2-(4-(2-carboxyethyl)phenethylamino)-5--n-ethylcarboxamidoadenosine has been researched along with Disease-Models--Animal* in 75 studies
2 trial(s) available for 2-(4-(2-carboxyethyl)phenethylamino)-5--n-ethylcarboxamidoadenosine and Disease-Models--Animal
Article | Year |
---|---|
Plasma glutamate-modulated interaction of A2AR and mGluR5 on BMDCs aggravates traumatic brain injury-induced acute lung injury.
The bone marrow-derived cell (BMDC)-associated inflammatory response plays a key role in the development of acute lung injury (ALI). Activation of adenosine A2A receptor (A2AR) is generally considered to be antiinflammatory, inhibiting BMDC activities to protect against ALI. However, in the present study, we found that in a mouse model of neurogenic ALI induced by severe traumatic brain injury (TBI), BMDC A2AR exerted a proinflammatory effect, aggravating lung damage. This is in contrast to the antiinflammatory effect observed in the mouse oleic acid-induced ALI model (a nonneurogenic ALI model.) Moreover, the A2AR agonist CGS21680 aggravated, whereas the antagonist ZM241385 attenuated, the severe TBI-induced lung inflammatory damage in mice. Further investigation of white blood cells isolated from patients or mouse TBI models and of cultured human or mouse neutrophils demonstrated that elevated plasma glutamate after severe TBI induced interaction between A2AR and the metabotropic glutamate receptor 5 (mGluR5) to increase phospholipase C-protein kinase C signaling, which mediated the proinflammatory effect of A2AR. These results are in striking contrast to the well-known antiinflammatory and protective role of A2AR in nonneurogenic ALI and indicate different therapeutic strategies should be used for nonneurogenic and neurogenic ALI treatment when targeting A2AR. Topics: Acute Lung Injury; Adenosine; Adenosine A2 Receptor Agonists; Adult; Animals; Bone Marrow Cells; Brain Injuries; Disease Models, Animal; Female; Glutamic Acid; Humans; Male; Mice; Mice, Knockout; Middle Aged; Phenethylamines; Protein Kinase C; Receptor, Adenosine A2A; Receptor, Metabotropic Glutamate 5; Receptors, Metabotropic Glutamate; Signal Transduction; Triazines; Triazoles; Type C Phospholipases | 2013 |
Adenosine modulation of neurotransmission in penile erection.
1. Adenosine inhibited the noradrenaline-induced contraction of rabbit corpus cavernosum in a dose-dependent manner. The effect of adenosine was greater in intact corpus cavernosa than in endothelium-denuded preparations. This finding indicates that the relaxing effect of adenosine is partially endothelium-dependent and involved in the release of endothelium-derived relaxing factors. 2. Adenosine and its analogues relaxed the noradrenaline-induced contractile response as well as inhibited the transmural nerve induced contraction with the potency order: NECA > R-PIA > adenosine. These data indicate that adenosine can modulate both the non-adrenergic non-cholinergic and adrenergic neurotransmission. DMPX, an adenosine antagonist selective for the A2 receptors, abolished the electrically elicited relaxation. However, CGS 21680, selective for A2a receptor, had no effect on relaxation. Therefore, adenosine receptors involved in the modulation of neurotransmission in rabbit corpus cavernosum appear to be A2b subtype. 3. Adenosine also induced an increase in human cavernosal arterial velocity and resistive index measured by colour duplex sonography. The combination of adenosine and 10 micrograms prostaglandin E1 was more effective in resistive index and erection grade than 20 micrograms prostaglandin E1 alone. Our results suggest that adenosine seems to be an important neuromodulator for penile erection and can be an effective and alternative combination in the treatment of impotence. Topics: Adenosine; Adenosine-5'-(N-ethylcarboxamide); Adult; Aged; Alprostadil; Animals; Antihypertensive Agents; Disease Models, Animal; Dose-Response Relationship, Drug; Erectile Dysfunction; Humans; Male; Middle Aged; Muscle Contraction; Muscle, Smooth; Nitric Oxide; Norepinephrine; Penile Erection; Penis; Phenethylamines; Phenylisopropyladenosine; Purinergic P1 Receptor Agonists; Rabbits; Synaptic Transmission; Theobromine; Vasodilator Agents | 1994 |
73 other study(ies) available for 2-(4-(2-carboxyethyl)phenethylamino)-5--n-ethylcarboxamidoadenosine and Disease-Models--Animal
Article | Year |
---|---|
Novel alendronate-CGS21680 conjugate reduces bone resorption and induces new bone formation in post-menopausal osteoporosis and inflammatory osteolysis mouse models.
Topics: Alendronate; Animals; Bone Resorption; Disease Models, Animal; Female; Humans; Mice; Mice, Inbred C57BL; Osteoclasts; Osteogenesis; Osteolysis; Osteoporosis, Postmenopausal; RANK Ligand | 2022 |
Adenosine A2a receptors modulate TrkB receptor-dependent respiratory plasticity in neonatal rats.
Neuroplasticity is a fundamental property of the respiratory control system, enabling critical adaptations in breathing to meet the challenges, but little is known whether neonates express neuroplasticity similar to adults. We tested the hypothesis that, similar to adults, tyrosine receptor kinase B (TrkB) or adenosine A2a receptor activation in neonates are independently sufficient to elicit respiratory motor facilitation, and that co-induction of TrkB and A2a receptor-dependent plasticity undermines respiratory motor facilitation. TrkB receptor activation with 7,8-dihydroxyflavone (DHF) in neonatal brainstem-spinal cord preparations induced a long-lasting increase in respiratory motor output in 55 % of preparations, whereas adenosine A2a receptor activation with CGS21680 only sporadically induced respiratory motor plasticity. CGS21680 and DHF co-application prevented DHF-dependent respiratory motor facilitation, whereas co-application of MSX-3 (adenosine A2a receptor antagonist) and DHF more rapidly induced respiratory motor plasticity. Collectively, these data suggest that mechanisms underlying respiratory neuroplasticity may be only partially operational in early neonatal life, and that adenosine A2a receptor activation undermines TrkB-induced respiratory plasticity. Topics: Adenosine; Adenosine A2 Receptor Agonists; Adenosine A2 Receptor Antagonists; Animals; Animals, Newborn; Brain Stem; Disease Models, Animal; Flavones; Neuronal Plasticity; Phenethylamines; Rats; Receptor, Adenosine A2A; Receptor, trkB; Respiratory Physiological Phenomena; Spinal Cord | 2021 |
Therapeutic candidates for the Zika virus identified by a high-throughput screen for Zika protease inhibitors.
When Zika virus emerged as a public health emergency there were no drugs or vaccines approved for its prevention or treatment. We used a high-throughput screen for Zika virus protease inhibitors to identify several inhibitors of Zika virus infection. We expressed the NS2B-NS3 Zika virus protease and conducted a biochemical screen for small-molecule inhibitors. A quantitative structure-activity relationship model was employed to virtually screen ∼138,000 compounds, which increased the identification of active compounds, while decreasing screening time and resources. Candidate inhibitors were validated in several viral infection assays. Small molecules with favorable clinical profiles, especially the five-lipoxygenase-activating protein inhibitor, MK-591, inhibited the Zika virus protease and infection in neural stem cells. Members of the tetracycline family of antibiotics were more potent inhibitors of Zika virus infection than the protease, suggesting they may have multiple mechanisms of action. The most potent tetracycline, methacycline, reduced the amount of Zika virus present in the brain and the severity of Zika virus-induced motor deficits in an immunocompetent mouse model. As Food and Drug Administration-approved drugs, the tetracyclines could be quickly translated to the clinic. The compounds identified through our screening paradigm have the potential to be used as prophylactics for patients traveling to endemic regions or for the treatment of the neurological complications of Zika virus infection. Topics: Animals; Antiviral Agents; Artificial Intelligence; Chlorocebus aethiops; Disease Models, Animal; Drug Evaluation, Preclinical; High-Throughput Screening Assays; Immunocompetence; Inhibitory Concentration 50; Methacycline; Mice, Inbred C57BL; Protease Inhibitors; Quantitative Structure-Activity Relationship; Small Molecule Libraries; Vero Cells; Zika Virus; Zika Virus Infection | 2020 |
The Adenosine A2A Receptor Agonist Accelerates Bone Healing and Adjusts Treg/Th17 Cell Balance through Interleukin 6.
The aim of this study was to explore the effect of adenosine A2A receptor agonists on fracture healing and the regulation of the immunity system after bone fracture. We implanted fibrin gel containing adenosine A2A receptor agonist CGS 21680/inhibitor ZM 241385/saline locally in rat tibial fracture models, finding that the adenosine A2A receptor agonist could promote fracture healing. At the same time, the adenosine A2A receptor agonist decreased the level of IL-6 in blood and the fracture area, increased Treg cells, and decreased Th17 cells in blood of bone fracture rats. Further, tibial fracture rats implanted with the adenosine A2A receptor agonist gel were injected with IL-6. We found that IL-6 could reverse the effect of adenosine A2A receptor agonists on fracture healing and Treg/Th17 cells in blood. Through the above results, we believe that the adenosine A2A receptor agonist can promote fracture healing and regulate Treg/Th17 cells in blood of rats with fractures. These effects are related to IL-6. Topics: Adenosine; Adenosine A2 Receptor Agonists; Animals; Disease Models, Animal; Female; Fracture Healing; Interleukin-6; Phenethylamines; Rats; Rats, Sprague-Dawley; Receptor, Adenosine A2A; T-Lymphocytes, Regulatory; Th17 Cells; Tibial Fractures | 2020 |
Pharmacological targeting of striatal indirect pathway neurons improves subthalamic nucleus dysfunction and reduces repetitive behaviors in C58 mice.
Repetitive behaviors (e.g., stereotypic movements, compulsions, rituals) are common features of a number of neurodevelopmental disorders. Clinical and animal model studies point to the importance of cortical-basal ganglia circuitry in the mediation of repetitive behaviors. In the current study, we tested whether a drug cocktail (dopamine D2 receptor antagonist + adenosine A2A receptor agonist + glutamate mGlu5 positive allosteric modulator) designed to activate the indirect basal ganglia pathway would reduce repetitive behavior in C58 mice after both acute and sub-chronic administration. In addition, we hypothesized that sub-chronic administration (i.e. 7 days of twice-daily injections) would increase the functional activation of the subthalamic nucleus (STN), a key node of the indirect pathway. Functional activation of STN was indexed by dendritic spine density, analysis of GABA, glutamate, and synaptic plasticity genes, and cytochrome oxidase activity. The drug cocktail used significantly reduced repetitive motor behavior in C58 mice after one night as well as seven nights of twice-nightly injections. These effects did not reflect generalized motor behavior suppression as non-repetitive motor behaviors such as grooming, digging and eating were not reduced relative to vehicle. Sub-chronic drug treatment targeting striatopallidal neurons resulted in significant changes in the STN, including a four-fold increase in brain-derived neurotrophic factor (BDNF) mRNA expression as well as a significant increase in dendritic spine density. The present findings are consistent with, and extend, our prior work linking decreased functioning of the indirect basal ganglia pathway to expression of repetitive motor behavior in C58 mice and suggest novel therapeutic targets. Topics: Adenosine; Animals; Autism Spectrum Disorder; Basal Ganglia; Behavior, Animal; Benzamides; Compulsive Behavior; Corpus Striatum; Disease Models, Animal; Indoles; Male; Mice; Mice, Inbred Strains; Neural Pathways; Neurons; Phenethylamines; Piperidines; Pyrazoles; Stereotyped Behavior; Subthalamic Nucleus | 2020 |
The adenosine A(2A) receptor agonist CGS 21680 alleviates auditory sensorimotor gating deficits and increases in accumbal CREB in rats neonatally treated with quinpirole.
The adenosine A(2A) receptor forms a mutually inhibitory heteromer with the dopamine D. Male and female Sprague-Dawley rats were neonatally treated with saline (NS) or quinpirole HCl (NQ; 1 mg/kg) from postnatal days (P) 1-21. Animals were raised to P44 and behaviorally tested on auditory sensorimotor gating as measured through prepulse inhibition (PPI) from P44 to P48. Approximately 15 min before each session, animals were given an ip administration of saline or the adenosine A(2A) agonist CGS 21680 (0.03 or 0.09 mg/kg). One day after PPI was complete on P49, animals were administered a locomotor activity test in the open field after saline or CGS 21680 treatment, respectively. On P50, the nucleus accumbens (NAc) was evaluated for CREB protein.. NQ-treated rats demonstrated a deficit in PPI that was alleviated to control levels by either dose of CGS 21680. The 0.03 mg/kg dose of CGS 21680 increased startle amplitude in males. The 0.09 mg/kg dose of CGS 21680 resulted in an overall decrease in locomotor activity. NQ treatment significantly increased NAc CREB that was attenuated to control levels by either dose of CGS 21680.. This study revealed that an adenosine A(2A) receptor agonist was effective to alleviate PPI deficits in the NQ model of SZ in both male and female rats. Topics: Adenosine; Adenosine A2 Receptor Agonists; Animals; Animals, Newborn; Cyclic AMP Response Element-Binding Protein; Disease Models, Animal; Dopamine Agonists; Dose-Response Relationship, Drug; Female; Male; Motor Activity; Nucleus Accumbens; Phenethylamines; Prepulse Inhibition; Quinpirole; Rats; Rats, Sprague-Dawley; Receptor, Adenosine A2A; Receptors, Dopamine D2; Schizophrenia; Sensory Gating | 2020 |
Intraarticular injection of liposomal adenosine reduces cartilage damage in established murine and rat models of osteoarthritis.
Osteoarthritis (OA) affects nearly 10% of the population of the United States and other industrialized countries and, at present, short of surgical joint replacement, there is no therapy available that can reverse the progression of the disease. Adenosine, acting at its A2A receptor (A2AR), is a critical autocrine factor for maintenance of cartilage homeostasis and here we report that injection of liposomal suspensions of either adenosine or a selective A2AR agonist, CGS21680, significantly reduced OA cartilage damage in a murine model of obesity-induced OA. The same treatment also improved swelling and preserved cartilage in the affected knees in a rat model of established post-traumatic OA (PTOA). Differential expression analysis of mRNA from chondrocytes harvested from knees of rats with PTOA treated with liposomal A2AR agonist revealed downregulation of genes associated with matrix degradation and upregulation of genes associated with cell proliferation as compared to liposomes alone. Studies in vitro and in affected joints demonstrated that A2AR ligation increased the nuclear P-SMAD2/3/P-SMAD1/5/8 ratio, a change associated with repression of terminal chondrocyte differentiation. These results strongly suggest that targeting the A2AR is an effective approach to treat OA. Topics: Adenosine; Animals; Cartilage; Cartilage, Articular; Cell Differentiation; Chondrocytes; Disease Models, Animal; Injections, Intra-Articular; Liposomes; Male; Mice; Mice, Inbred C57BL; Osteoarthritis; Phenethylamines; Rats; Rats, Sprague-Dawley; Signal Transduction | 2020 |
Inhibition of natriuretic peptide receptor 1 reduces itch in mice.
There is a major clinical need for new therapies for the treatment of chronic itch. Many of the molecular components involved in itch neurotransmission are known, including the neuropeptide NPPB, a transmitter required for normal itch responses to multiple pruritogens in mice. Here, we investigated the potential for a novel strategy for the treatment of itch that involves the inhibition of the NPPB receptor NPR1 (natriuretic peptide receptor 1). Because there are no available effective human NPR1 (hNPR1) antagonists, we performed a high-throughput cell-based screen and identified 15 small-molecule hNPR1 inhibitors. Using in vitro assays, we demonstrated that these compounds specifically inhibit hNPR1 and murine NPR1 (mNPR1). In vivo, NPR1 antagonism attenuated behavioral responses to both acute itch- and chronic itch-challenged mice. Together, our results suggest that inhibiting NPR1 might be an effective strategy for treating acute and chronic itch. Topics: Animals; Behavior, Animal; Cell-Free System; Dermatitis, Contact; Disease Models, Animal; Ganglia, Spinal; Humans; Mice, Inbred C57BL; Mice, Knockout; Neurons; Pruritus; Receptors, Atrial Natriuretic Factor; Reproducibility of Results; Signal Transduction; Small Molecule Libraries | 2019 |
Adenosine 2a Receptor Signal Blockade of Murine Autoimmune Arthritis via Inhibition of Pathogenic Germinal Center-Follicular Helper T Cells.
CD4 germinal center (GC)-follicular helper T (Tfh) cells are important in the pathogenesis of autoimmune arthritis. Previous studies have shown that adenosine 2a receptor (A2aR; Adora2a) signaling can divert CD4 T cells away from the GC-Tfh cell lineage during the primary response to foreign antigens. This study was undertaken to examine the effects of A2aR signaling on CD4 T cells during the recognition of self antigen in a murine model of autoimmune arthritis.. Wild-type and Adora2a-deficient mouse KRN T cell receptor-transgenic CD4 T cells specific for glucose-6-phosphate isomerase (GPI)/I-A. CGS treatment inhibited the development of arthritis and differentiation of KRN GC-Tfh cells, blocked the appearance of high-affinity GPI-specific and IgG1 isotype class-switched polyclonal plasmablasts, and led to a reduction in serum titers of anti-GPI IgG1. In addition, therapeutic administration of CGS after the onset of arthritis blocked further disease progression in association with reductions in the number of KRN GC-Tfh cells and anti-GPI IgG1 serum titers.. Strong A2aR signaling diverts autoreactive CD4 T cell differentiation away from the GC-Tfh cell lineage, thus reducing help for the differentiation of dangerous autoreactive B cells that promote arthritis. These data in a mouse model of autoimmune arthritis suggest that A2aR and its downstream signaling pathways in CD4 T cells may be promising therapeutic targets for interfering with potentially dangerous autoreactive GC-Tfh cell differentiation. Topics: Adenosine; Adenosine A2 Receptor Agonists; Adoptive Transfer; Animals; Arthritis, Experimental; Autoantigens; Autoimmune Diseases; CD4-Positive T-Lymphocytes; Cytokines; Disease Models, Animal; Germinal Center; Glucose-6-Phosphate Isomerase; Mice; Mice, Knockout; Mice, Transgenic; Phenethylamines; Receptor, Adenosine A2A; Receptors, Antigen, T-Cell, alpha-beta; Signal Transduction; T-Lymphocytes, Helper-Inducer | 2019 |
Time-Dependent Bidirectional Neuroprotection by Adenosine 2A Receptor in Experimental Traumatic Brain Injury.
Traumatic brain injury (TBI) results in both focal and diffuse brain pathological features that become severely exacerbated after the initial injury. Owing to this disease complexity, no effective therapeutic measure has yet been devised aimed directly at these pathological processes. We developed a clinically relevant model of TBI and tested the bidirectional neuroprotective role of adenosine 2A receptors (A2ARs) at different times.. Wistar rats were divided into 4 treatment groups (sham, TBI, A2AR agonist [CGS-21680], and A2AR antagonist [SCH-58261]) and 4 post-TBI intervals (15 minutes and 1, 12, and 24 hours). A2AR agonist and antagonist effects were tested by the neurological functional score (NFS) and levels of cyclic adenosine monophosphate, interleukin-1β, oxidative stress antioxidant markers, and caspase-3.. The A2AR agonist-treated group showed significant NFS improvement at 15 minutes and 1 hour after TBI compared with the TBI group. However, no improvement was observed at 12 and 24 hours. The A2AR antagonists resulted in no NFS improvement at 15 minutes and 1 hour, and significant improvement observed at 12 and 24 hours. Significant neuroprotective effect with an A2AR agonist were observed with cyclic adenosine monophosphate, interleukin-1β, oxidative stress markers, catalase, and caspase-3 levels at 15 minutes and 1 hour after TBI. The A2AR antagonist showed no effect at these intervals but showed a protective effect at 12 and 24 hours after TBI.. The A2AR agonist showed a beneficial neuroprotective effect at the early stages after TBI, and the A2AR antagonist showed a benefit at the later stages after TBI. These findings suggest that A2AR agonists and antagonists should be used in accordance with the point at which the TBI occurred. Topics: Adenosine; Animals; Brain Injuries; Brain Injuries, Traumatic; Disease Models, Animal; Male; Neuroprotection; Neuroprotective Agents; Phenethylamines; Pyrimidines; Rats, Wistar; Receptors, Purinergic P1; Signal Transduction; Time Factors; Triazoles | 2019 |
Modulating P1 Adenosine Receptors in Disease Progression of SOD1
Amyotrophic lateral sclerosis (ALS) is a fatal progressing neurodegenerative disease; to date, despite the intense research effort, only two therapeutic options, with very limited effects, are available. The purinergic system has been indicated as a possible new therapeutic target for ALS, but the results are often contradictory and generally confused. The present study was designed to determine whether P1 adenosine receptor ligands affected disease progression in a transgenic model of ALS. SOD1 Topics: Adenosine; Amyotrophic Lateral Sclerosis; Animals; Disease Models, Animal; Mice, Transgenic; Microglia; Motor Neurons; Phenethylamines; Receptors, Purinergic P1; Spinal Cord; Superoxide Dismutase-1 | 2019 |
Adenosine receptor agonism protects against NETosis and thrombosis in antiphospholipid syndrome.
Potentiation of neutrophil extracellular trap (NET) release is one mechanism by which antiphospholipid antibodies (aPL Abs) effect thrombotic events in patients with antiphospholipid syndrome (APS). Surface adenosine receptors trigger cyclic AMP (cAMP) formation in neutrophils, and this mechanism has been proposed to regulate NETosis in some contexts. Here we report that selective agonism of the adenosine A Topics: Adenosine; Adenosine A2 Receptor Agonists; Animals; Antibodies, Antiphospholipid; Antiphospholipid Syndrome; Cyclic AMP; Dipyridamole; Disease Models, Animal; Extracellular Traps; Fibrinolytic Agents; Gene Expression Regulation; Humans; Immunoglobulin G; Male; Mice; Mice, Inbred C57BL; Neutrophils; Phenethylamines; Receptor, Adenosine A2A; Signal Transduction; Vena Cava, Inferior; Venous Thrombosis | 2019 |
The A
Allogeneic hematopoietic stem cell transplantation (HSCT) is a curative method for blood cancers and other blood disorders, but is limited by the development of graft-versus-host disease (GVHD). GVHD results in inflammatory damage to the host liver, gastrointestinal tract and skin, resulting in high rates of morbidity and mortality in HSCT recipients. Activation of the A Topics: Adenosine; Adenosine A2 Receptor Agonists; Animals; Body Weight; Cytokines; Disease Models, Animal; Female; Graft vs Host Disease; Humans; Mice; Phenethylamines; T-Lymphocytes, Regulatory | 2019 |
Adenosine A2a receptor agonist CGS21680 treatment attenuates cardiopulmonary bypass‑associated inflammatory lung injury in juvenile rats.
The adenosine A2a receptor agonist CGS21680 has been suggested to act as an anti‑inflammatory agent that protects against cardiopulmonary bypass (CPB)‑induced organ injury. However, the therapeutic effects of CGS21680 for CPB‑induced lung injury have not been comprehensively evaluated. Using a juvenile rat model, the present study was designed to evaluated whether CGS21680 attenuates CPB‑induced lung injury. Our juvenile rat CPB model was established by 60 min CPB with or without CGS21680 pretreatment (100 µg/kg, in the CPB priming solution). Rats in the Sham group only underwent cannulation and heparinization. Serum and pulmonary levels of inflammatory markers and histological features of pulmonary tissues were analyzed. All juvenile rats survived following CPB. Significantly elevated serum levels of tumor necrosis factor‑α (TNF‑α), myeloperoxidase (MPO) and interleukin‑1β (IL‑1β), and decreased glutathione peroxidase (GSH‑PX) levels were observed in the CPB group compared to the Sham group (all P<0.05). TNF‑α, MPO and IL‑1β were significantly decreased, while GSH‑PX was markedly increased in the CGS group when compared to the CPB group. Consistently, pulmonary tissues from rats in the CPB group showed considerable amounts of damaged pneumocytes, severe edema, and increased alveolar macrophages, and significantly higher lung injury scores compared to the controls. Collectively, these changes were all further attenuated by CGS21680. Pretreatment with CGS21680 before CPB attenuated pulmonary injury, which may be related to the anti‑inflammatory effects of CGS21680 downstream of A2a receptor activation. Topics: Adenosine; Adenosine A2 Receptor Agonists; Animals; Cardiopulmonary Bypass; Disease Models, Animal; Humans; Inflammation; Interleukin-1beta; Lung; Lung Injury; Peroxidase; Phenethylamines; Rats; Rats, Sprague-Dawley; Receptor, Adenosine A2A; Tumor Necrosis Factor-alpha | 2019 |
Adenosine A
Tau is a microtubule-associated protein, and the oligomeric and hyperphosphorylated forms of tau are increased significantly after neurotrauma and considered important factors in mediating cognitive dysfunction. Blockade of adenosine A Topics: Adenosine; Adenosine A2 Receptor Agonists; Adult; Aged; Animals; Brain Injuries, Traumatic; Cognitive Dysfunction; Disease Models, Animal; Female; Glycogen Synthase Kinase 3 beta; Glycogen Synthase Kinases; Hippocampus; Humans; Indoles; Isoquinolines; Male; Maleimides; Mice; Mice, Knockout; Middle Aged; Neurons; Phenethylamines; Phosphorylation; Protein Kinase Inhibitors; Receptor, Adenosine A2A; Signal Transduction; Sulfonamides; tau Proteins; Triazines; Triazoles | 2017 |
Dual roles of the adenosine A2a receptor in autoimmune neuroinflammation.
Conditions of inflammatory tissue distress are associated with high extracellular levels of adenosine, due to increased adenosine triphosphate (ATP) degradation upon cellular stress or the release of extracellular ATP upon cell death, which can be degraded to adenosine by membrane-bound ecto-enzymes like CD39 and CD73. Adenosine is recognised to mediate anti-inflammatory effects via the adenosine A2a receptor (A2aR), as shown in experimental models of arthritis. Here, using pharmacological interventions and genetic inactivation, we investigated the roles of A2aR in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS).. We used two independent mouse EAE variants, i.e. active immunization in C57BL/6 with myelin oligodendrocyte glycoprotein (MOG)35-55 or transfer-EAE by proteolipid protein (PLP)139-155-stimulated T lymphocytes and EAE in mice treated with A2aR-agonist CGS21680 at different stages of disease course and in mice lacking A2aR (A2aR(-/-)) compared to direct wild-type littermates. In EAE, we analysed myelin-specific proliferation and cytokine synthesis ex vivo, as well as inflammation and demyelination by immunohistochemistry. In vitro, we investigated the effect of A2aR on migration of CD4(+) T cells, macrophages and microglia, as well as the impact of A2aR on phagocytosis of macrophages and microglia. Statistical tests were Mann-Whitney U and Student's t test.. We found an upregulation of A2aR in the central nervous system (CNS) in EAE, predominantly detected on T cells and macrophages/microglia within the inflamed tissue. Preventive EAE treatment with A2aR-specific agonist inhibited myelin-specific T cell proliferation ex vivo and ameliorated disease, while application of the same agonist after disease onset exacerbated non-remitting EAE progression and resulted in more severe tissue destruction. Accordingly, A2aR-deficient mice showed accelerated and exacerbated disease manifestation with increased frequencies of IFN-γ-, IL-17- and GM-CSF-producing CD4(+) T helper cells and higher numbers of inflammatory lesions in the early stage. However, EAE quickly ameliorated and myelin debris accumulation was lower in A2aR(-/-) mice. In vitro, activation of A2aR inhibited phagocytosis of myelin by macrophages and primary microglia as well as migration of CD4(+) T cells, macrophages and primary microglia.. A2aR activation exerts a complex pattern in chronic autoimmune neurodegeneration: while providing anti-inflammatory effects on T cells and thus protection at early stages, A2aR seems to play a detrimental role during later stages of disease and may thus contribute to sustained tissue damage within the inflamed CNS. Topics: Adenosine; Animals; Anti-Inflammatory Agents; CD4-Positive T-Lymphocytes; Cell Movement; Cell Proliferation; Cells, Cultured; Cytokines; Disease Models, Animal; Encephalomyelitis, Autoimmune, Experimental; Macrophages; Mice; Mice, Inbred C57BL; Mice, Transgenic; Microglia; Myelin Proteolipid Protein; Myelin-Oligodendrocyte Glycoprotein; Peptide Fragments; Phagocytosis; Phenethylamines; Receptor, Adenosine A2A; Time Factors; Up-Regulation | 2016 |
Activation of the adenosine A2A receptor exacerbates experimental autoimmune neuritis in Lewis rats in association with enhanced humoral immunity.
Accumulated evidence demonstrated that Adenosine A2A receptor (A2AR) is involved in the inflammatory diseases. In the present study, we showed that a selective A2AR agonist, CGS21680, exacerbated experimental autoimmune neuritis in Lewis rats induced with bovine peripheral myelin. The exacerbation was accompanied with reduced CD4(+)Foxp3(+) T cells, increased CD4(+)CXCR5(+) T cells, B cells, dendritic cells and antigen-specific autoantibodies, which is possibly due to the inhibition of IL-2 induced by CGS21680. Combined with previous studies, our data indicate that the effects of A2AR stimulation in vivo are variable in different diseases. Caution should be taken in the use of A2AR agonists. Topics: Adenosine; Adenosine A2 Receptor Agonists; Animals; Cattle; CD4-Positive T-Lymphocytes; Cytokines; Dendritic Cells; Disease Models, Animal; Female; Immunity, Humoral; Immunoglobulin G; Leukocytes, Mononuclear; Lymph Nodes; Lymphocytes; Myelin Sheath; Nervous System Diseases; Neuritis, Autoimmune, Experimental; Phenethylamines; Rats; Rats, Inbred Lew; Receptors, Adenosine A2; Sciatic Nerve | 2016 |
Optogenetic activation of intracellular adenosine A2A receptor signaling in the hippocampus is sufficient to trigger CREB phosphorylation and impair memory.
Human and animal studies have converged to suggest that caffeine consumption prevents memory deficits in aging and Alzheimer's disease through the antagonism of adenosine A2A receptors (A2ARs). To test if A2AR activation in the hippocampus is actually sufficient to impair memory function and to begin elucidating the intracellular pathways operated by A2AR, we have developed a chimeric rhodopsin-A2AR protein (optoA2AR), which retains the extracellular and transmembrane domains of rhodopsin (conferring light responsiveness and eliminating adenosine-binding pockets) fused to the intracellular loop of A2AR to confer specific A2AR signaling. The specificity of the optoA2AR signaling was confirmed by light-induced selective enhancement of cAMP and phospho-mitogen-activated protein kinase (p-MAPK) (but not cGMP) levels in human embryonic kidney 293 (HEK293) cells, which was abolished by a point mutation at the C terminal of A2AR. Supporting its physiological relevance, optoA2AR activation and the A2AR agonist CGS21680 produced similar activation of cAMP and p-MAPK signaling in HEK293 cells, of p-MAPK in the nucleus accumbens and of c-Fos/phosphorylated-CREB (p-CREB) in the hippocampus, and similarly enhanced long-term potentiation in the hippocampus. Remarkably, optoA2AR activation triggered a preferential p-CREB signaling in the hippocampus and impaired spatial memory performance, while optoA2AR activation in the nucleus accumbens triggered MAPK signaling and modulated locomotor activity. This shows that the recruitment of intracellular A2AR signaling in the hippocampus is sufficient to trigger memory dysfunction. Furthermore, the demonstration that the biased A2AR signaling and functions depend on intracellular A2AR loops prompts the possibility of targeting the intracellular A2AR-interacting partners to selectively control different neuropsychiatric behaviors. Topics: Adenosine; Adenosine A2 Receptor Agonists; Animals; Cell Membrane; Cyclic AMP Response Element-Binding Protein; Disease Models, Animal; Exploratory Behavior; HEK293 Cells; Hippocampus; Humans; In Vitro Techniques; Light; Memory Disorders; Mice; Mice, Inbred C57BL; Phenethylamines; Phosphorylation; Receptors, Adenosine A2; Signal Transduction; Synaptosomes; Transfection | 2015 |
Promotion of Wound Healing by an Agonist of Adenosine A2A Receptor Is Dependent on Tissue Plasminogen Activator.
Impaired wound healing, as it occurs in diabetes mellitus or long-term corticoid treatment, is commonly associated with disability, diminished quality of life, and high economic costs. Selective agonists of the A2A receptor subtype of adenosine, an endogenous regulator of inflammation, promote tissue repair in animal models, both healthy and with impaired healing. Plasmin-mediated proteolysis of fibrin and other matrix proteins is essential for cell migration at sites of injury. Since adenosine A2A receptor activation increases plasminogen activator release from macrophages and mast cells, we studied the effect of a selective agonist, CGS-21680, on full-thickness excisional wound closure in wild-type, urokinase plasminogen activator (uPA)-deficient, and tissue plasminogen activator (tPA)-deficient mice. Wound closure was impaired in tPA- and uPA-deficient mice as compared with wild-type mice, and topical application of CGS-21680 significantly increased the rate at which wounds closed in wild-type mice and uPA-deficient mice, but not in tPA-deficient mice. Immunostaining of tissue sections showed that tPA was present in endothelial cells and histiocytes by day 3 post-wound and also by day 6. In contrast, uPA was more prominent in these cell types only by day 6 post-wound. Our results confirm that plasminogen activation contributes to wound repair and are consistent with the hypothesis that adenosine A2A receptor activation promotes wound closure by a mechanism that depends upon tPA, but not uPA. Moreover, our results suggest that topical adenosine A2A receptor agonists may be useful in promotion of wound closure in patients with impaired wound healing. Topics: Adenosine; Adenosine A2 Receptor Agonists; Administration, Cutaneous; Animals; Disease Models, Animal; Endothelial Cells; Histiocytes; Mice, Inbred C57BL; Mice, Knockout; Phenethylamines; Receptor, Adenosine A2A; Skin; Time Factors; Tissue Plasminogen Activator; Urokinase-Type Plasminogen Activator; Wound Healing; Wounds, Penetrating | 2015 |
Low doses of the selective adenosine A2A receptor agonist CGS21680 are protective in a rat model of transient cerebral ischemia.
Evidence indicate that adenosine A2A receptor subtype is of critical importance in stroke. An overexpression of A2A adenosine receptors occurs at central level on neurons and microglia of ischemic striatum and cortex after focal ischemia. Adenosine A2A receptor subtype is localized not only at central level but also peripherally on blood cells, where it is known to exert antiinflammatory effect. Purpose of the present work was to investigate the putative neuroprotective effect of the adenosine A2A receptor agonist CGS21680 in a rat model of transient medial cerebral artery occlusion (MCAo). Transient cerebral ischemia was induced by 1h occlusion of MCA. CGS21680 (0.01 and 0.1mg/kg, i.p.) was administered starting 4h after ischemia according to a chronic protocol (twice/day for 7 days). CGS21680, at the dose of 0.1mg/kg transiently increased heart frequency but did not modify blood pressure. At the dose of 0.01mg/kg the drug did not modify either heart frequency or blood pressure. Following transient MCAo, CGS21680 at both doses protected from neurological deficit from the first day up to 7 days thereafter. At this time, it has reduced microgliosis, astrogliosis and improved myelin organization in the striatum and cytoarchitecture of the ischemic cortex and striatum. Two days after transient MCAo, CGS21680 has reduced the number of infiltrated granulocytes into the ischemic tissue. Data indicate that CGS21680 systemically administered is protective by immunosuppressive effects. Topics: Adenosine; Adenosine A2 Receptor Agonists; Animals; Blood Pressure; Body Weight; Brain Ischemia; Cerebral Cortex; Corpus Striatum; Disease Models, Animal; Gliosis; Heart Rate; Male; Motor Activity; Neuroprotective Agents; Phenethylamines; Rats; Rats, Wistar; Weight Loss | 2014 |
Topical application of the adenosine A2A receptor agonist CGS-21680 prevents phorbol-induced epidermal hyperplasia and inflammation in mice.
The nucleoside adenosine is a known regulator of immunity and inflammation that mediates, at least in part, the anti-inflammatory effect of methotrexate, an immunosuppressive agent widely used to treat autoimmune inflammatory diseases. Adenosine A2A receptors play a key role in the inhibition of the inflammatory process besides promoting wound healing. Therefore, we aimed to determine the topical effect of a selective agonist, CGS-21680, on a murine model of skin hyperplasia with a marked inflammatory component. Pretreatment with either CGS-21680 (5 μg per site) or the reference agent dexamethasone (200 μg/site) prevented the epidermal hyperplasia and inflammatory response induced by topical application of 12-O-tetradecanoylphorbol-13-acetate (TPA, 2 nmol/site) for three consecutive days. The histological analysis showed that both CGS-21680 and dexamethasone produced a marked reduction of inflammatory cell infiltrate, which correlated with diminished myeloperoxidase (MPO) activity in skin homogenates. Both treatments reduced the levels of the chemotactic mediators LTB4 and CXCL-1, and the inflammatory cytokine TNF-α, through the suppression of NFκB phosphorylation. The immunohistochemical analysis of the hyperproliferative markers cytokeratin 6 (CK6) and Ki67 revealed that while both agents inhibit the number of proliferating cells in the epidermis, CGS-21680 treatment promoted dermal fibroblasts proliferation. Consistently, increased collagen deposition in dermis was observed in tissue sections from agonist-treated mice. Our results showed that CGS 21680 efficiently prevents phorbol-induced epidermal hyperplasia and inflammation in mice without the deleterious atrophic effect of topical corticosteroids. Topics: Adenosine; Adenosine A2 Receptor Agonists; Administration, Topical; Animals; Anti-Inflammatory Agents; Cell Proliferation; Collagen; Cytokines; Dexamethasone; Disease Models, Animal; Epidermis; Female; Hyperplasia; Inflammation; Mice; Peroxidase; Phenethylamines; Skin Diseases; Tetradecanoylphorbol Acetate | 2014 |
Adenosine A2A receptor: a target for regulating renal interstitial fibrosis in obstructive nephropathy.
Renal interstitial fibrosis (RIF) is the common pathological process of chronic kidney diseases leading inevitably to renal function deterioration. RIF and its preceding epithelial-mesenchymal transition (EMT) are commonly triggered by an early occurring renal inflammation. However, an effective approach to prevent EMT and RIF is still lacking and of urgent need. Recently, the adenosine A2A receptor (A2AR) emerges as a novel inflammation regulator, therefore manipulation of A2AR may suppress the EMT process and as such protect against RIF. To test this hypothesis we applied a unilateral ureteral obstruction (UUO) model of RIF on A2AR knockout mice and their wild-type littermates, combined with the intervention of a selective A2AR agonist, CGS 21680. On days 3, 7 and 14 post-UUO we evaluated the effects of A2AR manipulation on the molecular pathological progresses of RIF, including the cellular component of interstitial infiltration, expression of profibrotic factors, cellular biomarkers of EMT, and collagen deposition of extracellular matrix. Our data demonstrated that activation of A2AR significantly suppressed the deposition of collagen types I and III, reduced the infiltration of CD4+ T lymphocytes, and attenuated the expression of TGF-β1 and ROCK1, which in turn inhibited and postponed the EMT progress. Conversely, genetic inactivation of A2AR exacerbated the aforementioned pathological processes of UUO-induced RIF. Together, activation of A2AR effectively alleviated EMT and RIF in mice, suggesting A2AR as a potential therapeutic target for the treatment of RIF. Topics: Adenosine; Adenosine A2 Receptor Agonists; Animals; Biomarkers; CD4-Positive T-Lymphocytes; Cell Movement; Disease Models, Animal; Epithelial-Mesenchymal Transition; Fibrosis; Gene Expression; Humans; Kidney; Mice; Mice, Knockout; Nephritis; Phenethylamines; Receptor, Adenosine A2A; rho-Associated Kinases; Transforming Growth Factor beta1; Ureteral Obstruction | 2013 |
Adenosine A2A receptor contributes to ischemic brain damage in newborn piglet.
Pharmacologic inactivation or genetic deletion of adenosine A2A receptors protects ischemic neurons in adult animals, but studies in neonatal hypoxia-ischemia (H-I) are inconclusive. The present study in neonatal piglets examined the hypothesis that A2A receptor signaling after reoxygenation from global H-I contributes to injury in highly vulnerable striatal neurons where A2A receptors are enriched. A2A receptor immunoreactivity was detected in striatopallidal neurons. In nonischemic piglets, direct infusion of the selective A2A receptor agonist CGS 21680 through microdialysis probes into putamen increased phosphorylation of N-methyl-D-aspartic acid (NMDA) receptor NR1 subunit and Na(+),K(+)-ATPase selectively at protein kinase A (PKA)-sensitive sites. In ischemic piglets, posttreatment with SCH 58261, a selective A2A receptor antagonist, improved early neurologic recovery and preferentially protected striatopallidal neurons. SCH 58261 selectively inhibited the ischemia-induced phosphorylation of NR1, Na(+),K(+)-ATPase, and cAMP-regulated phosphoprotein 32 KDa (DARPP32) at PKA-sensitive sites at 3 hours of recovery and improved Na(+),K(+)-ATPase activity. SCH 58261 also suppressed ischemia-induced protein nitration and oxidation. Thus, A2A receptor activation during reoxygenation contributes to the loss of a subpopulation of neonatal putamen neurons after H-I. Its toxic signaling may be related to DARPP32-dependent phosphorylation of PKA-sensitive sites on NR1 and Na(+),K(+)-ATPase, thereby augmenting excitotoxicity-induced oxidative stress after reoxygenation. Topics: Adenosine; Adenosine A2 Receptor Agonists; Adenosine A2 Receptor Antagonists; Animals; Animals, Newborn; Blood Flow Velocity; Cerebrovascular Circulation; Corpus Striatum; Disease Models, Animal; Hypoxia-Ischemia, Brain; Immunohistochemistry; Laser-Doppler Flowmetry; Male; Neurons; Oxidative Stress; Phenethylamines; Pyrimidines; Receptor, Adenosine A2A; Sus scrofa; Triazoles | 2013 |
The adenosine A2A receptor agonist CGS 21680 decreases ethanol self-administration in both non-dependent and dependent animals.
There is emerging evidence that the adenosinergic system might be involved in drug addiction and alcohol dependence. We have already demonstrated the involvement of A2A receptors (A2AR) in ethanol-related behaviours in mice. Here, we investigated whether the A2AR agonist CGS 21680 can reduce ethanol operant self-administration in both non-dependent and ethanol-dependent Wistar rats. To rule out a potential involvement of the A1R in the effects of CGS 21680, we also tested its effectiveness to reduce ethanol operant self-administration in both heterozygous and homozygous A1R knockout mice. Our results demonstrated that CGS 21680 (0.065, 0.095 and 0.125 mg/kg, i.p.) had a bimodal effect on 10% ethanol operant self-administration in non-dependent rats. The intermediate dose was also effective in reducing 2% sucrose self-administration. Interestingly, the intermediate dose reduced 10% ethanol self-administration in dependent animals more effectively (75% decrease) when compared with non-dependent animals (57% decrease). These results suggest that the A2AR are involved in CGS 21680 effects since the reduction of ethanol self-administration was not dependent upon the presence of A1R in mice. In conclusion, our findings demonstrated the effectiveness of the A2AR agonist CGS 21680 in a preclinical model of alcohol addiction and suggested that the adenosinergic pathway is a promising target to treat alcohol addiction. Topics: Adenosine; Adenosine A2 Receptor Agonists; Alcoholism; Analysis of Variance; Animals; Conditioning, Operant; Disease Models, Animal; Dose-Response Relationship, Drug; Drug-Seeking Behavior; Ethanol; Food Preferences; Male; Mice; Mice, Knockout; Motivation; Phenethylamines; Rats; Rats, Wistar; Receptor, Adenosine A1; Receptors, Adenosine A2; Reinforcement Schedule; Reward; Self Administration; Sucrose | 2013 |
A(2A) adenosine receptors are differentially modulated by pharmacological treatments in rheumatoid arthritis patients and their stimulation ameliorates adjuvant-induced arthritis in rats.
A(2A) adenosine receptors (ARs) play a key role in the inhibition of the inflammatory process. The purpose of this study was to evaluate the modulation of A(2A)ARs in rheumatoid arthritis (RA) patients after different pharmacological treatments and to investigate the effect of A(2A)AR stimulation in a rat model of arthritis. We investigated A(2A)AR density and functionality in RA progression by using a longitudinal study in RA patients before and after methotrexate (MTX), anti-TNFα agents or rituximab treatments. A(2A)ARs were analyzed by saturation binding assays in lymphocytes from RA patients throughout the 24-month study timeframe. In an adjuvant-induced arthritis model in rats we showed the efficacy of the A(2A)AR agonist, CGS 21680 in comparison with standard therapies by means of paw volume assessment, radiographic and ultrasonographic imaging. Arthritic-associated pain was investigated in mechanical allodynia and thermal hyperalgesia tests. IL-10 release following A(2A)AR stimulation in lymphocytes from RA patients and in serum from arthritic rats was measured. In lymphocytes obtained from RA patients, the A(2A)AR up-regulation was gradually reduced in function of the treatment time and the stimulation of these receptors mediated a significant increase of IL-10 production. In the same cells, CGS 21680 did not affected cell viability and did not produced cytotoxic effects. The A(2A)AR agonist CGS 21680 was highly effective, as suggested by the marked reduction of clinical signs, in rat adjuvant-induced arthritis and associated pain. This study highlighted that A(2A)AR agonists represent a physiological-like therapeutic alternative for RA treatment as suggested by the anti-inflammatory role of A(2A)ARs in lymphocytes from RA patients. The effectiveness of A(2A)AR stimulation in a rat model of arthritis supported the role of A(2A)AR agonists as potential pharmacological treatment for RA. Topics: Adenosine; Animals; Anti-Inflammatory Agents; Antibodies, Monoclonal, Murine-Derived; Antirheumatic Agents; Arthritis, Experimental; Arthritis, Rheumatoid; Disease Models, Animal; Humans; Inflammation; Longitudinal Studies; Lymphocytes; Male; Methotrexate; Phenethylamines; Purinergic P1 Receptor Agonists; Rats; Receptors, Purinergic P1; Rituximab; Tumor Necrosis Factor-alpha; Up-Regulation | 2013 |
Adenosine A2A receptors in the nucleus accumbens bi-directionally alter cocaine seeking in rats.
Repeated cocaine administration enhances dopamine D(2) receptor sensitivity in the mesolimbic dopamine system, which contributes to drug relapse. Adenosine A(2A) receptors are colocalized with D(2) receptors on nucleus accumbens (NAc) medium spiny neurons where they antagonize D(2) receptor activity. Thus, A(2A) receptors represent a target for reducing enhanced D(2) receptor sensitivity that contributes to cocaine relapse. The aim of these studies were to determine the effects of adenosine A(2A) receptor modulation in the NAc on cocaine seeking in rats that were trained to lever press for cocaine. Following at least 15 daily self-administration sessions and 1 week of abstinence, lever pressing was extinguished in daily extinction sessions. We subsequently assessed the effects of intra-NAc core microinjections of the A(2A) receptor agonist, CGS 21680 (4-[2-[[6-amino-9-(N-ethyl-b-D-ribofuranuronamidosyl)-9H-purin-2-yl]amino]ethyl]benzenepropanoic acid hydrochloride), and the A(2A) receptor antagonist, MSX-3 (3,7-dihydro-8-[(1E)-2-(3-methoxyphenyl)ethenyl]-7-methyl-3-[3-(phosphonooxy)propyl-1-(2-propynyl)-1H-purine-2,6-dione disodium salt hydrate), in modulating cocaine- and quinpirole-induced reinstatement to cocaine seeking. Intra-NAc pretreatment of CGS 21680 reduced both cocaine- and quinpirole-induced reinstatement. These effects were specific to cocaine reinstatement as intra-NAc CGS 21680 had no effect on sucrose seeking in rats trained to self-administer sucrose pellets. Intra-NAc treatment with MSX-3 modestly reinstated cocaine seeking when given alone, and exacerbated both cocaine- and quinpirole-induced reinstatement. Interestingly, the exacerbation of cocaine seeking produced by MSX-3 was only observed at sub-threshold doses of cocaine and quinpirole, suggesting that removing tonic A(2A) receptor activity enables behaviors mediated by dopamine receptors. Taken together, these findings suggest that A(2A) receptor stimulation reduces, while A(2A) blockade amplifies, D(2) receptor signaling in the NAc that mediates cocaine relapse. Topics: Adenosine; Adenosine A2 Receptor Agonists; Analysis of Variance; Animals; Cocaine; Cocaine-Related Disorders; Conditioning, Operant; Disease Models, Animal; Dopamine Agonists; Dopamine Uptake Inhibitors; Dose-Response Relationship, Drug; Extinction, Psychological; Locomotion; Male; Nucleus Accumbens; Phenethylamines; Purinergic P2X Receptor Antagonists; Quinpirole; Rats; Rats, Sprague-Dawley; Receptor, Adenosine A2A; Reinforcement, Psychology; Self Administration; Sucrose; Sweetening Agents; Xanthines | 2012 |
A2A adenosine receptor agonists reduce both high-palatability and low-palatability food intake in female rats.
The present study examined the effect of two A(2A) adenosine receptor (AR) agonists, CGS 21680 and VT 7, on high-palatability food (HPF) intake in a model of binge eating in sated rats and on low-palatability food (LPF) intake in food-deprived rats. Binge eating was induced in female rats by three 8-day cycles of food restriction/refeeding, followed by acute stress. Two groups of rats were used: NR+NS rats normally fed and not stressed and R+S rats exposed to cycles of food restriction/refeeding and then stressed. R+S rats had higher intake of HPF than the NR+NS controls. The two A(2A)AR agonists were tested at doses of 0.1 and 0.05 mg/kg intraperitoneally; VT 7 did not modify locomotor activity at either dose, whereas CGS 21680 only slightly reduced it at the higher dose tested. The injection of 0.1 mg/kg of both agonists markedly reduced HPF intake both in R+S and in NR+NS rats. The dose of 0.05 mg/kg was inactive. CGS 21680 and VT 7, 0.1 mg/kg, also reduced the standard LPF intake in 24 h food-deprived rats; however, they did not reduce water intake, indicating that their effect on food intake is selective. The dose of 0.05 mg/kg was inactive. Thus, A(2A)AR agonists exert a rather general effect on food intake, inhibiting both HPF intake in sated rats and LPF intake in food-deprived rats. They may potentially be useful pharmacological agents to control binge-related eating disorders and to reduce food overconsumption associated with obesity. Topics: Adenosine; Adenosine A2 Receptor Agonists; Animals; Appetite Depressants; Appetite Regulation; Behavior, Animal; Binge-Eating Disorder; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Therapy, Combination; Energy Intake; Feeding Behavior; Female; Food Preferences; Nerve Tissue Proteins; Phenethylamines; Rats; Rats, Sprague-Dawley; Receptor, Adenosine A2A; Satiety Response; Thionucleosides | 2012 |
Deletion of the BDNF truncated receptor TrkB.T1 delays disease onset in a mouse model of amyotrophic lateral sclerosis.
Brain Derived Neurotrophic Factor (BDNF) exerts strong pro-survival effects on developing and injured motoneurons. However, in clinical trials, BDNF has failed to benefit patients with amyotrophic lateral sclerosis (ALS). To date, the cause of this failure remains unclear. Motoneurons express the TrkB kinase receptor but also high levels of the truncated TrkB.T1 receptor isoform. Thus, we investigated whether the presence of this receptor may affect the response of diseased motoneurons to endogenous BDNF. We deleted TrkB.T1 in the hSOD1(G93A) ALS mouse model and evaluated the impact of this mutation on motoneuron death, muscle weakness and disease progression. We found that TrkB.T1 deletion significantly slowed the onset of motor neuron degeneration. Moreover, it delayed the development of muscle weakness by 33 days. Although the life span of the animals was not affected we observed an overall improvement in the neurological score at the late stage of the disease. To investigate the effectiveness of strategies aimed at bypassing the TrkB.T1 limit to BDNF signaling we treated SOD1 mutant mice with the adenosine A2A receptor agonist CGS21680, which can activate motoneuron TrkB receptor signaling independent of neurotrophins. We found that CGS21680 treatment slowed the onset of motor neuron degeneration and muscle weakness similarly to TrkB.T1 removal. Together, our data provide evidence that endogenous TrkB.T1 limits motoneuron responsiveness to BDNF in vivo and suggest that new strategies such as Trk receptor transactivation may be used for therapeutic intervention in ALS or other neurodegenerative disorders. Topics: Adenosine; Adenosine A2 Receptor Agonists; Amyotrophic Lateral Sclerosis; Animals; Brain-Derived Neurotrophic Factor; Disease Models, Animal; Disease Progression; Mice; Phenethylamines; Receptor, trkB; Superoxide Dismutase | 2012 |
Adenosine 2A receptor is protective against renal injury in MRL/lpr mice.
Adenosine is considered as a potent endogenous anti-inflammatory and immunosuppressive molecule. We examined the roles of A2A-adenosine receptor (A(2A)R) in the progression of lupus nephritis.. MRL/lpr mice were given a selective A(2A)R agonist, CGS21680 (0.4 mg/kg per day, i.p.) while control mice received saline only. After 8 weeks of treatment, mice were sacrificed for assessment of functional and histological parameters as well as inflammatory infiltration in the kidneys. MCP-1, IFN-γ, MHC-II and A(2A)R mRNA expression was evaluated by RT-PCR. Expression of A(2A)R and nuclear NFκB p65 protein was determined by Western blot analysis. Levels of anti-dsDNA antibody and IFN-γ were measured by ELISA.. CGS21680 treatment resulted in significant decrease in proteinuria, blood urea and creatinine as well as improvement in renal histology. Renal macrophage and T-cell infiltration were significantly attenuated in association with suppressed expression of MCP-1, IFN-γ and MHC-II. CGS21680 treatment reduced the level of serum anti-dsDNA and renal immune complex deposition. CGS21680 inhibited the activation of NFκB and suppressed the expression of IFN-γ, MCP-1 and MHC-II in MRL/lpr splenocytes.. A(2A)R activation suppressed inflammation in the kidneys of MRL/lpr mice and can be considered as a novel therapeutic approach for human lupus nephritis. Topics: Adenosine; Adenosine A2 Receptor Agonists; Animals; Blotting, Western; Disease Models, Animal; Disease Progression; Female; Gene Expression; Inflammation; Lupus Erythematosus, Systemic; Lupus Nephritis; Mice; Mice, Inbred MRL lpr; Phenethylamines; Receptor, Adenosine A2A; Reverse Transcriptase Polymerase Chain Reaction; RNA | 2011 |
Adenosine A(2A) receptor activation prevents progressive kidney fibrosis in a model of immune-associated chronic inflammation.
Crescentic glomerulonephritis (GN) in Wistar-Kyoto rats progresses to lethal kidney failure by macrophage (Mφ)-mediated mechanisms. Mφs in nephritic glomeruli express adenosine A(2A) receptors (A(2A)Rs), the activation of which suppresses inflammation. Here, we pharmacologically activated the A(2A)Rs with a selective agonist, CGS 21680, and inactivated them with a selective antagonist, ZM241385, to test the effects on established GN. When activation was delayed until antiglomerular basement membrane GN and extracellular matrix deposition were established, glomerular Mφ infiltration was reduced by 83%. There was also a marked improvement in glomerular lesion histology, as well as decreased proteinuria. A(2A)R activation significantly reduced type I, III, and IV collagen deposition, and E-cadherin expression was restored in association with a reduction of α-smooth muscle actin-positive myofibroblasts in the interstitium and glomeruli. In contrast, pharmacological inactivation of A(2A)Rs increased glomerular crescent formation, type I, III, and IV collagen expression, and enhanced E-cadherin loss. Activation of A(2A)Rs suppressed the expression of the Mφ-linked glomerular damage mediators, transforming growth factor-β, osteopontin-1, thrombospondin-1, and tissue inhibitor of metalloproteinase-1. Thus, A(2A)R activation can arrest GN and prevent progressive fibrosis in established pathological lesions. Topics: Adenosine; Adenosine A2 Receptor Agonists; Adenosine A2 Receptor Antagonists; Animals; Anti-Inflammatory Agents; Autoantibodies; Disease Models, Animal; Extracellular Matrix Proteins; Fibrosis; Glomerulonephritis; Inflammation Mediators; Kidney; Macrophages; Male; Phenethylamines; Rats; Rats, Inbred WKY; Receptor, Adenosine A2A; Severity of Illness Index; Time Factors; Triazines; Triazoles | 2011 |
Post exposure administration of A(1) adenosine receptor agonists attenuates noise-induced hearing loss.
Adenosine is a constitutive cell metabolite with a putative role in protection and regeneration in many tissues. This study was undertaken to determine if adenosine signalling pathways are involved in protection against noise injury. A(1) adenosine receptor expression levels were altered in the cochlea exposed to loud sound, suggesting their involvement in the development of noise injury. Adenosine and selective adenosine receptor agonists (CCPA, CGS-21680 and Cl-IB-MECA) were applied to the round window membrane of the cochlea 6h after noise exposure. Auditory brainstem responses measured 48h after drug administration demonstrated partial recovery of hearing thresholds (up to 20dB) in the cochleae treated with adenosine (non-selective adenosine receptor agonist) or CCPA (selective A(1) adenosine receptor agonist). In contrast, the selective A(2A) adenosine receptor agonist CGS-21680 and A(3) adenosine receptor agonist Cl-IB-MECA did not protect the cochlea from hearing loss. Sound-evoked cochlear potentials in control rats exposed to ambient noise were minimally altered by local administration of the adenosine receptor agonists used in the noise study. Free radical generation in the cochlea exposed to noise was reduced by administration of adenosine and CCPA. This study pinpoints A(1) adenosine receptors as attractive targets for pharmacological interventions to reduce noise-induced cochlear injury after exposure. Topics: Acoustic Stimulation; Adenosine; Adenosine A1 Receptor Agonists; Animals; Auditory Threshold; Cochlea; Cochlear Microphonic Potentials; Disease Models, Animal; Evoked Potentials, Auditory, Brain Stem; Hearing Loss, Noise-Induced; Male; Oxidative Stress; Phenethylamines; Rats; Rats, Wistar; Receptor, Adenosine A1; Time Factors; Tyrosine | 2010 |
Adenosine A1 receptor agonists reduce hyperalgesia after spinal cord injury in rats.
An in vivo study using a spinal cord compression model in rats.. To evaluate the effect of adenosine on thermal hyperalgesia after spinal cord injury (SCI).. After SCI, some patients suffer dysesthesia that is unresponsive to conventional treatments. We previously established a rat thoracic spinal cord mild-compression model by which we were able to induce thermal hyperalgesia in the hind limbs.. The thoracic spinal cord was compressed gently using a 20-g weight for 20 min. The withdrawal latency in response to thermal stimulation was monitored bilaterally in the hind limbs using Hargreaves' Plantar test apparatus.. SCI-induced thermal hyperalgesia was mimicked by the intrathecal application of 8-cyclopentyl-1,3-dipropylxanthine (DPCPX), a selective adenosine A1 receptor antagonist. Hyperalgesia induced by SCI was significantly inhibited by the intrathecal application of 10-30 nmol chloro-adenosine (Cl-adenosine), a nonselective adenosine receptor agonist. The effect of Cl-adenosine (10 nmol) on hyperalgesia after SCI was blocked by the simultaneous application of DPCPX. Intrathecal application of R(-)N6-(2phenylisopropyl) adenosine (R-PIA; 10 nmol), a selective A1 receptor agonist, also inhibited SCI-induced hyperalgesia. In contrast, intrathecal application of CGS21680, a selective adenosine A2a receptor agonist, did not inhibit SCI-induced hyperalgesia.. These results suggest that adenosine inhibits hyperalgesia through the stimulation of A1 receptors. Adenosine or adenosine A1 receptor agonists should be considered as candidates for new therapeutic methods for treating post-SCI dysesthesia. Topics: Adenosine; Adenosine A1 Receptor Agonists; Adenosine A1 Receptor Antagonists; Adenosine A2 Receptor Agonists; Analgesics; Animals; Disease Models, Animal; Female; Hyperalgesia; Phenethylamines; Rats; Rats, Wistar; Receptor, Adenosine A1; Spinal Cord; Spinal Cord Compression; Treatment Outcome; Xanthines | 2010 |
Influence of CGS 21680, a selective adenosine A(2A) receptor agonist, on NMDA receptor function and expression in the brain of Huntington's disease mice.
The effect of chronic treatment with the selective adenosine A(2A) receptor agonist CGS 21680 on N-Methyl-d-Aspartate (NMDA) receptor function and expression has been studied in the striatum and cortex of R6/2 mice, a genetic mouse model of Huntington's disease (HD). Starting from 8weeks of age, R6/2 and wild type (WT) mice were treated daily with CGS 21680 (0.5mg/kg i.p.) for 3weeks and the expression levels of NMDA receptor subunits were then evaluated. In addition, to study CGS 21680-induced changes in NMDA receptor function, NMDA-induced toxicity in corticostriatal slices from both R6/2 and WT mice was investigated. We found that CGS 21680 increased NR2A subunit expression and the NR2A/NR2B ratio in the cortex of R6/2 mice, having no effect in WT mice. In the striatum, CGS 21680 reduced NR1 expression in both R6/2 and WT mice while the effect on NR2A and NR2/NR2B expression was genotype-dependent, reducing and increasing their expression in WT and R6/2 mice, respectively. On the contrary, NMDA-induced toxicity in corticostriatal slices was not modified by the treatment in WT or HD mice. These results demonstrate that in vivo activation of A(2A) receptors modulates the subunit composition of NMDA receptors in the brain of HD mice. Topics: Adenosine; Adenosine A2 Receptor Agonists; Animals; Blotting, Western; Cerebral Cortex; Corpus Striatum; Disease Models, Animal; Electrophysiology; Female; Huntington Disease; Male; Mice; Neurons; Phenethylamines; Protein Subunits; Receptors, N-Methyl-D-Aspartate; Statistics, Nonparametric | 2010 |
Changes induced by formalin pain in central alpha1-adrenoceptor density are modulated by adenosine receptor agonists.
We aimed to elucidate the role of alpha(1)-adrenoceptors in adenosine analgesia in the formalin test. Formalin was injected into the hind paw of male CD-1 mice after injection of adenosine A(1) or A(2a) receptor agonists, CPA, [N(6)-cyclopentyladenosine], and CGS21680 [2-p-(2-carboxyethyl)-phenylethylamino-5'-N-ethylcarboxamidoadenosine hydrochloride]. In the behavioral experiment, alpha(1)-adrenoceptors were blocked by an alpha(1)-adrenoceptor antagonist prazosin, 0.01 mg/kg i.p., and the time mice spent paw licking was recorded for the early (0-15 min) and late (15-60 min) phase of formalin pain. In the neurochemical experiments, mice were killed 15 or 45 min after formalin injection. The density of alpha(1)-adrenoceptors was assessed in various brain areas and in the lumbar spinal cord by [(3)H]prazosin autoradiography. Adenosine agonists produced analgesia in both phases of formalin pain, while prazosin showed a tendency to pronociceptive action in the late phase, and antagonized the effect of CGS21680. After formalin injection, alpha(1)-adrenoceptor density was elevated in some brain areas, mainly in the late phase (some contralateral amygdaloid and ipsilateral thalamic nuclei) and depressed in others (early phase in the ipsilateral spinal cord and late phase in both ipsi- and contralateral sensorimotor cortex). Elevation of alpha(1)-adrenoceptor density, which may be interpreted as a defensive response, did not develop in several cases of CPA-pretreated mice. This suggests that the analgesic effect of adenosine A(1) receptor activation renders the defensive response unnecessary. The depression of alpha(1)-adrenoceptors may suggest development of hypersensitivity in a given structure, and this was antagonized by CGS21680, suggesting the role of A(2a) receptors in control of inflammatory formalin pain. Topics: Adenosine; Adenosine A2 Receptor Agonists; Adenosine A2 Receptor Antagonists; Analgesics; Animals; Autoradiography; Central Nervous System; Disease Models, Animal; Inflammation; Male; Mice; Nociceptors; Pain; Pain Measurement; Phenethylamines; Prazosin; Receptor, Adenosine A2A; Receptors, Adrenergic, alpha-1 | 2010 |
Contribution of adenosine A(2A) and A(2B) receptors to ischemic coronary dilation: role of K(V) and K(ATP) channels.
This study was designed to elucidate the contribution of adenosine A(2A) and A(2B) receptors to coronary reactive hyperemia and downstream K(+) channels involved. Coronary blood flow was measured in open-chest anesthetized dogs. Adenosine dose-dependently increased coronary flow from 0.72 ± 0.1 to 2.6 ± 0.5 mL/minute/g under control conditions. Inhibition of A(2A) receptors with SCH58261 (1 μm) attenuated adenosine-induced dilation by ∼50%, while combined administration with the A(2B) receptor antagonist alloxazine (3 μm) produced no additional effect. SCH58261 significantly reduced reactive hyperemia in response to a transient 15 second occlusion; debt/repayment ratio decreased from 343 ± 63 to 232 ± 44%. Alloxazine alone attenuated adenosine-induced increases in coronary blood flow by ∼30% but failed to alter reactive hyperemia. A(2A) receptor agonist CGS21680 (10 μg bolus) increased coronary blood flow by 3.08 ± 0.31 mL/minute/g. This dilator response was attenuated to 0.76 ± 0.14 mL/minute/g by inhibition of K(V) channels with 4-aminopyridine (0.3mm) and to 0.11 ± 0.31 mL/minute/g by inhibition of K(ATP) channels with glibenclamide (3 mg/kg). Combined administration abolished vasodilation to CGS21680. These data indicate that A(2A) receptors contribute to coronary vasodilation in response to cardiac ischemia via activation of K(V) and K(ATP) channels. Topics: 4-Aminopyridine; Adenosine; Adenosine A2 Receptor Agonists; Adenosine A2 Receptor Antagonists; Animals; Coronary Circulation; Disease Models, Animal; Dogs; Flavins; Glyburide; Hyperemia; KATP Channels; Male; Myocardial Ischemia; Phenethylamines; Potassium Channel Blockers; Potassium Channels, Voltage-Gated; Pyrimidines; Receptor, Adenosine A2A; Receptor, Adenosine A2B; Triazoles; Vasodilation | 2010 |
Blocking adenosine A2A receptor reduces peritoneal fibrosis in two independent experimental models.
Long-term peritoneal dialysis (PD) is associated with peritoneal fibrosis and loss of function. It has been shown that activation of the adenosine A(2A) receptor (A(2A)R) promotes tissue repair, wound healing and extracellular matrix (ECM) production. We have previously shown that adenosine is a potent regulator of inflammation in the peritoneum. In the current study, we explored the role of adenosine and the A(2A)R in two experimental models.. Collagen deposition was evaluated in primary peritoneal fibroblasts following treatment with an A(2A)R agonist and antagonist. In addition, peritoneal fibrosis was induced by i.p. injection of either chlorhexidine gluconate for 2 weeks or 4.25% glucose peritoneal dialysis fluid (PDF) for 1 month. The development of fibrosis was compared between wild-type (WT) and WT mice treated with caffeine (an A(2A)R antagonist) in drinking water or between (A(2A)R(+/+)) mice and A(2A)R-deficient mice (A(2A)R(-/-)).. Adenosine or the A(2A)R agonist CGS21680 stimulated collagen production by peritoneal fibroblasts in vitro and A(2A)R antagonists (ZM241385 and caffeine) blocked this effect. Consistent with these results, caffeine-treated WT or A(2A)R(-/-) mice had reduced submesothelial thickness, collagen deposition and mRNA levels of fibroblast-specific protein (FSP-1) and connective tissue growth factor (CTGF). In addition, treatment with caffeine in vitro and in vivo diminished A(2A)R and A(2B)R mRNA levels induced by CG or PDF while it upregulated A(1)R levels.. Our data suggest that adenosine through its A(2A)R promotes peritoneal fibrosis and therefore should be considered as a target for pharmacological intervention. Topics: Adenosine; Adenosine A2 Receptor Agonists; Adenosine A2 Receptor Antagonists; Animals; Caffeine; Cells, Cultured; Central Nervous System Stimulants; Chlorhexidine; Collagen; Disease Models, Animal; Fibroblasts; Fibrosis; Mice; Mice, Knockout; Peritoneal Cavity; Phenethylamines; Vasodilator Agents | 2009 |
A2A adenosine receptor deficiency leads to impaired tracheal relaxation via NADPH oxidase pathway in allergic mice.
A(2A) adenosine receptor (A(2A)AR) has been shown to suppress superoxide generation in leukocytes via the cAMP-protein kinase A (PKA) pathway. However, no study has yet explored the role of A(2A)AR in relation to NADPH oxidase in murine tracheas in vitro, which may lead to altered smooth muscle relaxation in asthma. Therefore, the present study evaluated the effects of A(2A)AR deficiency on the NADPH oxidase pathway in tracheas of A(2A) wild-type (WT) and A(2A) knockout (KO) mice. A(2A)WT mice were sensitized with ovalbumin (30 microg i.p.) on days 1 and 6, followed by 5% ovalbumin aerosol challenge on days 11, 12, and 13. A(2A)AR (gene and protein expression), cAMP, and phosphorylated PKA (p-PKA) levels were decreased in A(2A)WT sensitized mice compared with controls. A(2A)KO mice also showed decreased cAMP and p-PKA levels. A(2A)WT sensitized and A(2A)KO control mice had increased gene and protein expression of NADPH oxidase subunits (p47phox and gp91phox) compared with the controls. Tracheal relaxation to specific A(2A)AR agonist, 4-[2-[[6-amino-9-(N-ethyl-beta-d-ribofuranuronamidosyl)-9H-purin-2-yl]amino]ethyl]benzenepropanoic acid hydrochloride (CGS 21680), decreased in A(2A)WT sensitized mice compared with the controls, although it was absent in A(2A)KO mice. Pretreatment with NADPH oxidase inhibitors apocyanin/diphenyliodonium reversed the attenuated relaxation to CGS 21680 in A(2A)WT sensitized tracheas, whereas specific PKA inhibitor (9S,10S,12R)-2,3,9,10,11,12-hexahydro-10-hydroxy-9-methyl-1-oxo-9,12-epoxy-1H-diindolo[1,2,3-fg:3',2',1'-kl]pyrrolo[3,4-i] [1,6]benzodiazocine-10-carboxylic acid hexyl ester (KT 5720) blocked CGS 21680-induced relaxation. Tracheal reactive oxygen species (ROS) generation was also increased in A(2A)WT sensitized and A(2A)KO control mice compared with the controls. In conclusion, this study shows that A(2A)AR deficiency causes increased NADPH oxidase activation leading to decreased tracheal relaxation via altered cAMP-PKA signaling and ROS generation. Topics: Adenosine; Adenosine A2 Receptor Agonists; Animals; Asthma; Disease Models, Animal; Female; In Vitro Techniques; Male; Mice; Mice, Inbred C57BL; Mice, Knockout; Muscle Relaxation; Muscle, Smooth; NADPH Oxidases; Phenethylamines; Reactive Oxygen Species; Receptor, Adenosine A2A; Signal Transduction; Trachea | 2009 |
Spinal NMDA receptor activation is necessary for de novo, but not the maintenance of, A2a receptor-mediated phrenic motor facilitation.
Adenosine 2a (A2a) receptor agonists elicit persistent increases in phrenic nerve activity by transactivating the neurotrophin receptor, TrkB, near phrenic motoneurons. Our working model proposes that A2a receptor-mediated TrkB receptor activation strengthens glutamatergic synapses onto phrenic motoneurons. Activation of glutamate N-methyl d-aspartate (NMDA) receptors has been implicated in other models of phrenic motor plasticity. Thus we hypothesized that NMDA receptor activation also would contribute to A2a receptor-mediated phrenic motor facilitation. Adult male Sprague-Dawley rats were anesthetized with urethane, mechanically ventilated, neuromuscularly paralyzed, and bilaterally vagotomized. The A2a receptor agonist CGS-21680 and the NMDA receptor-channel blocker MK-801 were administered intrathecally over the C4 spinal segment. Phrenic nerve activity was recorded before, during, and after drug administration. MK-801 (concentration range 0.1, 1.0, 10.0, and 100 microM) was administered 30 min before CGS-21680 (50 microM). MK-801 dose-dependently blocked A2a receptor-mediated phrenic motor facilitation. When administered at 60 min post-CGS-21680, MK-801 prevented further increases in phrenic nerve activity compared with the CGS-21680 alone (CGS-21680 alone at 120 min: 114 +/- 19%; CGS-21680 and MK-801 at 60 min post-CGS-21680: 61 +/- 11%, above baseline, P < 0.05) but did not return phrenic motor output to baseline values. Our data suggest that NMDA receptor activation is necessary for de novo A2a receptor-mediated phrenic motor facilitation and that the maintenance of preexisting phrenic motor facilitation does not involve NMDA receptor-dependent mechanisms. Topics: Adenosine; Adenosine A2 Receptor Antagonists; Animals; Antihypertensive Agents; Disease Models, Animal; Dizocilpine Maleate; Drug Interactions; Male; Motor Neurons; Neuroprotective Agents; Phenethylamines; Phrenic Nerve; Rats; Rats, Sprague-Dawley; Receptor, Adenosine A2A; Receptor, trkB; Receptors, N-Methyl-D-Aspartate; Spinal Cord; Spinal Cord Injuries | 2009 |
The A2A adenosine receptor rescues the urea cycle deficiency of Huntington's disease by enhancing the activity of the ubiquitin-proteasome system.
Huntington's disease (HD) is an autosomal dominant neurodegenerative disease caused by a CAG trinucleotide expansion in the Huntingtin (Htt) gene. The resultant mutant Htt protein (mHtt) forms aggregates in the brain and several peripheral tissues (e.g. the liver) and causes devastating neuronal degeneration. Metabolic defects resulting from Htt aggregates in peripheral tissues also contribute to HD pathogenesis. Simultaneous improvement of defects in both the CNS and peripheral tissues is thus the most effective therapeutic strategy and is highly desirable. We earlier showed that an agonist of the A(2A) adenosine receptor (A(2A) receptor), CGS21680 (CGS), attenuates neuronal symptoms of HD. We found herein that the A(2A) receptor also exists in the liver, and that CGS ameliorated the urea cycle deficiency by reducing mHtt aggregates in the liver. By suppressing aggregate formation, CGS slowed the hijacking of a crucial transcription factor (HSF1) and two protein chaperons (Hsp27 and Hsp70) into hepatic Htt aggregates. Moreover, the abnormally high levels of high-molecular-mass ubiquitin conjugates in the liver of an HD mouse model (R6/2) were also ameliorated by CGS. The protective effect of CGS against mHtt-induced aggregate formation was reproduced in two cells lines and was prevented by an antagonist of the A(2A) receptor and a protein kinase A (PKA) inhibitor. Most importantly, the mHtt-induced suppression of proteasome activity was also normalized by CGS through PKA. Our findings reveal a novel therapeutic pathway of A(2A) receptors in HD and further strengthen the concept that the A(2A) receptor can be a drug target in treating HD. Topics: Adenosine; Adenosine A2 Receptor Antagonists; Animals; Cell Line; Disease Models, Animal; Female; Humans; Huntington Disease; Liver; Male; Mice; Phenethylamines; Proteasome Endopeptidase Complex; Receptor, Adenosine A2A; Serotonin Plasma Membrane Transport Proteins; Ubiquitin; Urea | 2009 |
Protective effects of adenosine A2A receptor agonist in ventilator-induced lung injury in rats.
Mechanical ventilation is associated with overwhelming inflammatory responses that are associated with ventilator-induced lung injury (VILI) in patients with acute respiratory distress syndrome. The activation of adenosine A2A receptors has been reported to attenuate inflammatory cascades.. The administration of A2A receptors agonist ameliorates VILI.. Rats were subjected to hemorrhagic shock and resuscitation as a first hit to induce systemic inflammation. The animals randomly received the selective A2A receptor agonist CGS-21680 or a vehicle control in a blinded fashion at the onset of resuscitation phase. They were then randomized to receive mechanical ventilation as a second hit with a high tidal volume of 20 mL/kg and zero positive end-expiratory pressure, or a low tidal volume of 6 mL/kg with positive end-expiratory pressure of 5 cm H2O.. The administration of CGS-21680 attenuated lung injury as evidenced by a decrease in respiratory elastance, lung edema, lung injury scores, neutrophil recruitment in the lung, and production of inflammatory cytokines, compared with the vehicle-treated animals.. The selective A2A receptor agonist may have a place as a novel therapeutic approach in reducing VILI. Topics: Adenosine; Adenosine A2 Receptor Agonists; Animals; Blood Gas Analysis; Blood Pressure; Cytokines; Disease Models, Animal; Male; Phenethylamines; Rats; Rats, Sprague-Dawley; Respiratory Distress Syndrome; Respiratory Mechanics; Shock, Hemorrhagic; Ventilator-Induced Lung Injury | 2009 |
Altered distribution and function of A2A adenosine receptors in the brain of WAG/Rij rats with genetic absence epilepsy, before and after appearance of the disease.
The involvement of excitatory adenosine A(2A) receptors (A(2A)Rs), which probably contribute to the pathophysiology of convulsive seizures, has never been investigated in absence epilepsy. Here, we examined the distribution and function of A(2A)Rs in the brain of Wistar Albino Glaxo/Rijswijk (WAG/Rij) rats, a model of human absence epilepsy in which disease onset occurs 2-3 months after birth. In the cerebral areas that are mostly involved in the generation of absence seizures (somatosensory cortex, reticular and ventrobasal thalamic nuclei), A(2A)R density was lower in presymptomatic WAG/Rij rats than in control rats, as evaluated by immunohistochemistry and western blotting. Accordingly, in cortical/thalamic slices prepared from the brain of these rats, A(2A)R stimulation with the agonist 2-[4-(-2-carboxyethyl)-phenylamino]-5'-N-ethylcarboxamido-adenosine failed to modulate either cAMP formation, mitogen-activated protein kinase system, or K(+)-evoked glutamate release. In contrast, A(2A)R expression, signalling and function were significantly enhanced in brain slices from epileptic WAG/Rij rats as compared with matched control animals. Additionally, the in vivo injection of the A(2A)R agonist CGS21680, or the antagonist 5-amino-7-(2-phenylethyl)-2-(2-fuyl)-pyrazolo-(4,3-c)1,2,4-triazolo(1,5-c)-pyrimidine, in the examined brain areas of epileptic rats, increased and decreased, respectively, the number/duration of recorded spontaneous spike-wave discharges in a dose-dependent manner during a 1-5 h post-treatment period. Our results support the hypothesis that alteration of excitatory A(2A)R is involved in the pathogenesis of absence seizures and might represent a new interesting target for the therapeutic management of this disease. Topics: Adenosine; Age Factors; Analysis of Variance; Animals; Blotting, Western; Disease Models, Animal; Disease Progression; Electroencephalography; Epilepsy, Absence; Glutamic Acid; Immunohistochemistry; Male; MAP Kinase Signaling System; Microinjections; Phenethylamines; Pyrimidines; Rats; Receptor, Adenosine A2A; Somatosensory Cortex; Thalamus; Triazoles | 2009 |
Effects of binge alcohol exposure in the second trimester on intracerebral arteriolar function in third trimester fetal sheep.
Fetal alcohol syndrome is a leading cause of mental retardation, but mechanisms of alcohol-associated brain damage remain elusive. Chronic alcohol exposure attenuates fetal and neonatal hypoxic cerebral vasodilation in sheep. We therefore hypothesized that alcohol could alter development of cerebrovascular responses to adenosine, a putative mediator of hypoxic cerebral vasodilation. The objective of this study was to examine the effect of earlier fetal alcohol exposure on later reactivity to adenosine in fetal sheep cerebral arterioles. Penetrating intracerebral arterioles were harvested from the brains of third trimester fetal sheep previously exposed in the second trimester to maternal alcohol "binges" (1.5 g/kg IV over 90 min, 5 days/week for 4 weeks) or same-volume saline infusions. Arterioles were cannulated with a micropipette system and luminally pressurized. Fetal alcohol exposure did not affect spontaneous myogenic tone, but enhanced the dilator response of penetrating arterioles to extraluminal acidosis (pH 6.8). Alcohol exposure also resulted in an increase in maximal vessel response to CGS-21680, an adenosine A2A receptor agonist, but did not alter the concentration-dependent response curves to adenosine. Our results suggest that earlier alcohol exposure does not impair the subsequent responsiveness of fetal cerebral arterioles to vasodilator agents. Thus, alteration in cerebral vascular response to hypoxia in fetal sheep may not be attributed to changes in vascular reactivity to adenosine. Topics: Adenosine; Animals; Antihypertensive Agents; Arterioles; Cerebrovascular Circulation; Disease Models, Animal; Dose-Response Relationship, Drug; Embryo, Mammalian; Ethanol; Female; Fetal Alcohol Spectrum Disorders; Hypoxia; Phenethylamines; Pregnancy; Prenatal Exposure Delayed Effects; Sheep; Vasodilation | 2008 |
Adenosine A2A receptor activation reduces infarct size in the isolated, perfused mouse heart by inhibiting resident cardiac mast cell degranulation.
Mast cells are found in the heart and contribute to reperfusion injury following myocardial ischemia. Since the activation of A2A adenosine receptors (A2AARs) inhibits reperfusion injury, we hypothesized that ATL146e (a selective A2AAR agonist) might protect hearts in part by reducing cardiac mast cell degranulation. Hearts were isolated from five groups of congenic mice: A2AAR+/+ mice, A2AAR(-/-) mice, mast cell-deficient (Kit(W-sh/W-sh)) mice, and chimeric mice prepared by transplanting bone marrow from A2AAR(-/-) or A2AAR+/+ mice to radiation-ablated A2AAR+/+ mice. Six weeks after bone marrow transplantation, cardiac mast cells were repopulated with >90% donor cells. In isolated, perfused hearts subjected to ischemia-reperfusion injury, ATL146e or CGS-21680 (100 nmol/l) decreased infarct size (IS; percent area at risk) from 38 +/- 2% to 24 +/- 2% and 22 +/- 2% in ATL146e- and CGS-21680-treated hearts, respectively (P < 0.05) and significantly reduced mast cell degranulation, measured as tryptase release into reperfusion buffer. These changes were absent in A2AAR(-/-) hearts and in hearts from chimeric mice with A2AAR(-/-) bone marrow. Vehicle-treated Kit(W-sh/W-sh) mice had lower IS (11 +/- 3%) than WT mice, and ATL146e had no significant protective effect (16 +/- 3%). These data suggest that in ex vivo, buffer-perfused hearts, mast cell degranulation contributes to ischemia-reperfusion injury. In addition, our data suggest that A2AAR activation is cardioprotective in the isolated heart, at least in part by attenuating resident mast cell degranulation. Topics: Adenosine; Adenosine A2 Receptor Agonists; Animals; Bone Marrow Transplantation; Cardiovascular Agents; Cell Degranulation; Cells, Cultured; Cyclohexanecarboxylic Acids; Disease Models, Animal; Mast Cells; Mice; Mice, Inbred C57BL; Mice, Knockout; Mice, Mutant Strains; Myocardial Infarction; Myocardial Reperfusion Injury; Myocardium; Perfusion; Phenethylamines; Purines; Receptor, Adenosine A2A; Time Factors; Tryptases; Whole-Body Irradiation | 2008 |
Fetal alcohol exposure alters cerebrovascular reactivity to vasoactive intestinal peptide in adult sheep.
Chronic fetal alcohol exposure impairs neural and vascular development. We have previously shown that fetal alcohol exposure is associated with attenuated hypoxic cerebral vasodilation and reduced neuronal vasoactive intestinal peptide (VIP) expression in fetal sheep. In the present study, we tested the hypothesis that fetal alcohol exposure alters vascular development, leading to altered cerebral vascular reactivity to VIP in adulthood. Penetrating intracerebral arterioles were harvested from the brains of adult (10-13 months old) offspring of ewes that had received intravenous infusions of alcohol (1.5 g/kg) or same-volume saline (90 min/day, 5 days/week) during days 30-82 of gestation (full term = 145 days). The isolated arterioles were cannulated with a micropipette system that allowed luminal perfusion and control of luminal pressure and developed spontaneous tone at 40 degrees C and 60 mm Hg luminal pressure. There was no difference in myogenic tone between arterioles exposed prenatally to alcohol (n = 18) and saline controls (n = 17). However, fetal alcohol exposure significantly (p = 0.03) enhanced the dilator responses of adult intracerebral arterioles to VIP [0.1 nM to 1 microM, logEC(50): -8.6 +/- 0.2 (alcohol) vs. -7.4 +/- 0.8 (saline)]. In contrast, there was no difference in dilator responses to H(+) (pH 6.8 buffer), to adenosine (10 nM to 0.1 mM), or to CGS21680 (an adenosine A(2A) receptor agonist, 0.01 nM to 10 microM). Thus, fetal alcohol exposure alters vasomotor sensitivity to VIP in adult intracerebral arterioles - perhaps a compensatory response to alcohol-induced underdevelopment of neurotransmitter pathways involved in cerebral vascular regulation. Topics: Adenosine; Animals; Animals, Newborn; Arterioles; Brain; Cerebrovascular Circulation; Disease Models, Animal; Ethanol; Female; Fetal Alcohol Spectrum Disorders; In Vitro Techniques; Male; Phenethylamines; Pregnancy; Prenatal Exposure Delayed Effects; Purinergic P1 Receptor Agonists; Sheep; Vasoactive Intestinal Peptide; Vasodilation; Vasodilator Agents | 2008 |
Regulation of A(2A) adenosine receptor expression and functioning following permanent focal ischemia in rat brain.
Ischemia, through modulation of adenosine receptors (ARs), may influence adenosine-mediated-cellular responses. In the present study, we investigated the modulation of rat A(2A) receptor expression and functioning, in rat cerebral cortex and striatum, following in vivo focal ischemia (24 h). In cortex, middle cerebral artery occlusion did not induce any alterations in A(2A) receptor binding and functioning. On the contrary, in striatum, a significant decrease in A(2A) ligand affinity, associated with an increase in receptor density, were detected. In striatum, ischemia also induced a significant reduction both in G protein pool and in A(2A) receptor-G protein coupling. On the contrary, A(2A) receptor functional responsiveness, measured as stimulation of adenylyl cyclise, was not affected by ischemia, suggesting receptor up-regulation may represent a compensatory mechanism to maintain receptor functioning during cerebral damage. Immunohistochemical study showed that following 24 h middle cerebral artery occlusion, A(2A) ARs were definitely expressed both on neurons and activated microglia in ischemic striatum and cortex, but were not detected on astrocytes. In the non-ischemic hemisphere and in sham-operated rats A(2A) ARs were barely detected. Modifications of ARs may play a significant role in determining adenosine effects during ischemia and therefore should be taken into account when evaluating time-dependent protective effects of specific A(2A) active compounds. Topics: Adenosine; Adenosine A2 Receptor Agonists; Animals; Astrocytes; Behavior, Animal; Brain; Brain Ischemia; Disease Models, Animal; Gene Expression Regulation; Guanosine 5'-O-(3-Thiotriphosphate); Male; Motor Activity; Neurologic Examination; Phenethylamines; Rats; Rats, Wistar; Receptors, Adenosine A2 | 2008 |
[Different effects of adenosine A2A receptors in the models of traumatic brain injury and peripheral tissue injury].
Recently, activation of the adenosine A2A receptors has been shown to exert protection against peripheral tissue injuries but aggravation in the central nervous system (CNS) injuries. To explore the different effects of adenosine A2A receptors and try to perform some new treatment strategies for peripheral tissue and CNS traumas, we constructed the mouse models of skin trauma, skin combined radiation-impaired wound and traumatic brain injury (TBI), respectively. Wild type mice and A2A receptor gene knockout mice were both used in the experiments. In skin trauma and combined radiation-impaired wound models, the time of wound healing was observed, while in TBI model, neurological deficit scores, water content in injured brain and glutamate concentration in cerebral spinal fluid (CSF) were detected at 24 h after TBI. The results showed that in skin trauma and combined radiation-impaired wound models, CGS21680 (an agonist of the A2A receptors) promoted while A2A receptor gene knockout delayed the course of skin wound healing. On the contrary, in TBI model, A2A receptor gene knockout, not CGS21680, showed a protective role by inhibition of glutamate release. These data further indicate that promoting glutamate release may account for the different effects of A2A receptor activation in CNS injury and peripheral tissue injury models. These findings may provide some experimental evidence and a new strategy for clinical treatment of peripheral tissue damages by agonists of A2A receptors, while treatment of CNS injuries by antagonists of A2A receptors. Topics: Adenosine; Animals; Brain; Brain Injuries; Disease Models, Animal; Glutamic Acid; Mice; Mice, Knockout; Phenethylamines; Receptor, Adenosine A2A; Wound Healing | 2008 |
Opposite effects of the A2A receptor agonist CGS21680 in the striatum of Huntington's disease versus wild-type mice.
Huntington's disease (HD) is an inherited neurodegenerative disorder. Adenosine A(2A) receptors (A(2A)Rs) are involved in excitotoxic/neurodegenerative processes, and A(2A)R ligands may be neuroprotective in models of HD. However, changes in the transcription, expression and function of A(2A)Rs have been reported to occur in HD models. The aim of the present work was to verify whether A(2A)R-mediated effects are altered in the striatum of transgenic HD (R6/2) versus wild-type (WT) mice. Extracellular field potentials (FPs) were recorded in corticostriatal slices from R6/2 mice in early (7-8 weeks) or frankly (12-13 weeks) symptomatic phases, and age-matched WT. In 12-13 weeks aged WT animals, the application of 75 microM NMDA induced a transient disappearance of the FP followed by an almost complete recovery at washout. In slices from HD mice, the mean FP recovery was significantly reduced (P<0.01 versus WT). A(2A)R activation oppositely modulated NMDA-induced toxicity in the striatum of HD versus WT mice. Indeed, the A(2A)R agonist CGS21680 reduced the FP recovery in slices from WT mice, while it significantly increased it in slices from R6/2 mice. In early symptomatic (7-8 weeks) mice, no differences were observed between WT and HD animals in terms of basal synaptic transmission and response to NMDA. At the same age, the behavioural effects elicited by CGS21680 were qualitatively identical in WT and HD mice. These findings may have very important implications for the neuroprotective potential of A(2A)R ligands in HD. Topics: Action Potentials; Adenosine; Adenosine A2 Receptor Agonists; Animals; Antihypertensive Agents; Corpus Striatum; Disease Models, Animal; Huntington Disease; Mice; Mice, Transgenic; Neuroprotective Agents; Neurotoxins; Organ Culture Techniques; Phenethylamines; Receptor, Adenosine A2A; Receptors, N-Methyl-D-Aspartate; Recovery of Function; Species Specificity; Synaptic Transmission | 2007 |
Attenuation of experimental subarachnoid hemorrhage--induced cerebral vasospasm by the adenosine A2A receptor agonist CGS 21680.
Impaired endothelium-dependent relaxation is present in vasospastic cerebral vessels after subarachnoid hemorrhage (SAH) and may result from deficient production of endothelial nitric oxide synthase (eNOS) or increased production and/or activity of inducible NOS (iNOS). Accumulating evidence demonstrates that adenosine A2A receptors increase the production of NO by human and porcine arterial endothelial cells, which in turn leads to vasodilation. This study was designed to examine the effects of an adenosine A2A receptor agonist, (2(4-[2-carboxyethyl]phenyl)ethylamino)-5'-N-ethylcarboxamidoadenosine (CGS 21680), in the prevention of SAH-induced vasospasm.. . Experimental SAH was induced in Sprague-Dawley rats by injecting 0.3 ml of autologous blood into the cisterna magna of each animal. Intraperitoneal injections of CGS 21680 or vehicle were administered 5 minutes and 24 hours after induction of SAH. The degree of vasospasm was determined by averaging measurements of cross-sectional areas of the basilar artery (BA) 48 hours after SAH. Expression of eNOS and iNOS in the BA was also evaluated. Prior to perfusion-fixation, there were no significant differences among animals in the control and treated groups in any physiological parameter that was recorded. The CGS 21680 treatment significantly attenuated SAH-induced vasospasm. Induction of iNOS mRNA and protein in the BA by the SAH was significantly diminished by administration of CGS 21680. The SAH-induced suppression of eNOS mRNA and protein was also relieved by the CGS 21680 treatment.. This is the first evidence that adenosine A2A receptor agonism is effective in preventing SAH-induced vasospasm without significant complications. The beneficial effect of adenosine A2A receptor agonists may be, at least in part, related to the prevention of augmented expression of iNOS and the preservation of normal eNOS expression following SAH. Adenosine A2A receptor agonism holds promise in the treatment of cerebral vasospasm following SAH and merits further investigation. Topics: Adenosine; Adenosine A2 Receptor Agonists; Animals; Antihypertensive Agents; Disease Models, Animal; Nitric Oxide Synthase Type II; Nitric Oxide Synthase Type III; Phenethylamines; Rats; Rats, Sprague-Dawley; RNA, Messenger; Subarachnoid Hemorrhage; Vasospasm, Intracranial | 2007 |
The role of adenosine A1 and A2A receptors of entorhinal cortex on piriform cortex kindled seizures in rats.
In this research the role of adenosine A1 and A2A receptors of the entorhinal cortex on piriform cortex kindled seizures was investigated. In piriform cortex kindled rats, N6-cyclohexyladenosine (CHA), a selective A1 receptor agonist, 1,3-dimethyl-8 cyclopenthylxanthine (CPT), a selective A1 receptor antagonist, CGS21680 hydrochloride (CGS), a selective A2A receptor agonist and ZM241385 (ZM), a selective A2A receptor antagonist were injected into the entorhinal cortex bilaterally. Five minutes later, animals were stimulated and seizure parameters were recorded. CHA (10 and 100microM) decreased the afterdischarge duration (ADD), stage 5 seizure duration (S5D), and seizure duration (SD), and increased the latency to stage 4 of the seizure (S4L) significantly. Bilateral microinjection of CPT (100microM) increased ADD, S(5)D, and SD, and reduced S(4)L significantly. Pretreatment of animals with CPT (50microM) before CHA (100microM), reduced the effect of CHA on seizure parameters. On the other hand, CGS (1mM) increased only ADD. Bilateral microinjection of ZM had no effect on seizure parameters. However, pretreatment of animals with ZM (200microM) before CGS (1mM), eliminated the excitatory effect of CGS on seizure parameters. These results suggest that activation of A1 receptors of the entorhinal cortex has an anticonvulsant, but activation of A2A receptors of this region has a proconvulsive effect on piriform cortex kindled seizures. Topics: Adenosine; Animals; Anticonvulsants; Convulsants; Disease Models, Animal; Electric Stimulation; Electrodes, Implanted; Entorhinal Cortex; Kindling, Neurologic; Male; Microinjections; Phenethylamines; Rats; Rats, Sprague-Dawley; Receptor, Adenosine A1; Receptor, Adenosine A2A; Seizures; Triazines; Triazoles; Xanthines | 2007 |
Activation of central adenosine A(2A) receptors enhances superior laryngeal nerve stimulation-induced apnea in piglets via a GABAergic pathway.
Activation of the laryngeal mucosa results in apnea that is mediated through, and can be elicited via electrical stimulation of, the superior laryngeal nerve (SLN). This potent inhibitory reflex has been suggested to play a role in the pathogenesis of apnea of prematurity and sudden infant death syndrome, and it is attenuated by theophylline and blockade of GABA(A) receptors. However, the interaction between GABA and adenosine in the production of SLN stimulation-induced apnea has not been previously examined. We hypothesized that activation of adenosine A(2A) receptors will enhance apnea induced by SLN stimulation while subsequent blockade of GABA(A) receptors will reverse the effect of A(2A) receptor activation. The phrenic nerve responses to increasing levels of SLN stimulation were measured before and after sequential intracisternal administration of the adenosine A(2A) receptor agonist CGS (n = 10) and GABA(A) receptor blocker bicuculline (n = 7) in ventilated, vagotomized, decerebrate, and paralyzed newborn piglets. Increasing levels of SLN stimulation caused progressive inhibition of phrenic activity and lead to apnea during higher levels of stimulation. CGS caused inhibition of baseline phrenic activity, hypotension, and enhancement of apnea induced by SLN stimulation. Subsequent bicuculline administration reversed the effects of CGS and prevented the production of apnea compared with control at higher SLN stimulation levels. We conclude that activation of adenosine A(2A) receptors enhances SLN stimulation-induced apnea probably via a GABAergic pathway. We speculate that SLN stimulation causes endogenous release of adenosine that activates A(2A) receptors on GABAergic neurons, resulting in the release of GABA at inspiratory neurons and subsequent respiratory inhibition. Topics: Adenosine; Animals; Animals, Newborn; Apnea; Bicuculline; Blood Pressure; Cisterna Magna; Disease Models, Animal; Drug Antagonism; Electric Stimulation; GABA Antagonists; gamma-Aminobutyric Acid; Heart Rate; Injections, Intraventricular; Laryngeal Nerves; Phenethylamines; Phrenic Nerve; Receptor, Adenosine A2A; Swine | 2007 |
Protective roles of adenosine A1, A2A, and A3 receptors in skeletal muscle ischemia and reperfusion injury.
Although adenosine exerts cardio-and vasculoprotective effects, the roles and signaling mechanisms of different adenosine receptors in mediating skeletal muscle protection are not well understood. We used a mouse hindlimb ischemia-reperfusion model to delineate the function of three adenosine receptor subtypes. Adenosine A(3) receptor-selective agonist 2-chloro-N(6)-(3-iodobenzyl)adenosine-5'-N-methyluronamide (Cl-IBMECA; 0.07 mg/kg ip) reduced skeletal muscle injury with a significant decrease in both Evans blue dye staining (5.4 +/- 2.6%, n = 8 mice vs. vehicle-treated 28 +/- 6%, n = 7 mice, P < 0.05) and serum creatine kinase level (1,840 +/- 910 U/l, n = 13 vs. vehicle-treated 12,600 +/- 3,300 U/l, n = 14, P < 0.05), an effect that was selectively blocked by an A(3) receptor antagonist 3-ethyl-5-benzyl-2-methyl-6-phenyl-4-phenylethynyl-1,4-(+/-)-dihydropyridine-3,5-dicarboxylate (MRS-1191; 0.05 mg/kg). The adenosine A(1) receptor agonist 2-chloro-N(6)-cyclopentyladenosine (CCPA; 0.05 mg/kg) also exerted a cytoprotective effect, which was selectively blocked by the A(1) antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX; 0.2 mg/kg). The adenosine A(2A) receptor agonist 2-p-(2-carboxyethyl)phenethylamino-5'-N-ethylcarboxamidoadenosine (CGS-21680; 0.07 mg/kg)-induced decrease in skeletal muscle injury was selectively blocked by the A(2A) antagonist 2-(2-furanyl)-7-[3-(4-methoxyphenyl)propyl]-7H-pyrazolo[4,3-e] [1,2,4]triazolo[1,5-C]pyrimidin-5-amine (SCH-442416; 0.017 mg/kg). The protection induced by the A(3) receptor was abrogated in phospholipase C-beta2/beta3 null mice, but the protection mediated by the A(1) or A(2A) receptor remained unaffected in these animals. The adenosine A(3) receptor is a novel cytoprotective receptor that signals selectively via phospholipase C-beta and represents a new target for ameliorating skeletal muscle injury. Topics: Adenosine; Animals; Dihydropyridines; Disease Models, Animal; Hindlimb; Mice; Mice, Inbred C57BL; Mice, Knockout; Muscle, Skeletal; Phenethylamines; Phospholipase C beta; Pyrazoles; Pyrimidines; Receptor, Adenosine A1; Receptor, Adenosine A2A; Receptor, Adenosine A3; Reperfusion Injury; Signal Transduction; Xanthines | 2007 |
A2a receptors mediate inhibitory effects of adenosine on colonic motility in the presence of experimental colitis.
Adenosine regulates immunity and inflammation, and acts also as a modulator of gut functions. In this study, we investigated the role of adenosine A2a receptors on colonic motility in a rat model of experimental colitis.. Colitis was induced by 2,4-dinitrobenzenesulfonic acid. The effects of ZM 241385 (A2a receptor antagonist) and CGS 21680 (A2a receptor agonist) were assayed on cholinergic contractions of colonic longitudinal muscle preparations evoked by transmural electrical stimulation (TES) or carbachol. A2a receptor expression in colonic neuromuscular layers was assessed by reverse transcription-polymerase chain reaction.. ZM 241385 increased TES-induced contractions in the absence or in the presence of colitis, the drug being more effective in colonic preparations from inflamed animals. The enhancing effects of ZM 241385 were unaffected by guanethidine or alpha-chimotrypsin, whereas being prevented by Nomega-propyl-L-arginine (neuronal nitric oxide synthase inhibitor) or adenosine 5'-(alpha,beta-methylene) diphosphate (ecto-5'-nucleotidase inhibitor). Upon exposure of colonic tissues from normal or inflamed rats to dipyridamole plus adenosine deaminase, to abate endogenous adenosine levels, CGS 21680 evoked concentration-dependent reductions of contractile responses to TES, which were more intense in preparations from inflamed rats, and were antagonized by ZM 241385. Neither CGS 21680 nor ZM 241385 affected carbachol-induced contractions. Reverse transcription-polymerase chain reaction showed an increase in A2a receptor expression in colonic tissues isolated from inflamed animals.. The adenosine system is involved in neuroplastic changes occurring in inflamed gut. A2a receptors modulate the activity of colonic excitatory cholinergic nerves via facilitatory control on inhibitory nitrergic pathways, and such a regulatory function is enhanced in the presence of bowel inflammation. Topics: Adenosine; Animals; Colitis; Disease Models, Animal; Gastrointestinal Motility; Intestinal Mucosa; Male; Muscle Contraction; Muscle, Smooth; Nitric Oxide; Phenethylamines; Rats; Rats, Sprague-Dawley; Receptor, Adenosine A2A; Reverse Transcriptase Polymerase Chain Reaction; Sensitivity and Specificity; Tissue Culture Techniques; Triazines; Triazoles | 2006 |
Experimental studies and theoretical aspects on A2A/D2 receptor interactions in a model of Parkinson's disease. Relevance for L-dopa induced dyskinesias.
Dual probe microdialysis was used to study A2A/D2 receptor interactions in the striato-pallidal GABA pathway in a model of Parkinson's Disease. The A2A agonist CGS21680 and/or the D2-like agonist quinpirole were perfused via reverse microdialysis into the DA denervated striatum and the effects on globus pallidus (GP) extracellular GABA levels were evaluated. CGS21680 alone produced in the DA denervated striatum a transient rise of GP GABA levels. Quinpirole perfused alone into the DA denervated striatum reduced GP GABA levels, which was not only counteracted by coperfused CGS21680, but led to an enhancement of the GABA levels, which was larger than that seen with CGS21680 alone. These results may reflect existence not only of antagonistic A2A/D2 interactions but also of the appearance of D2/A2A interactions increasing the A2A signaling at the level of the adenylate cyclase. Such actions diminish the therapeutic efficacy of L-dopa and D2 agonists. L-dopa induced dyskinesias could be caused by changes in the balance of A2A/D2 heteromers vs A2A homomers expressed at the surface membrane, where A2A homomers dominate with abnormal increases in A2A signaling. This may lead to stabilization of abnormal receptor mosaics (high order hetero-oligomers) leading to formation of abnormal motor programs contributing to dyskinesia development. Topics: Adenosine; Adenosine A2 Receptor Antagonists; Adrenergic Agents; Animals; Chromatography, High Pressure Liquid; Disease Models, Animal; Dopamine Agonists; Dose-Response Relationship, Drug; Drug Interactions; Electrochemistry; gamma-Aminobutyric Acid; Globus Pallidus; Male; Microdialysis; Models, Biological; Oxidopamine; Parkinson Disease; Phenethylamines; Quinpirole; Rats; Rats, Sprague-Dawley; Receptor, Adenosine A2A; Receptors, Dopamine D2 | 2006 |
Adenosine is upregulated during peritonitis and is involved in downregulation of inflammation.
Loss of function of the peritoneal membrane is associated with peritonitis. Adenosine levels in sites of inflammation were shown to increase and exhibit immunoregulatory effects. Our aim was to elucidate the regulatory role of adenosine during peritonitis and to test the involvement of peritoneal mesothelial cells (PMC) in adenosine regulation. In a mice model of Escherichia coli peritonitis, the adenosine A(2A) receptor (A(2A)R) agonist (CGS21680) prevented leukocyte recruitment and reduced tumor necrosis factor alpha (TNF-alpha) and interleukin-6 (IL-6) levels. Peritonitis induced the elevation of adenosine with a peak at 24 h. Analysis of adenosine receptor levels on peritoneum showed that A(1) receptor (A(1)R) protein levels peak at 12 h after inoculation and then return to baseline at 24 h, whereas high affinity A(2A)R protein levels peak at 24 h concomitantly with the peak of adenosine concentration. Low affinity A(2B) receptor (A(2B)R) levels elevated slowly, remaining elevated up to 48 h. In human PMC (HPMC), the early cytokines, IL-1-alpha, and TNF-alpha upregulated the A(2B) and A(2A) receptors. However, interferon-gamma (IFN-gamma) upregulated the A(2B)R and decreased A(2A)R levels. Treatment with the A(2A)R agonist reduced IL-1-dependent IL-6 secretion from HPMC. In conclusion, the kinetics of adenosine receptors suggest that at early stage of peritonitis, the A(1)R dominates, and later its dominance is replaced by the G stimulatory (Gs) protein-coupled A(2A)R that suppresses inflammation. Early proinflammatory cytokines are an inducer of the A(2A)R and this receptor reduces their production and leukocyte recruitment. Future treatment with adenosine agonists should be considered for attenuating the damage to mesothelium during the course of acute peritonitis. Topics: Adenosine; Adenosine A1 Receptor Agonists; Adenosine A2 Receptor Agonists; Adenosine A2 Receptor Antagonists; Animals; Antihypertensive Agents; Cells, Cultured; Disease Models, Animal; Down-Regulation; Epithelium; Escherichia coli; Female; Humans; Inflammation; Interleukin-6; Leukocytes; Mice; Mice, Inbred Strains; Peritonitis; Phenethylamines; Purinergic P1 Receptor Agonists; Purinergic P1 Receptor Antagonists; Receptor, Adenosine A1; Receptor, Adenosine A2A; Receptors, Purinergic P1; RNA, Messenger; Theobromine; Tumor Necrosis Factor-alpha; Up-Regulation; Xanthines | 2006 |
The impact of adenosine and A(2B) receptors on glucose homoeostasis.
Adenosine and adenosine receptor antagonists are involved in glucose homoeostasis. The participating receptors are not known, mainly due to a lack of specific agonists and antagonists, but are reasonable targets for anti-diabetic therapy. The stable, albeit nonselective, adenosine analogue NECA (5'-N-ethylcarboxamidoadenosine) (10 microM) reduced glucose-stimulated insulin release from INS-1 cells. This was mimicked by A(1)-(CHA), A(2A)-(CGS-21680) and A(3)-receptor agonists (Cl-IB-MECA). Two newly synthesized A(2B)-receptor antagonists, PSB-53 and PSB-1115, counteracted the inhibitory effect of NECA. These in-vitro effects were mirrored by in-vivo data with respect to CHA, CGS and Cl-IB-MECA. Distinct concentrations of either PSB-53 or PSB-1115 reversed the decrease in plasma insulin induced by NECA. This was not mimicked by a corresponding change in blood glucose. The effect of PSB-1115 was also obvious in diabetic GotoKakizaki rats: plasma insulin was increased whereas blood glucose was unchanged. During most experiments the effects on blood glucose were not impressive probably because of the physiologically necessary homoeostasis. The adenosine levels were not different in normal Wistar rats and in diabetic GotoKakzaki rats. Altogether the A(2B)-receptor antagonists showed an anti-diabetic potential mainly by increasing plasma insulin levels under conditions when the adenosine tonus was elevated in-vivo and increased insulin release in-vitro. Topics: Adenosine; Adenosine A2 Receptor Agonists; Adenosine A2 Receptor Antagonists; Adenosine A3 Receptor Agonists; Adenosine A3 Receptor Antagonists; Adenosine-5'-(N-ethylcarboxamide); Analysis of Variance; Animals; Antineoplastic Agents; Blood Glucose; Cell Line, Tumor; Diabetes Mellitus, Type 2; Disease Models, Animal; Dose-Response Relationship, Drug; Female; Homeostasis; Insulin; Male; Phenethylamines; Radioimmunoassay; Rats; Rats, Wistar; Receptor, Adenosine A2B; Sulfonic Acids; Xanthines | 2006 |
The role of adenosine A2a receptors in experimental acute pancreatitis.
The role of adenosine and its receptors in acute pancreatitis remains unelucidated. The aim was to evaluate the effects of the adenosine A2a receptor agonist and antagonist in the severe, taurocholate-induced experimental acute pancreatitis (EAP).. The experiments were performed on 80 male Wistar rats, subdivided into 4 groups: C--the control rats, I--the EAP group, IIA--EAP group treated with the A2a adenosine receptor agonist CGS 21680, IIB--EAP group treated with the A2a adenosine receptor antagonist ZM 241385. The blood for alpha-amylase and lipase and tissues samples for the morphological examinations and immunohistochemistry for A2a receptors were collected in 2, 6, 24 hours of the experiment.. The serum alpha-amylase tended to decrease in the group IIA as compared to EAP untreated after 6 and 24 h. No significant effect of both treatments on serum lipase was noted. The administration of CGS 21680 resulted in favorable decrease of the inflammatory cell infiltration, hemorrhagic changes, necrosis and vacuolization of acinar cells, without an evident effect on the edema of the interstitial tissue. The administration of ZM 241385 did not affect the scores of necro-hemorrhagic changes and inflammatory infiltration, whereas it decreased the scores of vacuolization and edema. In all groups the expression of A2a receptors was similar.. Our findings suggest, that A2a adenosine receptors are involved in the course of sodium taurocholate EAP. It is probable that the modulation of some subgroups of adenosine receptors could alleviate the course of severe experimental AP. Topics: Acute Disease; Adenosine; Adenosine A2 Receptor Agonists; Adenosine A2 Receptor Antagonists; Amylases; Animals; Disease Models, Animal; Immunohistochemistry; Lipase; Male; Pancreas; Pancreatitis; Phenethylamines; Rats; Rats, Wistar; Receptor, Adenosine A2A; Taurocholic Acid; Triazines; Triazoles | 2006 |
A novel A1 adenosine receptor antagonist, L-97-1 [3-[2-(4-aminophenyl)-ethyl]-8-benzyl-7-{2-ethyl-(2-hydroxy-ethyl)-amino]-ethyl}-1-propyl-3,7-dihydro-purine-2,6-dione], reduces allergic responses to house dust mite in an allergic rabbit model of asthma.
Adenosine, an important signaling molecule in asthma, produces bronchoconstriction in asthmatics. Adenosine produces bronchoconstriction in allergic rabbits, primates, and humans by activating A1 adenosine receptors (ARs). Effects of L-97-1 [3-[2-(4-aminophenyl)-ethyl]-8-benzyl-7-{2-ethyl-(2-hydroxyethyl)-amino]-ethyl}-1-propyl-3,7-dihydro-purine-2,6-dione] a water-soluble, small molecule A1 AR antagonist were investigated on early and late phase allergic responses (EAR and LAR) in a hyper-responsive rabbit model of asthma. Rabbits were made allergic by intraperitoneal injections of house dust mite [HDM; 312 allergen units (AU)] extract within 24 h of their birth. Booster HDM injections were given weekly for 1 month, biweekly for 4 months, and continued monthly thereafter. Hyperresponsiveness was monitored by measuring lung dynamic compliance (Cdyn), after histamine or adenosine aerosol challenge in allergic rabbits. Hyper-responsive rabbits were subjected to aerosol of HDM (2500 AU), 1 h after intragastric administration of L-97-1 (10 mg/kg) solution or an equivalent volume of saline. Cdyn was significantly higher after treatment with L-97-1 compared with untreated controls (p < 0.05 n = 5). Histamine PC30 was significantly higher (p < 0.05; n = 5) after L-97-1 at 24 h compared with histamine PC30 at 24 h after HDM. Adenosine PC30 was significantly higher at 15 min and 6 h after L-97-1 compared with control (p < 0.05; n = 5). L-97-1 showed strong affinity for human A1 ARs in radioligand binding studies and no inhibition toward human phosphodiesterase II, III, IV, and V enzymes. These data suggest that L-97-1 produces a significant reduction of histamine or adenosine-induced hyper-responsiveness and HDM-induced EAR and LAR in allergic rabbits by blocking A1 ARs and may be beneficial as an oral therapy for human asthma. Topics: Adenosine; Adenosine A1 Receptor Antagonists; Animals; Anti-Asthmatic Agents; Asthma; Bronchial Hyperreactivity; Disease Models, Animal; Dose-Response Relationship, Drug; Dust; Histamine; Mites; Phosphodiesterase Inhibitors; Purines; Rabbits; Radioligand Assay; Receptor, Adenosine A1 | 2005 |
CGS21680 attenuates symptoms of Huntington's disease in a transgenic mouse model.
Huntington's disease (HD) is an autosomal dominant neurodegenerative disease caused by a CAG trinucleotide expansion in exon 1 of the Huntingtin (Htt) gene. We show herein that in an HD transgenic mouse model (R6/2), daily administration of CGS21680 (CGS), an A(2A) adenosine receptor (A(2A)-R)-selective agonist, delayed the progressive deterioration of motor performance and prevented a reduction in brain weight. 3D-microMRI analysis revealed that CGS reversed the enlarged ventricle-to-brain ratio of R6/2 mice, with particular improvements in the left and right ventricles. (1)H-MRS showed that CGS significantly reduced the increased choline levels in the striatum. Immunohistochemical analyses further demonstrated that CGS reduced the size of ubiquitin-positive neuronal intranuclear inclusions (NIIs) in the striatum of R6/2 mice and ameliorated mutant Htt aggregation in a striatal progenitor cell line overexpressing mutant Htt with expanded polyQ. Moreover, chronic CGS treatment normalized the elevated blood glucose levels and reduced the overactivation of a major metabolic sensor [5'AMP-activated protein kinase (AMPK)] in the striatum of R6/2 mice. Since AMPK is a master switch for energy metabolism, modulation of energy dysfunction caused by the mutant Htt might contribute to the beneficial effects of CGS. Collectively, CGS is a potential drug candidate for the treatment of HD. Topics: Adenosine; Adenosine A2 Receptor Agonists; Animals; Brain; Disease Models, Animal; Huntington Disease; Male; Mice; Mice, Transgenic; Motor Activity; Phenethylamines | 2005 |
Stimulation of adenosine A2A receptors elicits zif/268 and NMDA epsilon2 subunit mRNA expression in cortex and striatum of the "weaver" mutant mouse, a genetic model of nigrostriatal dopamine deficiency.
Interaction between basal ganglia and cerebral cortex is critical for normal goal-directed behavior. In the present study we have used the immediate early gene zif/268, as functional marker to investigate how the stimulation of adenosine A2A receptors, i.e. of the "indirect" striatal output pathway, affects striatal and cortical function in "weaver" mouse, a genetic model of dopamine deficiency. Furthermore, we have examined the effect of A2A receptor stimulation on glutamate receptor expression in the "weaver" brain. A single injection of CGS21680 (A2A receptor agonist), induced strong expression of zif/268 mRNA, detected by in situ hybridization, not only in striatum but also in the motor cortex of the "weaver" mutant. This cortical response seems to be elicited through the basal-ganglia-thalamo-cortical circuit, rather than through a direct cortical effect, since A2A receptors are not detectable in cortex according to our autoradiographic study. Co-administration of CGS21680 and quinpirole (D2 receptor agonist) attenuated the expression of zif/268 mRNA in dorsal striatum but not in motor cortex, indicating that the cortical response is dopamine-D2-receptor-independent. However, this co-administration induced an increase in zif/268 mRNA expression in somatosensory cortex, which could rely on disinhibition of the thalamo-cortical pathway. The motor cortical response could be of clinical interest, as it would further stimulate the "indirect" striatal pathway in a feed forward circuit, thus worsening the parkinsonian symptoms. Furthermore, the up-regulation of epsilon2 subunit mRNA of the NMDA receptor, induced by CGS21680 administration, seen in striatum and cortex of the "weaver" mouse, would lead to overactivity of these receptors worsening dyskinesias. These results suggest adenosine to play a significant role in regulating striatal and cortical neurochemistry in a dopamine-depleted mouse. Blockade of these receptors by specific A2A antagonists could ameliorate parkinsonian symptoms. Topics: Adenosine; Adenosine A2 Receptor Antagonists; Animals; Autoradiography; Binding Sites; Cell Count; Cerebral Cortex; Corpus Striatum; Disease Models, Animal; DNA-Binding Proteins; Dopamine; Dopamine Agonists; Dopamine Antagonists; Early Growth Response Protein 1; Gene Expression; Immediate-Early Proteins; In Situ Hybridization; Mice; Mice, Neurologic Mutants; Phenethylamines; Protein Subunits; Quinpirole; Raclopride; Receptor, Adenosine A2B; Receptors, AMPA; Receptors, N-Methyl-D-Aspartate; RNA, Messenger; Staining and Labeling; Transcription Factors | 2004 |
Protection against cisplatin ototoxicity by adenosine agonists.
Cisplatin is a commonly used antineoplastic agent that causes ototoxicity through the formation of reactive oxygen species (ROS). Previous studies have shown that cisplatin causes an upregulation of A(1) adenosine receptor (A(1)AR) in the cochlea, and that application of the adenosine agonist, R-phenylisopropyladenosine (R-PIA), to the round window (RW) results in significant increases in cochlear glutathione peroxidase and superoxide dismutase. These data suggest that adenosine receptors (ARs) are an important part of the cytoprotective system of the cochlea in response to oxidative stress. The purpose of the current study was to investigate the effect of various adenosine agonists on cisplatin ototoxicity using RW application. Auditory brainstem response (ABR) thresholds were recorded in anesthetized chinchillas at 1, 2, 4, 8 and 16kHz. The auditory bullae were surgically opened, and 1mM R-PIA, 10microM 8-cyclopentyl-1,3-dipropylxanthine (DPCPX)/R-PIA (1mM) cocktail, 100microM 2-chloro-N-cyclopentyladenosine (CCPA), 2-[4-(2-p-carboxy-ethyl)phenylamino]-5'-N-ethylcarboxamidoadenosine (CGS) or vehicle were applied to the RW. After 90min, the remaining solution was removed and cisplatin was applied to the RW. The bullae were closed and the animals recovered for 72h, after which, follow-up ABRs were performed. Cochleae were harvested for scanning electron microscopy (SEM) and for lipid peroxides. Pre-administration of the A(1)AR agonists R-PIA or CCPA significantly reduced cisplatin-induced threshold changes at all but the highest test frequency. In addition, A(1)AR agonists protected against cisplatin-induced hair cell damage and significantly reduced cisplatin-induced lipid peroxidation. Co-administration of the A(1)AR antagonist, DPCPX, completely reversed the protective effects of R-PIA. In contrast, pretreatment with CGS-21680, an A(2A) adenosine receptor (A(2A)AR) agonist, significantly increased cisplatin-induced threshold changes. Our findings are consistent with the notion that the A(1)AR contributes significantly to cytoprotection in the cochlea, and thereby protects against hearing loss. Topics: Adenosine; Animals; Antineoplastic Agents; Chinchilla; Cisplatin; Cochlea; Disease Models, Animal; Hearing Disorders; Lipid Peroxidation; Male; Neuroprotective Agents; Phenethylamines; Receptors, Purinergic P1 | 2004 |
Neuroprotective effect of L-DOPA co-administered with the adenosine A2A receptor agonist CGS 21680 in an animal model of Parkinson's disease.
Adenosine A2A receptors are a new target for drug development in Parkinson's disease. Some experimental and clinical data suggest that A2A receptor antagonists can provide symptomatic improvement by potentiating the effects of L-DOPA as well as a decrease in secondary effects such as L-DOPA-induced dyskinesia. L-DOPA-induced behavioral sensitization in unilateral 6-hydroxydopamine-lesioned rats is frequently used as an experimental model of L-DOPA-induced dyskinesia. In the present work this model was used to evaluate the effect of the A2A receptor agonist CGS 21680 and the A2A receptor antagonist MSX-3 on L-DOPA-induced behavioral sensitization and 6-hydroxydopamine-induced striatal dopamine denervation. L-DOPA-induced behavioral sensitization was determined as an increase in L-DOPA-induced abnormal involuntary movements and enhancement of apomorphine-induced turning behavior. Striatal dopamine innervation was determined by measuring tyrosine-hydroxylase immunoreactivity. Chronic administration of MSX-3 was not found to be effective at counteracting L-DOPA-induced behavioral sensitization. On the other hand, CGS 21680 completely avoided the development of L-DOPA-induced behavioral sensitization. The analysis of the striatal dopamine innervation showed that L-DOPA-CGS 21680 co-treatment conferred neuroprotection to the toxic effects of 6-hydroxydopamine. This neuroprotective effect was dependent on A2A and D2 receptor stimulation, since it was counteracted by MSX-3 and by the D2 receptor antagonist haloperidol. These results open new therapeutic avenues in early events in Parkinson's disease. Topics: Adenosine; Adenosine A2 Receptor Agonists; Analysis of Variance; Animals; Antiparkinson Agents; Apomorphine; Cell Count; Corpus Striatum; Disease Models, Animal; Dopamine Agonists; Drug Therapy, Combination; Dyskinesias; Immunohistochemistry; Levodopa; Male; Oxidopamine; Parkinson Disease; Phenethylamines; Rats; Rats, Sprague-Dawley; Stereotyped Behavior; Sympatholytics; Time Factors; Tyrosine 3-Monooxygenase | 2004 |
Characterization of adenosine receptor(s) involved in adenosine-induced bronchoconstriction in an allergic mouse model.
We recently reported that adenosine caused bronchoconstriction and enhanced airway inflammation in an allergic mouse model. In this study, we further report the characterization of the subtype of adenosine receptor(s) involved in bronchoconstriction. 5'-(N-ethylcarboxamido)adenosine (NECA), a nonselective adenosine agonist, elicited bronchoconstriction in a dose-dependent manner. Little effects of N(6)-cyclopentyladenosine (A(1)-selective agonist) and 2-p-(2-carboxyethyl)phenethylamino-5'-N-ethylcarboxamidoadenosine (A(2A)-selective agonist) compared with NECA were observed in this model. 2-Chloro-N(6)-(3-iodobenzyl)-9-[5-(methylcarbamoyl)-beta-d-ribofuranosyl]adenosine, an A(3)-selective receptor agonist, produced a dose-dependent bronchoconstrictor response, which was blocked by selective A(3) antagonist 2,3-diethyl-4,5-dipropyl-6-phenylpyridine-3-thiocarboxylate-5-carboxylate (MRS1523). However, MRS1523 only partially inhibited NECA-induced bronchoconstriction. Neither selective A(1) nor A(2A) antagonists affected NECA-induced bronchoconstriction. Enprofylline, a relatively selective A(2B) receptor antagonist, blocked partly NECA-induced bronchoconstriction. Furthermore, a combination of enprofylline and MRS1523 completely abolished NECA-induced bronchoconstrictor response. Using RT-PCR, we found that all four adenosine receptor subtypes are expressed in control lungs. Allergen sensitization and challenge significantly increased transcript levels of the A(2B) and A(3) receptors, whereas the A(1) receptor message decreased. No change in transcript levels of A(2A) receptors was observed after allergen sensitization and challenge. These findings suggest that A(2B) and A(3) adenosine receptors play an important role in adenosine-induced bronchoconstriction in our allergic mouse model. Finally, whether the airway effects of the receptor agonists/antagonists are direct or indirect needs further investigations. Topics: Adenosine; Adenosine-5'-(N-ethylcarboxamide); Adrenergic Agonists; Adrenergic Antagonists; Animals; Asthma; Bronchoconstriction; Disease Models, Animal; Hypersensitivity; Lung; Male; Mice; Mice, Inbred BALB C; Phenethylamines; Pyridines; Receptors, Purinergic P1; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Vasodilator Agents | 2003 |
A dual role of adenosine A2A receptors in 3-nitropropionic acid-induced striatal lesions: implications for the neuroprotective potential of A2A antagonists.
Reduction of A2A receptor expression is one of the earliest events occurring in both Huntington's disease (HD) patients and mice overexpressing the N-terminal part of mutated huntingtin. Interestingly, increased activity of A2A receptors has been found in striatal cells prone to degenerate in experimental models of this neurodegenerative disease. However, the role of A2A receptors in the pathogenesis of HD remains obscure. In the present study, using A2A-/- mice and pharmacological compounds in rat, we demonstrate that striatal neurodegeneration induced by the mitochondrial toxin 3-nitropropionic acid (3NP) is regulated by A2A receptors. Our results show that the striatal outcome induced by 3NP depends on a balance between the deleterious activity of presynaptic A2A receptors and the protective activity of postsynaptic A2A receptors. Moreover, microdialysis data demonstrate that this balance is anatomically determined, because the A2A presynaptic control on striatal glutamate release is absent within the posterior striatum. Therefore, because blockade of A2A receptors has differential effects on striatal cell death in vivo depending on its ability to modulate presynaptic over postsynaptic receptor activity, therapeutic use of A2A antagonists in Huntington's as well as in other neurodegenerative diseases could exhibit undesirable biphasic neuroprotective-neurotoxic effects. Topics: Adenosine; Animals; Body Weight; Cell Death; Corpus Striatum; Disease Models, Animal; Drug Administration Schedule; Encephalitis; Genetic Predisposition to Disease; Glutamic Acid; Huntington Disease; Male; Mice; Mice, Knockout; Neuroprotective Agents; Nitro Compounds; Phenethylamines; Propionates; Rats; Rats, Inbred Lew; Rats, Wistar; Receptor, Adenosine A2A; Receptors, Purinergic P1; RNA, Messenger; Signal Transduction; Survival Rate; Synapses; Xanthines | 2003 |
Discovery of nonxanthine adenosine A2A receptor antagonists for the treatment of Parkinson's disease.
During a program to investigate the biochemical basis of side effects associated with the antimalarial drug mefloquine, the authors made the unexpected discovery that the (-)-(R,S)-enantiomer of the drug is a potent adenosine A2A receptor antagonist. Although the compound was ineffective in in vivo animal models of central adenosine receptor function, it provided a unique nonxanthine adenosine A2A receptor antagonist lead structure and encouraged the initiation of a medicinal chemistry program to develop novel adenosine A2A antagonists for the management of Parkinson's disease (PD). The authors have synthesized and screened more than 2,000 chemically diverse and novel adenosine A(2A antagonists. Early examples from two distinct chemical series are the thieno[3,2-dy]pyrimidine VER-6623 and the purine compounds VER-6947 and VER-7835, which have high affinity at adenosine A2A receptors (K(i) values 1.4, 1.1, and 1.7 nmol/L, respectively) and act as competitive antagonists. In particular, VER-6947 and VER-7835 demonstrate potent in vivo activity reversing the locomotor deficit caused by the D2 receptor antagonist haloperidol, with minimum effective doses comparable with that of KW6002 (0.3 to 1 mg/kg). In conclusion, the authors have discovered potent, selective, and in vivo active nonxanthine adenosine A2A antagonists that have considerable promise as a new therapy for PD. Topics: Adenosine; Adenosine A2 Receptor Antagonists; Animals; Antiparkinson Agents; Binding, Competitive; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Design; Drug Evaluation, Preclinical; Humans; Ligands; Mefloquine; Mice; Motor Activity; Neuroprotective Agents; Parkinsonian Disorders; Phenethylamines; Purines; Pyrimidines; Radioligand Assay; Rats; Triazines; Triazoles | 2003 |
[Role of G-protein-coupled adenosine receptors in downregulation of inflammation and protection from tissue damage].
Topics: Adenosine; Animals; Concanavalin A; Disease Models, Animal; Hepatitis, Animal; Hepatitis, Autoimmune; Liver; Mice; Phenethylamines; Purinergic P1 Receptor Agonists | 2002 |
Effects of adenosine receptor agonists and antagonists in a genetic animal model of primary paroxysmal dystonia.
1. Recent studies have shown beneficial effects of an adenosine A(2A) receptor agonist in dt(sz) mutant hamsters, an animal model of paroxysmal dystonia, in which stress and consumption of coffee can precipitate dystonic attacks. This prompted us to examine the effects of adenosine receptor agonists and antagonists on severity of dystonia in dt(sz) hamsters in more detail. 2. The non-selective adenosine A(1)/A(2A) receptor antagonists, caffeine (10 - 20 mg kg(-1) i.p.) and theophylline (10 - 30 mg kg(-1) s.c.), worsened the dystonia in dt(sz) hamsters. 3. Aggravation of dystonia was also caused by the selective adenosine A(1)/A(2A) antagonist CGS 15943 (9-chloro2-2-furyl)[1,2,4]triazolo[1,5-c]quinazolin-5-amine) at a dose of 30 mg kg(-1) i.p. and by the adenosine A(1) antagonist DPCPX (8-cyclopentyl-1,3-dipropylxanthine; 20 - 30 mg kg(-1) i.p.), while the A(2) antagonist DMPX (3,7-dimethyl-1-propargylxanthine; 2 - 4 mg kg(-1) i.p.) and the highly selective A(2A) antagonist ZM 241385 (4-(2-[7-amino-2-(2-furyl)[1,2,4]triazolo[2,3-a][1,3,5]triazin-5-ylamino]ethyl)phenol; 2 - 5 mg kg(-1) i.p.) failed to exert any effects on dystonia. 4. In contrast to the antagonists, both the adenosine A(1) receptor agonist CPA (N(6)-cyclopentyladenosine; 0.1 - 1.0 mg kg(-1) i.p.) and the A(2A) agonist CGS 21680 (2p-(2carboxyethylphen-ethylamino-5'-N-ethylcarboxamindoadenosine; 0.1 - 2.0 mg kg(-1) i.p.) exerted a striking improvement of dystonia. 5. These data suggest that the precipitating effects of methylxanthines are, at least in part, related to their adenosine receptor antagonistic action. 6. Although adenosine receptor agonists can be regarded as interesting candidates for the therapy of paroxysmal dystonia, adverse effects may limit the therapeutic potential of adenosine A(1) agonists, while beneficial effects of the adenosine A(2A) agonist CGS 21680 were already found at well tolerated doses. Topics: Adenosine; Animals; Caffeine; Cricetinae; Disease Models, Animal; Dose-Response Relationship, Drug; Dystonia; Female; Male; Mesocricetus; Muscle Contraction; Mutation; Phenethylamines; Purinergic P1 Receptor Agonists; Purinergic P1 Receptor Antagonists; Quinazolines; Theobromine; Theophylline; Triazines; Triazoles; Xanthines | 2001 |
Adenosine A2A, 5-HT1A and 5-HT7 receptor in neonatally pregnenolone-treated rats.
Steroid hormones synthesized in the brain, called 'neurosteroids', modulate neuronal activity. We treated neonatal rats with a main precursor of the neurosteroidogenesis, pregnenolone, and examined adenosine A2A receptor, 5- hydroxytryptamine (5-HT)1A and 5-HT7 receptor densities in the front-parietal cortex in juvenile and adult rats. In receptor binding assay using [3H]CGS21680 and [3H]8-OH-DPAT, it was shown that neonatal pregnenolone-treatment induced a significant decrease in the adenosine A2A receptor density with no significant effects on the 5-HT1A and 5-HT7 receptor densities. Topics: 8-Hydroxy-2-(di-n-propylamino)tetralin; Adenosine; Aging; Animals; Animals, Newborn; Antihypertensive Agents; Binding, Competitive; Cerebral Cortex; Disease Models, Animal; Dopamine; Female; Male; Mental Disorders; Phenethylamines; Pregnancy; Pregnenolone; Radioligand Assay; Rats; Rats, Sprague-Dawley; Receptor, Adenosine A2A; Receptors, Purinergic P1; Receptors, Serotonin; Receptors, Serotonin, 5-HT1; Serotonin; Serotonin Receptor Agonists; Sex Characteristics; Synaptic Transmission | 2001 |
CGS 21680 exerts marked antidystonic effects in a genetic model of paroxysmal dyskinesia.
The effect of the adenosine A(2A) receptor agonist CGS 21680 (2-carboxyethyl)phenylethylamino]-5'-N-ethylcarbonyamido-ade nosine) on severity of dystonia was examined in genetically dystonic hamsters which exhibit attacks of dystonic and choreoathetotic disturbances in response to mild stress. CGS 21680 significantly reduced the severity of dystonia (0.5, 1.0 and 2.0 mg/kg i.p.). The marked antidystonic effects of CGS 21680 in the hamster model suggest that this compound may represent an interesting candidate for the therapy of paroxysmal dystonia. Furthermore, the present data indicate that the precipitating effect of caffeine in patients with paroxysmal dystonia is probably due to its adenosine receptor antagonistic action. Topics: Adenosine; Animals; Chorea; Cricetinae; Disease Models, Animal; Drug Evaluation, Preclinical; Dystonia; Models, Genetic; Phenethylamines; Purinergic P1 Receptor Agonists; Receptors, Purinergic P1; Stress, Psychological | 2000 |
Electrophysiological and behavioural evidence for an antagonistic modulatory role of adenosine A2A receptors in dopamine D2 receptor regulation in the rat dopamine-denervated striatum.
It has been shown that striatal adenosine A2A receptors can antagonistically interact with dopamine D2 receptors at the membrane level leading to a decrease in the affinity and efficacy of D2 receptors. Extracellular recordings and rotational behaviour were employed to obtain a correlate to these findings in an animal model of Parkinson's disease (PD). The recordings were performed in rats with unilateral 6-hydroxydopamine (6-OHDA)-induced catecholamine depletion. While recording in the dopamine-depleted striatum, local applications of the dopamine D2 agonist quinpirole reduced neuronal activity. However, when the adenosine A2A antagonist MSX-3 was applied simultaneously with quinpirole, the inhibition of neuronal firing seen after quinpirole alone was significantly potentiated (P< 0.001, n = 11). In contrast, local application of CGS 21680 attenuated the effect of quinpirole. The doses of MSX-3 and CGS 21680 used to achieve the modulation of quinpirole action had no effect per se on striatal neuronal firing. Furthermore, rotational behaviour revealed that MSX-3 dose-dependently increased the number of turns when administrated together with a threshold dose of quinpirole while no enhancement was achieved when MSX-3 was combined with SKF 38393. MSX-3 alone did not induce rotational behaviour. In conclusion, this study shows that low ineffective doses of MSX-3 enhance the effect of quinpirole on striatal firing rate, while the A2A agonist exerts the opposite action. This mechanism gives a therapeutic potential to A2A antagonists in the treatment of PD by enhancing D2 receptor function. Topics: Adenosine; Animals; Corpus Striatum; Denervation; Disease Models, Animal; Dopamine; Functional Laterality; Humans; Male; Oxidopamine; Parkinson Disease; Phenethylamines; Purinergic P1 Receptor Antagonists; Quinpirole; Rats; Rats, Sprague-Dawley; Receptor, Adenosine A2A; Receptors, Dopamine D2; Receptors, Purinergic P1; Reference Values | 2000 |
Antinociception by adenosine analogs and inhibitors of adenosine metabolism in an inflammatory thermal hyperalgesia model in the rat.
The present study examined the spinal antinociceptive effects of adenosine analogs and inhibitors of adenosine kinase and adenosine deaminase in the carrageenan-induced thermal hyperalgesia model in the rat. The possible enhancement of the antinociceptive effects of adenosine kinase inhibitors by an adenosine deaminase inhibitor also was investigated. Unilateral hindpaw inflammation was induced by an intraplantar injection of lambda carrageenan (2 mg/100 microl), which consistently produced significant paw swelling and thermal hyperalgesia. Drugs were administered intrathecally, either by acute percutaneous lumbar puncture (individual agents and combinations) or via an intrathecal catheter surgically implanted 7-10 days prior to drug testing (antagonist experiments). N6-cyclohexyladenosine (CHA; adenosine A1 receptor agonist; 0.01-1 nmol), 2-[p-(2-carboxyethyl)phenylethylamino]-5'-N-ethylcarboxamidoadenos ine (CGS21680; adenosine A2A receptor agonist; 0.1-10 nmol), 5'-amino-5'-deoxyadenosine (NH2dAdo; adenosine kinase inhibitor: 10-300 nmol), and 5-iodotubercidin (ITU; adenosine kinase inhibitor; 0.1-100 nmol) produced, to varying extents, dose-dependent antinociception. No analgesia was seen following injection of 2'-deoxycoformycin (dCF; an adenosine deaminase inhibitor; 100-300 nmol). Reversal of drug effects by caffeine (non-selective adenosine A1/A2 receptor antagonist; 515 nmol) confirmed the involvement of the adenosine receptor, while antagonism by 8-cyclopentyl-1,3-dimethylxanthine (CPT; adenosine A1 receptor antagonist; 242 nmol), but not 3,7-dimethyl-1-propargylxanthine (DMPX; adenosine A2A receptor antagonist; 242 nmol), evidenced an adenosine A1 receptor mediated spinal antinociception by NH2dAdo. dCF (100 nmol), which was inactive by itself, enhanced the effects of 10 nmol and 30 nmol NH2dAdo. Enhancement of the antinociceptive effect of ITU by dCF was less pronounced. None of the antinociceptive drug regimens had any effect on paw swelling. These results demonstrate that both directly and indirectly acting adenosine agents, when administered spinally, produce antinociception through activation of spinal adenosine A1 receptors in an inflammatory model of thermal hyperalgesia. The spinal antinociceptive effects of selected adenosine kinase inhibitors can be significantly augmented when administered simultaneously with an adenosine deaminase inhibitor. Topics: Adenosine; Adenosine Deaminase; Animals; Antihypertensive Agents; Carrageenan; Deoxyadenosines; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Interactions; Edema; Enzyme Inhibitors; Excipients; Hot Temperature; Hyperalgesia; Male; Neuritis; Neurons; Nociceptors; Pentostatin; Phenethylamines; Purinergic P1 Receptor Antagonists; Rats; Rats, Sprague-Dawley; Tubercidin | 1998 |
Adenosine A2A agonists: a potential new type of atypical antipsychotic.
The systemic intraperitoneal (i.p.) administration of the adenosine A2A agonist CGS 21680 was found to dose-dependently antagonize spontaneous and amphetamine-induced (1 mg/kg i.p.) motor activity with similar ED50 values (about 0.2 mg/kg). The ratios between the ED50 values for induction of catalepsy and for antagonizing amphetamine-induced motor activity for CGS 21680, haloperidol, and clozapine were 12, 2, and > 30, respectively. Furthermore, CGS 21680 was comparably much stronger than haloperidol or clozapine at antagonizing the motor activity induced by phencyclidine (2 mg/kg subcutaneously) than motor activity induced by amphetamine (1 mg/kg i.p.). In conclusion, the present results show a clear "atypical" antipsychotic profile of the adenosine A2A agonist CGS 21680 in animal models. Topics: Adenosine; Animals; Antihypertensive Agents; Antipsychotic Agents; Catalepsy; Disease Models, Animal; Dose-Response Relationship, Drug; Locomotion; Male; Phenethylamines; Rats; Rats, Sprague-Dawley; Receptors, Purinergic P1 | 1997 |
Pharmacology of the spinal adenosine receptor which mediates the antiallodynic action of intrathecal adenosine agonists.
The effects of intrathecally delivered adenosine agonists on allodynia induced by L5/L6 spinal nerve ligation in rats with lumbar intrathecal catheters were examined. Tactile allodynia was assessed by measuring the threshold for evoking withdrawal of the lesioned hind paw with calibrated von Frey hairs. Intrathecal administration of the A1 adenosine selective agonist, N6-(2-phenylisopropyl)-adenosine R-(-)isomer (R-PIA), produced a dose-dependent (0.3-3 nmol; ED50 = 0.6 nmol) antiallodynic action and evoked a delayed motor weakness at a dosage of 30 nmol. Intrathecal administration of the A2 adenosine selective agonist, CGS 21680 {2-p-(2-carboxyethyl) phenethylamino-5'-N-ethylcarboxamido adenosine hydrochloride}, also produced a dose-dependent reduction in allodynia (2-40 nmol; ED50 = 15 nmol), but this effect was associated at 40 nmol after a short interval with prominent hind limb weakness. Intrathecal pretreatment with A1/A2 adenosine antagonists, caffeine (20 mumol) and 8-sulfophenyltheophylline (60 nmol), blocked antiallodynic actions of R-PIA (1 nmol) and CGS 21680 (40 nmol). Intrathecal pretreatment with the A1 adenosine-selective antagonist, 8-cyclopentyl-1,3-dimethylxanthine (3 nmol), blocked the antiallodynic effect of R-PIA (1 nmol), but even a dose as high as 10 nmol did not block the effect of CGS 21680 (40 nmol). The A2 adenosine-selective antagonist, 3, 7-dimethyl-1-propargylxanthine (3 nmol), prevented the antiallodynic effects of R-PIA (1 nmol) and CGS 21680 (40 nmol). Pretreatment with caffeine (20 mumol), 8-sulfophenyltheophylline (60 nmol) and 3,7-dimethyl-1-propargylxanthine (3 nmol) prevented the motor dysfunction induced by R-PIA (30 nmol) and CGS 21680 (40 nmol), but 8-cyclopentyl-1,3-dimethylxanthine (3 or 10 nmol) did not. Based on these effects, we hypothesize that the antiallodynic effects are mediated through the activation of spinal A1 adenosine receptors and motor dysfunction effects are mediated through A2 adenosine receptors. Topics: Adenosine; Animals; Disease Models, Animal; Dose-Response Relationship, Drug; Injections, Spinal; Male; Pain; Phenethylamines; Purinergic P1 Receptor Agonists; Rats; Rats, Sprague-Dawley; Spinal Cord | 1996 |
CGS 21680 antagonizes motor hyperactivity in a rat model of Huntington's disease.
The influence of CGS 21680 (2-[p-(2-carboxyethyl)phenethylamino]-5'-N-ethylcarboxamido-adenos ine), an adenosine A2 receptor agonist, was tested in an animal model of Huntington's disease. Male Wistar rats received bilateral intrastriatal injections of quinolinic acid and then, 1 and 2 weeks later, they were treated with intrastriatal CGS 21680 (3 micrograms/2 microliters) or saline. While quinolinic acid-lesioned rats not treated with CGS 21680 showed the typical motor hyperresponsiveness to d-amphetamine (1 mg/kg i.p.), the intrastriatal injection of CGS 21680 completely prevented this effect. Topics: Adenosine; Amphetamine; Animals; Disease Models, Animal; Huntington Disease; Male; Motor Activity; Phenethylamines; Quinolinic Acid; Rats; Rats, Wistar | 1994 |