinetics and drug efficacy are factors influencing clinical success. With the exception of inhibitors of GPCR kinases, targeting of intracellular GPCR signaling or receptor cycling for therapeutic purposes remains a futuristic concept. New assay approaches are more efficient and multidimensional: cell-based, label-free, fluorescence-based assays, and biosensors. Tailoring GPCR drugs to a patient's genetic background is now being considered. Chemoinformatic tools can predict ADME-tox properties. New imaging technology visualizes drug action in vivo. Thus, there is reason to be optimistic that new technology for GPCR ligand discovery will help reverse the current narrowing of the pharmaceutical pipeline.
Collapse
Affiliation(s)
- Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bldg. 8A, Rm. B1A-19, Bethesda, Maryland 20892, USA.
| |
Collapse
|
39
|
Nazario LR, Antonioli RJ, Capiotti KM, Hallak JEC, Zuardi AW, Crippa JAS, Bonan CD, da Silva RS. Reprint of "Caffeine protects against memory loss induced by high and non-anxiolytic dose of cannabidiol in adult zebrafish (Danio rerio)". Pharmacol Biochem Behav 2015; 139 Pt B:134-40. [PMID: 26569549 DOI: 10.1016/j.pbb.2015.11.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 06/03/2015] [Accepted: 06/13/2015] [Indexed: 12/18/2022]
Abstract
Cannabidiol (CBD) has been investigated in a wide spectrum of clinical approaches due to its psychopharmacological properties. CBD has low affinity for cannabinoid neuroreceptors and agonistic properties to 5-HT receptors. An interaction between cannabinoid and purinergic receptor systems has been proposed. The purpose of this study is to evaluate CBD properties on memory behavioral and locomotor parameters and the effects of pre-treatment of adenosine receptor blockers on CBD impacts on memory using adult zebrafish. CBD (0.1, 0.5, 5, and 10mg/kg) was tested in the avoidance inhibitory paradigm and anxiety task. We analyzed the effect of a long-term caffeine pre-treatment (~20mg/L - four months). Also, acute block of adenosine receptors was performed in co-administration with CBD exposure in the memory assessment. CBD promoted an inverted U-shaped dose-response curve in the anxiety task; in the memory assessment, CBD in the dose of 5mg/Kg promoted the strongest effects without interfering with social and aggressive behavior. Caffeine treatment was able to prevent CBD (5mg/kg) effects on memory when CBD was given after the training session. CBD effects on memory were partially prevented by co-treatment with a specific A2A adenosine receptor antagonist when given prior to or after the training session, while CBD effects after the training session were fully prevented by adenosine A1 receptor antagonist. These results indicated that zebrafish have responses to CBD anxiolytic properties that are comparable to other animal models, and high doses changed memory retention in a way dependent on adenosine.
Collapse
Affiliation(s)
- Luiza Reali Nazario
- Laboratório de Neuroquímica e Psicofarmacologia, Departamento de Biologia Celular e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, Caixa Postal 1429, 90619-900 Porto Alegre, RS, Brazil
| | - Régis Junior Antonioli
- Laboratório de Neuroquímica e Psicofarmacologia, Departamento de Biologia Celular e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, Caixa Postal 1429, 90619-900 Porto Alegre, RS, Brazil
| | - Katiucia Marques Capiotti
- Laboratório de Neuroquímica e Psicofarmacologia, Departamento de Biologia Celular e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, Caixa Postal 1429, 90619-900 Porto Alegre, RS, Brazil
| | - Jaime Eduardo Cecílio Hallak
- Departamento de Neurociências e Ciências do Comportamento, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Brazil; Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), 90035-003 Porto Alegre, RS, Brazil
| | - Antonio Waldo Zuardi
- Departamento de Neurociências e Ciências do Comportamento, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Brazil; Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), 90035-003 Porto Alegre, RS, Brazil
| | - José Alexandre S Crippa
- Departamento de Neurociências e Ciências do Comportamento, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Brazil; Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), 90035-003 Porto Alegre, RS, Brazil
| | - Carla Denise Bonan
- Laboratório de Neuroquímica e Psicofarmacologia, Departamento de Biologia Celular e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, Caixa Postal 1429, 90619-900 Porto Alegre, RS, Brazil; Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), 90035-003 Porto Alegre, RS, Brazil
| | - Rosane Souza da Silva
- Laboratório de Neuroquímica e Psicofarmacologia, Departamento de Biologia Celular e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, Caixa Postal 1429, 90619-900 Porto Alegre, RS, Brazil; Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), 90035-003 Porto Alegre, RS, Brazil.
| |
Collapse
|
40
|
Pedata F, Dettori I, Coppi E, Melani A, Fusco I, Corradetti R, Pugliese AM. Purinergic signalling in brain ischemia. Neuropharmacology 2015; 104:105-30. [PMID: 26581499 DOI: 10.1016/j.neuropharm.2015.11.007] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 11/04/2015] [Accepted: 11/06/2015] [Indexed: 12/18/2022]
Abstract
Ischemia is a multifactorial pathology characterized by different events evolving in the time. After ischemia a primary damage due to the early massive increase of extracellular glutamate is followed by activation of resident immune cells, i.e microglia, and production or activation of inflammation mediators. Protracted neuroinflammation is now recognized as the predominant mechanism of secondary brain injury progression. Extracellular concentrations of ATP and adenosine in the brain increase dramatically during ischemia in concentrations able to stimulate their respective specific P2 and P1 receptors. Both ATP P2 and adenosine P1 receptor subtypes exert important roles in ischemia. Although adenosine exerts a clear neuroprotective effect through A1 receptors during ischemia, the use of selective A1 agonists is hampered by undesirable peripheral effects. Evidence up to now in literature indicate that A2A receptor antagonists provide protection centrally by reducing excitotoxicity, while agonists at A2A (and possibly also A2B) and A3 receptors provide protection by controlling massive infiltration and neuroinflammation in the hours and days after brain ischemia. Among P2X receptors most evidence indicate that P2X7 receptor contribute to the damage induced by the ischemic insult due to intracellular Ca(2+) loading in central cells and facilitation of glutamate release. Antagonism of P2X7 receptors might represent a new treatment to attenuate brain damage and to promote proliferation and maturation of brain immature resident cells that can promote tissue repair following cerebral ischemia. Among P2Y receptors, antagonists of P2Y12 receptors are of value because of their antiplatelet activity and possibly because of additional anti-inflammatory effects. Moreover strategies that modify adenosine or ATP concentrations at injury sites might be of value to limit damage after ischemia. This article is part of the Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.
Collapse
Affiliation(s)
- Felicita Pedata
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy.
| | - Ilaria Dettori
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Elisabetta Coppi
- Department of Health Sciences, University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Alessia Melani
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Irene Fusco
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Renato Corradetti
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Anna Maria Pugliese
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| |
Collapse
|
41
|
Caffeine protects against memory loss induced by high and non-anxiolytic dose of cannabidiol in adult zebrafish (Danio rerio). Pharmacol Biochem Behav 2015; 135:210-6. [DOI: 10.1016/j.pbb.2015.06.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 06/03/2015] [Accepted: 06/13/2015] [Indexed: 02/02/2023]
|
42
|
Chiu GS, Freund GG. Modulation of neuroimmunity by adenosine and its receptors: metabolism to mental illness. Metabolism 2014; 63:1491-8. [PMID: 25308443 PMCID: PMC4252699 DOI: 10.1016/j.metabol.2014.09.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Revised: 08/14/2014] [Accepted: 09/19/2014] [Indexed: 12/20/2022]
Abstract
Adenosine is a pleiotropic bioactive with potent neuromodulatory properties. Due to its ability to easily cross the blood-brain barrier, it can act as a signaling molecule between the periphery and the brain. It functions through four (A1, A2A, A2B, and A3) cell surface G protein-coupled adenosine receptors (ARs) that are expressed in some combination on nearly all cells types within the CNS. By regulating the activity of adenylyl cyclase and changing the intracellular concentration of cAMP, adenosine can alter neuronal function and neurotransmission. A variety of illnesses related to metabolic dysregulation, such as type 1 diabetes and Alzheimer's disease, are associated with an elevated serum concentration of adenosine and a pathogenesis rooted in inflammation. This review describes the accepted physiologic function of adenosine in neurological disease and explores its new potential as a peripheral to central danger signal that can activate the neuroimmune system and contribute to symptoms of sickness and psychopathologies.
Collapse
Affiliation(s)
- Gabriel S Chiu
- Division of Nutritional Sciences, University of Illinois, Urbana IL, USA; Department of Pathology, Program in Integrative Immunology and Behavior, University of Illinois, Urbana IL, USA
| | - Gregory G Freund
- Division of Nutritional Sciences, University of Illinois, Urbana IL, USA; Department of Pathology, Program in Integrative Immunology and Behavior, University of Illinois, Urbana IL, USA; Department of Animal Sciences, University of Illinois, Urbana IL, USA.
| |
Collapse
|
43
|
Fardet A, Boirie Y. Associations between food and beverage groups and major diet-related chronic diseases: an exhaustive review of pooled/meta-analyses and systematic reviews. Nutr Rev 2014; 72:741-62. [PMID: 25406801 DOI: 10.1111/nure.12153] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Associations between food and beverage groups and the risk of diet-related chronic disease (DRCD) have been the subject of intensive research in preventive nutrition. Pooled/meta-analyses and systematic reviews (PMASRs) aim to better characterize these associations. To date, however, there has been no attempt to synthesize all PMASRs that have assessed the relationship between food and beverage groups and DRCDs. The objectives of this review were to aggregate PMASRs to obtain an overview of the associations between food and beverage groups (n = 17) and DRCDs (n = 10) and to establish new directions for future research needs. The present review of 304 PMASRs published between 1950 and 2013 confirmed that plant food groups are more protective than animal food groups against DRCDs. Within plant food groups, grain products are more protective than fruits and vegetables. Among animal food groups, dairy/milk products have a neutral effect on the risk of DRCDs, while red/processed meats tend to increase the risk. Among beverages, tea was the most protective and soft drinks the least protective against DRCDs. For two of the DRCDs examined, sarcopenia and kidney disease, no PMASR was found. Overweight/obesity, type 2 diabetes, and various types of cardiovascular disease and cancer accounted for 289 of the PMASRs. There is a crucial need to further study the associations between food and beverage groups and mental health, skeletal health, digestive diseases, liver diseases, kidney diseases, obesity, and type 2 diabetes.
Collapse
Affiliation(s)
- Anthony Fardet
- Unité de Nutrition Humaine, INRA de Theix & Université d'Auvergne, 63122, Saint-Genès-Champanelle, Auvergne, France
| | | |
Collapse
|
44
|
Dubroqua S, Yee BK, Singer P. Sensorimotor gating is disrupted by acute but not chronic systemic exposure to caffeine in mice. Psychopharmacology (Berl) 2014; 231:4087-98. [PMID: 24728602 DOI: 10.1007/s00213-014-3548-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 03/15/2014] [Indexed: 12/14/2022]
Abstract
RATIONALE Caffeine is a psychostimulant drug that blocks adenosine A₁ and A₂A receptors (A₁Rs and A₂ARs). However, its ability to disrupt early sensory gating as indexed by prepulse inhibition (PPI), which is consistently disrupted by other psychostimulant agents, has never been convincingly demonstrated. OBJECTIVES To compare the impact of caffeine on PPI expression in C57BL/6 mice by two dose-response experiments differing in terms of chronicity, regimen, and route of administration. To study separately the acute effect of selective antagonists against A₁R or A₂AR. METHODS Caffeine (10, 30, 100 mg/kg, intraperitoneal (i.p.)) was either administered shortly before testing or via caffeinated drinking water (0.3, 1.0, 2 g/l) in home cages over 3 weeks. Two separate dose-response studies tested the acute effect of the selective A₁R antagonist, 1,3 dipropyl-8 cyclopentyl xanthine (DPCPX), and the selective A₂AR antagonist, 5-amino-7-(2-phenylethyl)-2-(2-furyl)-pyrazolo-[4,3-e]-1,2,4-triazolo[1,5-c] (SCH 58261) (0.2, 1.0, 5.0 mg/kg, i.p.). The two drugs were combined in a final experiment to identify their potential synergistic interaction. RESULTS While the two lower acute doses of caffeine attenuated PPI, the highest dose potentiated PPI. By contrast, chronic caffeine exposure did not affect PPI. Neither DPCPX nor SCH 58261 altered PPI, and no synergism was observed when the two drugs were combined. CONCLUSIONS This is the first demonstration that acute caffeine disrupts PPI, but the relative contribution of A₁R and A₂AR blockade remains unclear, and possible non-adenosinergic mechanisms cannot be ruled out. The null effect under chronic caffeine exposure might involve the development of tolerance, but the precise receptor subtypes involved also warrant further investigation.
Collapse
Affiliation(s)
- Sylvain Dubroqua
- Laboratory of Behavioral Neuroscience, Legacy Research Institute, 1225 NE Second Avenue, Portland, OR, 97232, USA
| | | | | |
Collapse
|
45
|
Antonioli L, Csóka B, Fornai M, Colucci R, Kókai E, Blandizzi C, Haskó G. Adenosine and inflammation: what's new on the horizon? Drug Discov Today 2014; 19:1051-68. [DOI: 10.1016/j.drudis.2014.02.010] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 02/06/2014] [Accepted: 02/25/2014] [Indexed: 12/18/2022]
|
46
|
Tchekalarova JD, Kubová H, Mareš P. Different effects of postnatal caffeine treatment on two pentylenetetrazole-induced seizure models persist into adulthood. Pharmacol Rep 2014; 65:847-53. [PMID: 24145078 DOI: 10.1016/s1734-1140(13)71065-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 02/05/2013] [Indexed: 11/17/2022]
Abstract
BACKGROUND Postnatal treatment with caffeine from P7 to P11 (10 or 20 mg/kg daily) resulted in transient changes in two pentylenetetrazole (PTZ)-induced models of epileptic seizures characterized by spike-and-wave EEG rhythm in immature rats. To know if some changes persist into adulthood we studied these models in young adult Wistar rats. METHODS Caffeine treatment at a daily dose of 10 and/or 20 mg/kg, sc was executed during postnatal days 7-11. Rhythmic metrazol activity (RMA, model of human absences) was induced in 60-day old rats by two successive doses of PTZ (20 + 20 mg/kg, ip) while for induction of minimal clonic seizures (model of human myoclonic seizures) the second dose of PTZ was 40 mg/kg. RESULTS RMA episodes elicited by the 20 + 20 mg/kg dose of PTZ in adult rats exposed to caffeine at P7 to P11 were decreased. This effect was more pronounced in group treated with the higher dose of caffeine. In contrast, the lower dose of caffeine exacerbated minimal clonic seizures (both incidence and intensity were increased). In addition, some animals from the 20-mg/kg caffeine group exhibited transition to generalized tonic-clonic seizures. CONCLUSION Different effects of postnatal caffeine exposure persist into adulthood; the seizure ameliorating effects in a model of absences and seizure exacerbating action in a model of myoclonic seizures are dose-specific.
Collapse
Affiliation(s)
- Jana D Tchekalarova
- CZ-142 20, Academy of Sciences of Czech Republic, Institute of Physiology, Videňská 1083, Prague 4, Czech Republic.
| | | | | |
Collapse
|
47
|
Chen JF. Adenosine receptor control of cognition in normal and disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2014; 119:257-307. [PMID: 25175970 DOI: 10.1016/b978-0-12-801022-8.00012-x] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Adenosine and adenosine receptors (ARs) are increasingly recognized as important therapeutic targets for controlling cognition under normal and disease conditions for its dual roles of neuromodulation as well as of homeostatic function in the brain. This chapter first presents the unique ability of adenosine, by acting on the inhibitory A1 and facilitating A2A receptor, to integrate dopamine, glutamate, and BNDF signaling and to modulate synaptic plasticity (e.g., long-term potentiation and long-term depression) in brain regions relevant to learning and memory, providing the molecular and cellular bases for adenosine receptor (AR) control of cognition. This led to the demonstration of AR modulation of social recognition memory, working memory, reference memory, reversal learning, goal-directed behavior/habit formation, Pavlovian fear conditioning, and effort-related behavior. Furthermore, human and animal studies support that AR activity can also, through cognitive enhancement and neuroprotection, reverse cognitive impairments in animal models of Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease, and schizophrenia. Lastly, epidemiological evidence indicates that regular human consumption of caffeine, the most widely used psychoactive drug and nonselective AR antagonists, is associated with the reduced cognitive decline in aging and AD patients, and with the reduced risk in developing PD. Thus, there is a convergence of the molecular studies revealing AR as molecular targets for integrating neurotransmitter signaling and controlling synaptic plasticity, with animal studies demonstrating the strong procognitive impact upon AR antagonism in normal and disease brains and with epidemiological and clinical evidences in support of caffeine and AR drugs for therapeutic modulation of cognition. Since some of adenosine A2A receptor antagonists are already in phase III clinical trials for motor benefits in PD patients with remarkable safety profiles, additional animal and human studies to better understand the mechanism underlying the AR-mediated control of cognition under normal and disease conditions will provide the required rationale to stimulate the necessary clinical investigation to rapidly translate adenosine and AR drug as a novel strategy to control memory impairment in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jiang-Fan Chen
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA; The Molecular Medicine Institute, Wenzhou Medical University, Wenzhou, Zhejiang, PR China.
| |
Collapse
|
48
|
|
49
|
Tchekalarova J, Kubová H, Mareš P. Effects of caffeine on cortical epileptic afterdischarges in adult rats are modulated by postnatal treatment. Acta Neurol Belg 2013; 113:493-500. [PMID: 23864335 DOI: 10.1007/s13760-013-0233-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Accepted: 07/08/2013] [Indexed: 11/29/2022]
Abstract
We have previously shown that the effect of acute caffeine injection on cortical epileptic afterdischarges (ADs) in immature rats can be modulated by the postnatal period of repeated caffeine treatment and age of testing during development. To know if these changes persist into adulthood, in the present study adult rats, previously exposed to caffeine from postnatal day 7-11 (10 and 20 mg/kg), and injected with caffeine (10 and 20 mg/kg), respectively, at P67 were compared with groups of naive rats on cortical epileptic ADs. Low-frequency stimulation of sensorimotor cortical area was applied repeatedly with increasing intensity of stimulation current. The acute caffeine injection decreased the thresholds for both the spike-and-wave ADs and accompanying clonic seizures in naive adult rats. In contrast, the acute caffeine administration applied to rats with postnatal caffeine treatment did not change the thresholds for ADs and clonic seizures. In addition, a shorter duration of ADs was registered at some stimulation intensities in rats with early postnatal caffeine administration. Present results demonstrated failure of proconvulsant effects of caffeine after repeated caffeine administration during the early postnatal period persisting up to adulthood.
Collapse
Affiliation(s)
- Jana Tchekalarova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 23, 1113, Sofia, Bulgaria,
| | | | | |
Collapse
|
50
|
Pitchon DN, Zook M, Rhoads DE. A Pattern of Adolescent Caffeine Consumption that Reduces Alcohol Withdrawal Severity. JOURNAL OF CAFFEINE RESEARCH 2013. [DOI: 10.1089/jcr.2013.0011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Darsi N. Pitchon
- Department of Biology, Monmouth University, W. Long Branch, New Jersey
| | - Michelle Zook
- Department of Biology, Monmouth University, W. Long Branch, New Jersey
| | - Dennis E. Rhoads
- Department of Biology, Monmouth University, W. Long Branch, New Jersey
| |
Collapse
|