Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 303
Filter
1.
Mol Pharmacol ; 103(6): 311-324, 2023 06.
Article in English | MEDLINE | ID: mdl-36894319

ABSTRACT

KW-6356 is a novel adenosine A2A (A2A) receptor antagonist/inverse agonist, and its efficacy as monotherapy in Parkinson's disease (PD) patients has been reported. Istradefylline is a first-generation A2A receptor antagonist approved for use as adjunct treatment to levodopa/decarboxylase inhibitor in adult PD patients experiencing "OFF" episodes. In this study, we investigated the in vitro pharmacological profile of KW-6356 as an A2A receptor antagonist/inverse agonist and the mode of antagonism and compared them with istradefylline. In addition, we determined cocrystal structures of A2A receptor in complex with KW-6356 and istradefylline to explore the structural basis of the antagonistic properties of KW-6356. Pharmacological studies have shown that KW-6356 is a potent and selective ligand for the A2A receptor (the -log of inhibition constant = 9.93 ± 0.01 for human receptor) with a very low dissociation rate from the receptor (the dissociation kinetic rate constant = 0.016 ± 0.006 minute-1 for human receptor). In particular, in vitro functional studies indicated that KW-6356 exhibits insurmountable antagonism and inverse agonism, whereas istradefylline exhibits surmountable antagonism. Crystallography of KW-6356- and istradefylline-bound A2A receptor have indicated that interactions with His2506.52 and Trp2466.48 are essential for the inverse agonism, whereas the interactions at both deep inside the orthosteric pocket and the pocket lid stabilizing the extracellular loop conformation may contribute to the insurmountable antagonism of KW-6356. These profiles may reflect important differences in vivo and help predict better clinical performance. SIGNIFICANCE STATEMENT: KW-6356 is a potent and selective adenosine A2A receptor antagonist/inverse agonist and exhibits insurmountable antagonism, whereas istradefylline, a first-generation adenosine A2A receptor antagonist, exhibits surmountable antagonism. Structural studies of adenosine A2A receptor in complex with KW-6356 and istradefylline explain the characteristic differences in the pharmacological properties of KW-6356 and istradefylline.


Subject(s)
Adenosine A2 Receptor Antagonists , Drug Inverse Agonism , Parkinson Disease , Receptor, Adenosine A2A , Humans , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/therapeutic use , Levodopa/pharmacology , Levodopa/therapeutic use , Receptor, Adenosine A2A/physiology
2.
FASEB J ; 36(4): e22214, 2022 04.
Article in English | MEDLINE | ID: mdl-35230706

ABSTRACT

Adenosine is a local mediator that regulates changes in the cardiovascular system via activation of four G protein-coupled receptors (A1 , A2A , A2B , A3 ). Here, we have investigated the effect of A2A and A2B -selective agonists on vasodilatation in three distinct vascular beds of the rat cardiovascular system. NanoBRET ligand binding studies were used to confirm receptor selectivity. The regional hemodynamic effects of adenosine A2A and A2B selective agonists were investigated in conscious rats. Male Sprague-Dawley rats (350-450 g) were chronically implanted with pulsed Doppler flow probes on the renal artery, mesenteric artery, and the descending abdominal aorta. Cardiovascular responses were measured following intravenous infusion (3 min for each dose) of the A2A -selective agonist CGS 21680 (0.1, 0.3, 1 µg kg-1 min-1 ) or the A2B -selective agonist BAY 60-6583 (4,13.3, 40 µg kg-1 min-1 ) following predosing with the A2A -selective antagonist SCH 58261 (0.1 or 1 mg kg-1 min-1 ), the A2B /A2A antagonist PSB 1115 (10 mg kg-1 min-1 ) or vehicle. The A2A -selective agonist CGS 21680 produced a striking increase in heart rate (HR) and hindquarters vascular conductance (VC) that was accompanied by a significant decrease in mean arterial pressure (MAP) in conscious rats. In marked contrast, the A2B -selective agonist BAY 60-6583 significantly increased HR and VC in the renal and mesenteric vascular beds, but not in the hindquarters. Taken together, these data indicate that A2A and A2B receptors are regionally selective in their regulation of vascular tone. These results suggest that the development of A2B receptor agonists to induce vasodilatation in the kidney may provide a good therapeutic approach for the treatment of acute kidney injury.


Subject(s)
Adenosine A2 Receptor Agonists/pharmacology , Hemodynamics/drug effects , Receptor, Adenosine A2A/physiology , Receptor, Adenosine A2B/physiology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Aminopyridines/pharmacology , Animals , HEK293 Cells , Humans , Kidney/blood supply , Kidney/drug effects , Male , Phenethylamines/pharmacology , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Triazoles/pharmacology , Vasodilation/drug effects , Xanthines/pharmacology
3.
Cells ; 10(9)2021 09 07.
Article in English | MEDLINE | ID: mdl-34571993

ABSTRACT

Alzheimer's disease (AD) is one of the most common neurodegenerative pathologies. Its incidence is in dramatic growth in Western societies and there is a need of both biomarkers to support the clinical diagnosis and drugs for the treatment of AD. The diagnostic criteria of AD are based on clinical data. However, it is necessary to develop biomarkers considering the neuropathology of AD. The A2A receptor, a G-protein coupled member of the P1 family of adenosine receptors, has different functions crucial for neurodegeneration. Its activation in the hippocampal region regulates synaptic plasticity and in particular glutamate release, NMDA receptor activation and calcium influx. Additionally, it exerts effects in neuroinflammation, regulating the secretion of pro-inflammatory cytokines. In AD patients, its expression is increased in the hippocampus/entorhinal cortex more than in the frontal cortex, a phenomenon not observed in age-matched control brains, indicating an association with AD pathology. It is upregulated in peripheral blood cells of patients affected by AD, thus reflecting its increase at central neuronal level. This review offers an overview on the main AD biomarkers and the potential role of A2A adenosine receptor as a new marker and therapeutic target.


Subject(s)
Alzheimer Disease/genetics , Receptor, Adenosine A2A/physiology , Adenosine/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Biomarkers/metabolism , Brain/metabolism , Cerebral Cortex/metabolism , Entorhinal Cortex/metabolism , Glutamic Acid/metabolism , Hippocampus/metabolism , Humans , Neuronal Plasticity/physiology , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Receptors, N-Methyl-D-Aspartate
4.
Brain Res ; 1768: 147590, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34310936

ABSTRACT

Depression, rapid eye movement (REM) sleep behavior disorder, and altered olfaction are often present in Parkinson's disease. Our previous studies demonstrated the role of the olfactory bulb (OB) in causing REM sleep disturbances in depression. Furthermore, adenosine A2A receptors (A2AR) which are richly expressed in the OB, play an important role in the regulation of REM sleep. Caffeine, an adenosine A1 receptors and A2AR antagonist, and other A2AR antagonists were reported to improve olfactory function and restore age-related olfactory deficits. Therefore, we hypothesized that the A2AR neurons in the OB may regulate olfaction or odor-guided behaviors in mice. In the present study, we employed chemogenetics to specifically activate or inhibit neuronal activity. Then, buried food test and olfactory habituation/dishabituation test were performed to measure the changes in the mice's olfactory ability. We demonstrated that activation of OB neurons or OB A2AR neurons shortened the latency of buried food test and enhanced olfactory habituation to the same odors and dishabituation to different odors; inhibition of these neurons showed the opposite effects. Photostimulation of ChR2-expressing OB A2AR neuron terminals evoked inward current in the olfactory tubercle (OT) and the piriform cortex (Pir), which was blocked by glutamate receptor antagonists 2-amino-5-phosphonopentanoic acid and 6-cyano-7nitroquinoxaline-2,3-dione. Collectively, these results suggest that the OB mediates olfaction via A2AR neurons in mice. Moreover, the excitatory glutamatergic release from OB neurons to the OT and the Pir were found responsible for the olfaction-mediated effects of OB A2AR neurons.


Subject(s)
Receptor, Adenosine A2A/metabolism , Smell/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Odorants , Olfactory Bulb/metabolism , Olfactory Cortex/metabolism , Olfactory Perception/physiology , Piriform Cortex/metabolism , Receptor, Adenosine A2A/physiology
5.
ACS Chem Neurosci ; 11(20): 3377-3385, 2020 10 21.
Article in English | MEDLINE | ID: mdl-32976713

ABSTRACT

Adenosine is a neuromodulator, and rapid increases in adenosine in the brain occur spontaneously or after mechanical stimulation. However, the regulation of rapid adenosine by adenosine receptors is unclear, and understanding it would allow better manipulation of neuromodulation. The two main adenosine receptors in the brain are A1 receptors, which are inhibitory, and A2A receptors, which are excitatory. Here, we investigated the regulation of spontaneous adenosine and mechanically stimulated adenosine by adenosine receptors, using global A1 or A2A knockout mice. Results were compared in vivo and in brain slices' models. A1 KO mice have increased frequency of spontaneous adenosine events, but no change in the average concentration of an event, while A2A KO mice had no change in frequency but increased average event concentration. Thus, both A1 and A2A self-regulate spontaneous adenosine release; however, A1 acts on the frequency of events, while A2A receptors regulate concentration. The trends are similar both in vivo and slices, so brain slices are a good model system to study spontaneous adenosine release. For mechanically stimulated adenosine, there was no effect of A1 or A2A KO in vivo, but in brain slices, there was a significant increase in concentration evoked in A1KO mice. Mechanically stimulated release was largely unregulated by A1 and A2A receptors, likely because of a different release mechanism than spontaneous adenosine. Thus, A1 receptors affect the frequency of spontaneous adenosine transients, and A2A receptors affect the concentration. Therefore, future studies could probe drug treatments targeting A1 and A2A receptors to increase rapid adenosine neuromodulation.


Subject(s)
Adenosine , Caudate Nucleus/physiology , Receptor, Adenosine A1/physiology , Receptor, Adenosine A2A/physiology , Animals , Mice
6.
Med Hypotheses ; 144: 110012, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32590324

ABSTRACT

The most serious health issue today is the rapid outbreak of Coronavirus Disease 2019 (COVID-19). More than 6,973,427 confirmed cases were diagnosed in nearly 213 countries and territories around the world and two international conveyances, causing globally over 400,000 deaths. Epidemiology, risk factors, and clinical characteristics of COVID-19 patients have been identified, but the factors influencing the immune system against COVID-19 have not been well established. Upon infection or cell damage, high amounts of adenosine triphosphate (ATP) are released from damaged cells, which serve as mediators of inflammation through purinergic cell surface receptor signaling. As a protective mechanism to prevent excessive damage to host tissue, adenosine counteracts ATP's effects by adenosine receptor stimulation to suppress the pro-inflammatory response. Adenosine is seen as a major obstacle to the efficacy of immune therapies, and the adenosinergic axis components are critical therapeutic targets for cancer and microbial infections. Pharmacologic inhibitors or antibodies specific to adenosinergic pathway components or adenosine receptors in microbial and tumor therapy have shown efficacy in pre-clinical studies and are entering the clinical arena. In this review, we provide a novel hypothesis explaining the potential for improving the efficiency of innate and adaptive immune systems by targeting adenosinergic pathway components and adenosine A2A receptor signaling for the treatment of COVID-19.


Subject(s)
Adenosine A2 Receptor Antagonists/therapeutic use , COVID-19 Drug Treatment , Pandemics , Receptor, Adenosine A2A/physiology , 5'-Nucleotidase/metabolism , Adaptive Immunity/drug effects , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine Triphosphate/metabolism , Apyrase/metabolism , COVID-19/epidemiology , COVID-19/immunology , COVID-19/metabolism , GPI-Linked Proteins/metabolism , Humans , Immunity, Innate/drug effects , Interferon-beta/physiology , Models, Immunological , Molecular Targeted Therapy , Severe acute respiratory syndrome-related coronavirus/immunology , SARS-CoV-2/immunology , Signal Transduction/drug effects , Virus Replication/drug effects
7.
J Diabetes Res ; 2020: 7856503, 2020.
Article in English | MEDLINE | ID: mdl-32566683

ABSTRACT

Peripheral diabetic neuropathy (DPN) is a complication observed in up to half of all patients with type 2 diabetes. DPN has also been shown to be associated with obesity. High-fat diet (HFD) affects glucose metabolism, and the impaired glucose tolerance can lead to type 2 diabetes. There is evidence to suggest a role of adenosine 2A receptors (A2ARs) and semaphorin 3A (Sema3a) signaling in DPN. The link between the expression of Sema3a and A2AR in DPN was hypothesized, but the underlying mechanisms remain poorly understood. In this study, we investigated the regulation of Sema3a by A2AR in the spinal cord and the functional implications thereof in DPN. We examined the expression of A2ARs and Sema3a, as well as Neuropilin 1 and Plexin A, the coreceptors of Sema3a, in the dorsal horn of the lumbar spinal cord of an animal model with HFD-induced diabetes. Our results demonstrate that HFD dysregulates the A2AR-mediated Sema3a expression, with functional implications for the type 2 diabetes-induced peripheral neuropathy. These observations could stimulate clinical studies to improve our understanding on the subject.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Neuropathies , Receptor, Adenosine A2A/physiology , Animals , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetic Neuropathies/etiology , Diabetic Neuropathies/metabolism , Diabetic Neuropathies/pathology , Diet, High-Fat , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Male , Mice , Mice, Inbred C57BL , Nerve Fibers/metabolism , Nerve Fibers/pathology , Semaphorin-3A/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology
8.
Biol Pharm Bull ; 43(8): 1159-1171, 2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32448843

ABSTRACT

Regulating synaptic formation and transmission is critical for the physiology and pathology of psychiatric disorders. The adenosine A2A receptor subtype has attracted widespread attention as a key regulator of neuropsychiatric activity, neuroprotection and injury. In this study, we systematically investigated the regulatory effects of a novel A2A receptor agonist, PSB-0777, on the expression of synaptic proteins and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid glutamate receptors (AMPA receptors) at the cellular level in a time- and dose-dependent manner. After 30 min of high-dose PSB-0777 stimulation, the expression of Synapsin-1 (Syn-1), postsynaptic density protein 95 (PSD95), and AMPA receptors and the number of synapses were rapidly and significantly increased in rat primary cortical neurons compared with the control. Sustained elevation was found in the low and medium-dose groups after 24 h and 3 d of treatment. In contrast, after stimulation with PSB-0777 for 3 consecutive days, the expression of Syn-1 was decreased, and PSD95, AMPA receptors and the number of synapses were no longer increased in the high-dose group. Our study focuses on the detailed and systematic regulation of synaptic proteins and AMPA receptors by an A2A receptor agonist, PSB-0777, which may result in both beneficial and detrimental effects on neurotransmission and neuroprotection and may contribute to the pathophysiology of psychiatric disorders related to A2A receptors. These experimental data may contribute to understanding of the mechanisms for neuroprotective and therapeutic effect of A2A receptor agonists.


Subject(s)
Cerebral Cortex/drug effects , Furans/pharmacology , Neuroprotective Agents/pharmacology , Receptor, Adenosine A2A/physiology , Receptors, AMPA/genetics , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Disks Large Homolog 4 Protein/genetics , Dose-Response Relationship, Drug , Female , Rats , Rats, Sprague-Dawley , Synapses/drug effects , Synapses/physiology , Synapsins/genetics , Synaptic Transmission/drug effects
9.
Cells ; 9(5)2020 04 27.
Article in English | MEDLINE | ID: mdl-32349279

ABSTRACT

In the 1980s and 1990s, the concept was introduced that molecular integration in the Central Nervous System could develop through allosteric receptor-receptor interactions in heteroreceptor complexes presents in neurons. A number of adenosine-dopamine heteroreceptor complexes were identified that lead to the A2A-D2 heteromer hypothesis of schizophrenia. The hypothesis is based on strong antagonistic A2A-D2 receptor-receptor interactions and their presence in the ventral striato-pallidal GABA anti-reward neurons leading to reduction of positive symptoms. Other types of adenosine A2A heteroreceptor complexes are also discussed in relation to this disease, such as A2A-D3 and A2A-D4 heteroreceptor complexes as well as higher order A2A-D2-mGluR5 and A2A-D2-Sigma1R heteroreceptor complexes. The A2A receptor protomer can likely modulate the function of the D4 receptors of relevance for understanding cognitive dysfunction in schizophrenia. A2A-D2-mGluR5 complex is of interest since upon A2A/mGluR5 coactivation they appear to synergize in producing strong inhibition of the D2 receptor protomer. For understanding the future of the schizophrenia treatment, the vulnerability of the current A2A-D2like receptor complexes will be tested in animal models of schizophrenia. A2A-D2-Simag1R complexes hold the highest promise through Sigma1R enhancement of inhibition of D2R function. In line with this work, Lara proposed a highly relevant role of adenosine for neurobiology of schizophrenia.


Subject(s)
Receptor, Adenosine A2A/metabolism , Receptors, Dopamine D2/metabolism , Schizophrenia/metabolism , Adenosine/metabolism , Brain/metabolism , Central Nervous System/metabolism , Dopamine/metabolism , Humans , Neurons/metabolism , Receptor, Adenosine A2A/physiology , Receptors, Dopamine D2/physiology , Schizophrenia/physiopathology
10.
Prog Mol Biol Transl Sci ; 169: 247-277, 2020.
Article in English | MEDLINE | ID: mdl-31952688

ABSTRACT

The discovery of receptor-receptor interactions in the early 1980s, together with a more accurate focusing of allosteric mechanisms in proteins, expanded the knowledge on the G protein-coupled receptor (GPCR)-mediated signaling processes. GPCRs were seen to operate not only as monomers, but also as quaternary structures shaped by allosteric interactions. These integrative mechanisms can change the function of the GPCRs involved, leading to a sophisticated dynamic of the receptor assembly in terms of modulation of recognition and signaling. In this context, the heterodimeric complex formed by the adenosine A2A and the dopamine D2 receptors likely represents a prototypical example. The pharmacological evidence obtained, together with the tissue distribution of the A2A-D2 heteromeric complexes, suggested they could represent a target for new therapeutic strategies addressing significant disorders of the central nervous system. The research findings and the perspectives they offer from the therapeutic standpoint are the focus of the here presented discussion.


Subject(s)
Astrocytes/physiology , Neurons/physiology , Receptor, Adenosine A2A/physiology , Receptors, Dopamine D2/physiology , Adenosine/metabolism , Allosteric Site , Animals , Central Nervous System/metabolism , Computational Biology , Dopamine/metabolism , Humans , Mice , Parkinson Disease/metabolism , Protein Binding , Protein Conformation , Protein Multimerization , Schizophrenia/metabolism , Signal Transduction
11.
J Pharmacol Sci ; 141(1): 64-69, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31640919

ABSTRACT

Activation of both adenosine A2A and A2B receptors (A2BR) contributes to coronary vasodilation. We previously demonstrated that uridine adenosine tetraphosphate (Up4A) is a novel vasodilator in the porcine coronary microcirculation, acting mainly on A2AR in smooth muscle cells (SMC). We further investigated whether activation of A2BR is involved in Up4A-mediated coronary SMC relaxation. Both A2AR and A2BR may stimulate H2O2 production leading to activation of KATP channels in SMCs, we also studied the involvement of H2O2 and KATP channels in Up4A-mediated effect. Coronary small arteries dissected from the apex of porcine hearts were mounted on wire myograph for Up4A concentration responses. Up4A-induced coronary SMC relaxation was attenuated by A2AR but not A2BR antagonism or non-selective P2R antagonism, despite greater endogenous A2BR expression vs. A2AR in both coronary small arteries and primary cultured coronary SMCs. Moreover, Up4A-induced coronary SMC relaxation was blunted by H2O2 catabolism. This effect was not altered by KATP channel blockade. Combination of H2O2 catabolism and A2AR antagonism attenuated Up4A-induced coronary SMC relaxation to the similar extent as A2AR antagonism alone. Collectively, Up4A-induced porcine coronary SMC relaxation is mediated by activation of A2AR-H2O2 pathway. This process does not involve A2BR, P2R or KATP channels.


Subject(s)
Coronary Vessels/drug effects , Dinucleoside Phosphates/pharmacology , Muscle Relaxation/drug effects , Muscle, Smooth, Vascular/drug effects , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A2A/physiology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Cells, Cultured , Coronary Circulation/drug effects , Hydrogen Peroxide/metabolism , Microcirculation/drug effects , Receptor, Adenosine A2B , Swine
12.
Mol Nutr Food Res ; 63(23): e1900662, 2019 12.
Article in English | MEDLINE | ID: mdl-31512815

ABSTRACT

SCOPE: Platelet integrin αIIbß3 is the key mediator of atherothrombosis. Supplementation of coenzyme Q10 (CoQ10), a fat-soluble molecule that exists in various foods, exerts protective cardiovascular effects. This study aims to investigate whether and how CoQ10 acts on αIIbß3 signaling and thrombosis, the major cause of cardiovascular diseases. METHODS AND RESULTS: Using a series of platelet functional assays in vitro, it is demonstrated that CoQ10 reduces human platelet aggregation, granule secretion, platelet spreading, and clot retraction. It is further demonstrated that CoQ10 inhibits platelet integrin αIIbß3 outside-in signaling. These inhibitory effects are mainly mediated by upregulating cAMP/PKA pathway, where CoQ10 stimulates the A2A adenosine receptor and decreases phosphodiesterase 3A phosphorylation. Moreover, CoQ10 attenuates murine thrombus growth and vessel occlusion in a ferric chloride (FeCl3 )-induced thrombosis model in vivo. Importantly, the randomized, double-blind, placebo-controlled clinical trial in dyslipidemic patients demonstrates that 24 weeks of CoQ10 supplementation increases platelet CoQ10 concentrations, enhances the cAMP/PKA pathway, and attenuates αIIbß3 outside-in signaling, leading to decreased platelet aggregation and granule release. CONCLUSION: Through upregulating the platelet cAMP/PKA pathway, and attenuating αIIbß3 signaling and thrombus growth, CoQ10 supplementation may play an important protective role in patients with risks of cardiovascular diseases.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Cyclic AMP/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/physiology , Thrombosis/prevention & control , Ubiquinone/analogs & derivatives , Adult , Animals , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Double-Blind Method , Humans , Male , Mice , Mice, Inbred C57BL , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Receptor, Adenosine A2A/physiology , Signal Transduction/physiology , Ubiquinone/pharmacology , Up-Regulation
13.
Curr Pharm Des ; 25(26): 2859-2874, 2019.
Article in English | MEDLINE | ID: mdl-31291875

ABSTRACT

Adenosine is a purine nucleoside which is an effective controller of inflammation. The inflammatory effect of adenosine is expressed via its four receptor subtypes viz. A1, A2A, A2B and A3. The various inflammatory conditions including rheumatoid arthritis (RA) are initiated by adenosine receptors of which A2A and A3 play a vital role. RA primarily is an auto-immune disorder which is manifested as chronic inflammation in the synovial lining of joints. In order to develop an effective treatment, the role of cytokines, IL-1, TNF-α and IL-6 is crucial. Besides, the knowledge of PI3K-PKB/Akt and NF-kB signaling pathway is also important to understand the antiinflammatory targets. Methotrexate along with various other molecules like, NSAIDs and DMARDs are presently used as treatment lines for controlling RA. The enhanced knowledge of the preclinical stages and pathogenesis along with recent potent therapeutics raises the hopes that RA can be prevented in the near future.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Receptor, Adenosine A2A/physiology , Receptor, Adenosine A3/physiology , Adenosine , Adenosine A2 Receptor Antagonists , Adenosine A3 Receptor Antagonists , Humans , Methotrexate/therapeutic use , Signal Transduction
14.
Biochem Pharmacol ; 166: 313-321, 2019 08.
Article in English | MEDLINE | ID: mdl-31199895

ABSTRACT

Caffeine is the most consumed psychoactive drug worldwide and its intake in moderate amounts prevents neurodegenerative disorders. However, the molecular targets of caffeine to modulate activity in brain circuits are ill-defined. By electrophysiologically recording synaptic transmission and plasticity in Schaffer fibers-CA1 pyramid synapses of mouse hippocampal slices, we characterized the impact of caffeine using a concentration reached in the brain parenchyma upon moderate caffeine consumption. Caffeine (50 µM) facilitated synaptic transmission by 40%, while decreasing paired-pulse facilitation, and also decreased by 35% the amplitude of long-term potentiation (LTP). Clearance of extracellular adenosine with adenosine deaminase (2 U/mL) blunted all the effects of caffeine on synaptic transmission and plasticity. The A1R antagonist DPCPX (100 nM) only eliminated caffeine-induced facilitation of synaptic transmission while not affecting caffeine-induced depression of LTP; conversely, the genetic (using A2AR knockout mice) or the pharmacological blockade (with SCH58261, 50 nM) of A2AR eliminated the effect of caffeine on LTP while not affecting caffeine-induced facilitation of synaptic transmission. Finally, blockade of GABAA or of ryanodine receptors with bicuculline (10 µM) or dantrolene (10 µM), respectively, did not affect the ability of caffeine to alter synaptic transmission or plasticity. These results show that the effects of caffeine on synaptic transmission and plasticity in the hippocampus are selectively mediated by antagonizing adenosine receptors, where A1R are responsible for the impact of caffeine on synaptic transmission and A2AR regulate the impact of caffeine on LTP.


Subject(s)
Caffeine/pharmacology , Hippocampus/drug effects , Neuronal Plasticity/drug effects , Receptor, Adenosine A1 , Receptor, Adenosine A2A , Synaptic Transmission/drug effects , Adenosine A1 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Central Nervous System Stimulants/pharmacology , Hippocampus/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/physiology , Receptor, Adenosine A1/physiology , Receptor, Adenosine A2A/physiology , Synaptic Transmission/physiology
15.
Adv Exp Med Biol ; 1136: 113-121, 2019.
Article in English | MEDLINE | ID: mdl-31201720

ABSTRACT

Long-term studies of anti-pathogen and anti-tumor immunity have provided complementary genetic and pharmacological evidence for the immunosuppressive and immunomodulatory effects of Hypoxia-HIF-1α and adenosine-mediated suppression via the A2A adenosine receptor signaling pathway (Hypoxia-A2A-adenosinergic). This pathway is life saving when it protects inflamed tissues of vital organs from collateral damage by overactive anti-pathogen immune cells or enables the differentiation of cells of adaptive immunity. However, the Hypoxia-A2A-adenosinergic immunosuppression can also prevent tumor rejection by inhibiting the anti-tumor effects of T and NK cells. In addition, this suppressive pathway has been shown to mask tumors due to the hypoxia-HIF-α-mediated loss of MHC Class I molecules on tumor cells. It is suggested that it will be impossible to realize the full anti-tumor capacities of current cancer immunotherapies without simultaneous administration of anti-Hypoxia-A2A-Adenosinergic drugs that inactivate this tumor-protecting mechanism in hypoxic and adenosine-rich tumors.Here, we overview the supporting evidence for the conceptually novel immunotherapeutic motivation to breathe supplemental oxygen (40-60%) or to repurpose already available oxygenation agents in combination with current immunotherapies. Preclinical studies provide strong support for oxygen immunotherapy to enable much stronger tumor regression by weakening immunosuppression by A2A adenosine receptors and by the Hypoxia➔HIF-1α axis. The results of these studies emphasize the value of systemic oxygenation as clinically feasible, promising, and as a valuable tool for mechanistic investigations of tumor biology and cancer immunology. Perhaps the most effective and feasible among individual members of this novel class of anti-tumor drugs are oxygenation agents.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Immunosuppression Therapy , Neoplasms/pathology , Tumor Hypoxia , Cell Hypoxia , Cell Line, Tumor , Humans , Immune Tolerance , Receptor, Adenosine A2A/physiology , Signal Transduction
16.
Article in English | MEDLINE | ID: mdl-30946941

ABSTRACT

The prevalence of anxiety disorders in patients with Attention Deficit/Hyperactivity Disorder (ADHD) is around 15-40%, three times higher than in the general population. The dopaminergic system, classically associated with ADHD, interacts directly with the adenosinergic system through adenosine A2A receptors (A2A) and dopamine D2 receptors (D2) forming A2A-D2 heterodimers. Both dopaminergic and adenosinergic systems are implicated in anxiety disorders. Therefore, the aims of this study were: a) to investigate the main effects of ADORA2A and DRD2 gene variants on anxiety disorders in an ADHD sample of children and adolescents; b) to test potential synergism between ADORA2A and DRD2 genes on the same outcome; c) to explore ADORA2A variants functionality using an in silico approach. The sample consists of 478 children and adolescents with ADHD and their parents, totalizing 1.239 individuals. An association between the ADORA2A rs2298383 TT genotype with the presence of anxiety disorders (P = .004) and an interaction between ADORA2A-DRD2 risk haplotypes with the same outcome (P = .005) was detected. The in silico analyses showed that rs2298383 has the highest score for regulatory function among all variants in the ADORA2A gene described up to date. Altogether, the present findings suggested that the ADORA2A gene and the interaction of ADORA2A and DRD2 genes may play a role in anxiety disorders in children and adolescents with ADHD.


Subject(s)
Anxiety Disorders/genetics , Attention Deficit Disorder with Hyperactivity/genetics , Genetic Predisposition to Disease/genetics , Receptor, Adenosine A2A/genetics , Receptors, Dopamine D2/genetics , Adolescent , Anxiety Disorders/etiology , Attention Deficit Disorder with Hyperactivity/complications , Child , Female , Haplotypes , Humans , Male , Polymorphism, Single Nucleotide/genetics , Receptor, Adenosine A2A/physiology , Receptors, Dopamine D2/physiology
17.
Neuropharmacology ; 148: 229-243, 2019 04.
Article in English | MEDLINE | ID: mdl-30659840

ABSTRACT

Adenosine A2a receptors (A2aRs) are highly and selectively expressed in D2-medium spiny neurons (D2-MSNs) that also express a high level of dopamine D2 receptors (D2Rs). However, it was not established how A2aR activity affects D2-MSN excitability, let alone the ion channels involved. We have performed two sets of experiments to determine the potential A2aR agonistic effects on D2-MSN intrinsic excitability and the underlying ion channel mechanism. First, we have used the cAMP-producing, Gαs/olf coupled designer receptors exclusively activated by designer drug (Gs-DREADDs) to phenocopy cAMP-stimulating A2aR activation. We found that activation of Gs-DREADD inhibited the inwardly rectifying potassium current (Kir)-a key regulator of MSN excitability, caused a depolarization, increased input resistance, and substantially increased the intrinsic excitability of MSNs such that depolarizing inputs evoked many more action potentials. Second, we have determined that A2aR agonism produced these same excitatory effects on D2-MSN intrinsic excitability and spike firing, although at lower magnitudes than those induced by Gs-DREADD activation; furthermore, these A2aR-triggered excitatory effects were intact in the presence of a D2R antagonist. Taken together, these results clearly establish that in striatal D2-MSNs, A2aR activation can independently inhibit Kir and increase intrinsic excitability and spike and neurotransmitter output; our results also indicate that Gs-DREADD can serve as a broadly useful positive control for neurotransmitter receptors that increase intracellular cAMP levels and hence facilitate the determination of the cellular effects of these neurotransmitter receptors.


Subject(s)
Corpus Striatum/physiology , Potassium Channels, Inwardly Rectifying/physiology , Receptor, Adenosine A2A/physiology , Receptors, Dopamine D2/physiology , Action Potentials/drug effects , Adenosine A2 Receptor Agonists/pharmacology , Animals , Clozapine/analogs & derivatives , Clozapine/pharmacology , Dopamine D2 Receptor Antagonists/pharmacology , Electric Stimulation , Mice , Mice, Transgenic , Neurons/physiology , Receptor, Adenosine A2A/drug effects , Receptor, Adenosine A2A/genetics
18.
Pharmacology ; 103(1-2): 38-49, 2019.
Article in English | MEDLINE | ID: mdl-30380560

ABSTRACT

BACKGROUND/AIMS: In this study, we evaluated the functional impact of facilitatory presynaptic adenosine A2A and muscarinic M1 receptors in the recovery of neuromuscular tetanic depression caused by the blockage of high-affinity choline transporter (HChT) by hemicholinium-3 (HC-3), a condition that mimics a myasthenia-like condition. METHODS: Rat diaphragm preparations were indirectly stimulated via the phrenic nerve trunk with 50-Hz frequency trains, each consisting of 500-750 supramaximal intensity pulses. The tension at the beginning (A) and at the end (B) of the tetanus was recorded and the ratio (R) B/A calculated. RESULTS: Activation of A2A and M1 receptors with CGS21680 (CGS; 2 nmol/L) and McN-A-343c (McN; 3 µmol/L) increased R values. Similar facilitatory effects were obtained with forskolin (FSK; 3 µmol/L) and phorbol 12-myristate 13-acetate (PMA; 10 µmol/L), which activate adenylate cyclase and protein kinase C respectively. HC-3 (4 µmol/L) decreased transmitter exocytosis measured by real-time videomicroscopy with the FM4-64 fluorescent dye and prevented the facilitation of neuromuscular transmission caused by CGS, McN, and FSK, with a minor effect on PMA. The acetylcholinesterase inhibitor, neostigmine (NEO; 0.5 µmol/L), also decreased transmitter exocytosis. The paradoxical neuromuscular tetanic fade caused by NEO (0.5 µmol/L) was also prevented by HC-3 (4 µmol/L) and might result from the rundown of the positive feedback mechanism operated by neuronal nicotinic receptors (blocked by hexamethonium, 120 µmol/L). CONCLUSION: Data suggest that the recovery of tetanic neuromuscular facilitation by adenosine A2A and M1 receptors is highly dependent on HChT activity and may be weakened in myasthenic patients when HChT is inoperative.


Subject(s)
Membrane Transport Proteins/physiology , Receptor, Adenosine A2A/physiology , Receptor, Muscarinic M1/physiology , Refractory Period, Electrophysiological/drug effects , (4-(m-Chlorophenylcarbamoyloxy)-2-butynyl)trimethylammonium Chloride/pharmacology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Colforsin/pharmacology , Diaphragm/drug effects , Diaphragm/physiology , Hemicholinium 3/pharmacology , Neostigmine/pharmacology , Phenethylamines/pharmacology , Phrenic Nerve/drug effects , Phrenic Nerve/physiology , Rats , Rats, Wistar , Synaptic Transmission , Tetanus/drug therapy , Tetanus/physiopathology , Tetradecanoylphorbol Acetate/pharmacology
19.
Psychopharmacology (Berl) ; 235(12): 3509-3523, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30426181

ABSTRACT

RATIONALE AND OBJECTIVES: Many studies indicated that adenosine via its A2A receptors influences the behavioral effects of cocaine by modulating dopamine neurotransmission. The hypothesis was tested that A2A receptors in the nucleus accumbens (NAc) or the prefrontral cortex (PFc) may modulate cocaine reward and/or cocaine seeking behavior in rats. METHODS: The effects of local bilateral microinjections of the selective A2A receptor agonist CGS 21680 or the A2A receptor antagonists KW 6002 and SCH 58261 were investigated on cocaine self-administration on reinstatement of cocaine seeking. RESULTS: The intra-NAc shell, but not intra-infralimbic PFc, administration of CGS 21680 significantly reduced the number of active lever presses and the number of cocaine (0.25 mg/kg) infusions. However, tonic activation of A2A receptors located in the NAc or PFc did not play a role in modulating the rewarding actions of cocaine since neither KW 6002 nor SCH 58261 microinjections altered the cocaine (0.5 mg/kg) infusions. The intra-NAc but not intra-PFc microinjections of CGS 21680 dose- dependently attenuated the reinstatement of active lever presses induced by cocaine (10 mg/kg, i.p.) and the drug-associated combined conditioned stimuli using the subthreshold dose of cocaine (2.5 mg/kg, i.p.). On the other hand, the intra-NAc pretreatment with SCH 58261, but not with KW 6002, given alone evoked reinstatement of cocaine seeking behavior. CONCLUSION: The results strongly support the involvement of accumbal shell A2A receptors as a target, the activation of which exerts an inhibitory control over cocaine reward and cocaine seeking.


Subject(s)
Adenosine A2 Receptor Agonists/administration & dosage , Cocaine/administration & dosage , Drug-Seeking Behavior/physiology , Nucleus Accumbens/physiology , Prefrontal Cortex/physiology , Receptor, Adenosine A2A/physiology , Animals , Cocaine-Related Disorders/drug therapy , Conditioning, Operant/drug effects , Dopamine Uptake Inhibitors/administration & dosage , Drug-Seeking Behavior/drug effects , Ligands , Male , Microinjections/methods , Nucleus Accumbens/drug effects , Prefrontal Cortex/drug effects , Rats , Rats, Wistar , Reward , Self Administration
20.
Adv Exp Med Biol ; 1071: 89-93, 2018.
Article in English | MEDLINE | ID: mdl-30357738

ABSTRACT

Adenosine is one of the key neurotransmitters involved in hypoxic signaling in the carotid body (CB), and it was recently found to have a modulatory role in mediating hypercapnic sensitivity in the CB. Herein we have investigated the contribution of adenosine to the hypercapnic response in the rat CB and studied the adenosine receptors responsible for this effect. Experiments were performed in Wistar rats. Adenosine release in normoxia (21% O2) and in response to hypercapnia (10% CO2) was quantified by HPLC. Carotid sinus nerve (CSN) chemosensory activity was evaluated in response to hypercapnia in the absence and presence of ZM241385 (300 nM), an A2 antagonist, and SCH58261 (20 nM), a selective A2A antagonist. Hypercapnia increased the extracellular concentrations of adenosine by 50.01%. Both, ZM241385 and SCH58261, did not modify significantly the basal frequency of discharges of the CSN. Also, ZM241385 and SCH58261 did not modify the latency time and the time to peak in CSN chemosensory activity. CSN activity evoked by hypercapnia decreased by 58.82 and 33.59% in response to ZM241385 and to SCH58261, respectively. In conclusion, the effect of adenosine in mediating the hypercapnic response in the rat CB involves an effect on A2A and A2B adenosine receptors.


Subject(s)
Adenosine/physiology , Carotid Body/physiology , Hypercapnia/physiopathology , Hypoxia , Animals , Pyrimidines/pharmacology , Rats , Rats, Wistar , Receptor, Adenosine A2A/physiology , Receptor, Adenosine A2B/physiology , Triazines/pharmacology , Triazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...