Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Neurochem Res ; 46(5): 1081-1091, 2021 May.
Article in English | MEDLINE | ID: mdl-33616808

ABSTRACT

Propofol has shown strong addictive properties in rats and humans. Adenosine A2A receptors (A2AR) in the nucleus accumbens (NAc) modulate dopamine signal and addictive behaviors such as cocaine- and amphetamine-induced self-administration. However, whether A2AR can modulate propofol addiction remains unknown. AAV-shA2AR was intra-NAc injected 3 weeks before the propofol self-administration training to test the impacts of NAc A2AR on establishing the self-administration model with fixed ratio 1 (FR1) schedule. Thereafter, the rats were withdrawal from propofol for 14 days and tested cue-induced reinstatement of propofol seeking behavior on day 15. The propofol withdrawal rats received one of the doses of CGS21680 (A2AR agonist, 2.5-10.0 ng/site), MSX-3 (A2AR antagonist, 5.0-20.0 µg/site) or eticlopride (D2 receptor (D2R) antagonist, 0.75-3.0 µg/site) or vehicle via intra-NAc injection before relapse behavior test. The numbers of active and inactive nose-poke response were recorded. Focal knockdown A2AR by shA2AR did not affect the acquisition of propofol self-administration behavior, but enhance cue-induced reinstatement of propofol self-administration compared with the AAV-shCTRLgroup. Pharmacological activation of the A2AR by CGS21680 (≥ 5.0 ng/site) attenuated cue-induced reinstatement of propofol self-administration behavior. Similarly, pharmacological blockade of D2R by eticlopride (0.75-3.0 µg/site) attenuated propofol seeking behavior. These effects were reversed by the administration of MSX-3 (5.0-20.0 µg/site). The A2AR- and D2R-mediated effects on propofol relapse were not confounded by the learning process, and motor activity as the sucrose self-administration and locomotor activity were not affected by all the treatments. This study provides genetic and pharmacological evidence that NAc A2AR activation suppresses cue-induced propofol relapse in rats, possibly by interacting with D2R.


Subject(s)
Nucleus Accumbens/drug effects , Propofol/pharmacology , Receptor, Adenosine A2A/metabolism , Substance-Related Disorders/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Cues , Male , Nucleus Accumbens/metabolism , Phenethylamines/pharmacology , Propofol/administration & dosage , RNA, Small Interfering/pharmacology , Rats, Sprague-Dawley , Receptor, Adenosine A2A/deficiency , Recurrence , Self Administration , Xanthines/pharmacology
2.
Purinergic Signal ; 17(3): 439-448, 2021 09.
Article in English | MEDLINE | ID: mdl-33973110

ABSTRACT

Adenosine signaling plays a critical role in the maintenance of articular cartilage and may serve as a novel therapeutic for osteoarthritis (OA), a highly prevalent and morbid disease without effective therapeutics in the current market. Mice lacking adenosine A2A receptors (A2AR) develop spontaneous OA by 16 weeks of age, a finding relevant to human OA since loss of adenosine signaling due to diminished adenosine production (NT5E deficiency) also leads to development of OA in mice and humans. To better understand the mechanism by which A2AR and adenosine generation protect from OA development, we examined differential gene expression in neonatal chondrocytes from WT and A2AR null mice. Analysis of differentially expressed genes was analyzed by KEGG pathway analysis, and oPOSSUM and the flatiron database were used to identify transcription factor binding enrichment, and tissue-specific network analyses and patterns were compared to gene expression patterns in chondrocytes from patients with OA. There was a differential expression of 2211 genes (padj<0.05). Pathway enrichment analysis revealed that pro-inflammatory changes, increased metalloprotease, reduced matrix organization, and homeostasis are upregulated in A2AR null chondrocytes. Moreover, stress responses, including autophagy and HIF-1 signaling, seem to be important drivers of OA and bear marked resemblance to the human OA transcriptome. Although A2AR null mice are born with grossly intact articular cartilage, we identify here the molecular foundations for early-onset OA in these mice, further establishing their role as models for human disease and the potential use of adenosine as a treatment for human disease.


Subject(s)
Chondrocytes/metabolism , Osteoarthritis/metabolism , Receptor, Adenosine A2A/deficiency , Transcriptome/physiology , Animals , Animals, Newborn , Chondrocytes/pathology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoarthritis/genetics , Osteoarthritis/pathology , Receptor, Adenosine A2A/genetics , Sequence Analysis, RNA/methods , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Molecules ; 26(17)2021 Sep 02.
Article in English | MEDLINE | ID: mdl-34500787

ABSTRACT

The cannabis-derived molecules, ∆9 tetrahydrocannabinol (THC) and cannabidiol (CBD), are both of considerable therapeutic interest for a variety of purposes, including to reduce pain and anxiety and increase sleep. In addition to their other pharmacological targets, both THC and CBD are competitive inhibitors of the equilibrative nucleoside transporter-1 (ENT-1), a primary inactivation mechanism for adenosine, and thereby increase adenosine signaling. The goal of this study was to examine the role of adenosine A2A receptor activation in the effects of intraperitoneally administered THC alone and in combination with CBD or PECS-101, a 4'-fluorinated derivative of CBD, in the cannabinoid tetrad, elevated plus maze (EPM) and marble bury assays. Comparisons between wild-type (WT) and A2AR knock out (A2AR-KO) mice were made. The cataleptic effects of THC were diminished in A2AR-KO; no other THC behaviors were affected by A2AR deletion. CBD (5 mg/kg) potentiated the cataleptic response to THC (5 mg/kg) in WT but not A2AR-KO. Neither CBD nor THC alone affected EPM behavior; their combination produced a significant increase in open/closed arm time in WT but not A2AR-KO. Both THC and CBD reduced the number of marbles buried in A2AR-KO but not WT mice. Like CBD, PECS-101 potentiated the cataleptic response to THC in WT but not A2AR-KO mice. PECS-101 also reduced exploratory behavior in the EPM in both genotypes. These results support the hypothesis that CBD and PECS-101 can potentiate the cataleptic effects of THC in a manner consistent with increased endogenous adenosine signaling.


Subject(s)
Cannabidiol/pharmacology , Dronabinol/pharmacology , Receptor, Adenosine A2A/metabolism , Animals , Cannabidiol/analogs & derivatives , Dronabinol/administration & dosage , Exploratory Behavior/drug effects , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptor, Adenosine A2A/deficiency
4.
Biochim Biophys Acta ; 1863(7 Pt A): 1461-71, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27066978

ABSTRACT

Adenosine is known to reduce inflammation by suppressing the activity of most immune cells. Previous studies have shown that lipopolysaccharide (LPS) stimulated mouse macrophages produce adenosine, and the adenosine A2A receptor (A2AR) signaling activated in an autocrine manner attenuates LPS-induced pro-inflammatory cytokine formation. It has been suggested that A2AR signaling inhibits LPS-induced pro-inflammatory cytokine production through a unique cAMP-dependent, but PKA- and Epac-independent signaling pathway. However, the mechanism of inhibition was not identified so far. Here we report that LPS stimulation enhances A2AR expression in mouse bone marrow derived macrophages, and loss of A2ARs results in enhanced LPS-induced pro-inflammatory response. Loss of A2ARs in A2AR null macrophages did not alter the LPS-induced NF-κB activation, but an enhanced basal and LPS-induced phosphorylation of MAP kinases (especially that of JNKs) was detected in A2AR null cells. A2AR signaling did not alter the LPS-induced phosphorylation of their upstream kinases, but by regulating adenylate cyclase activity it enhanced the expression of dual specific phosphatase (DUSP)1, a negative regulator of MAP kinases. As a result, lower basal and LPS-induced DUSP1 mRNA and protein levels can be detected in A2AR null macrophages. Silencing of DUSP1 mRNA expression resulted in higher basal and LPS-induced JNK phosphorylation and LPS-induced pro-inflammatory cytokine formation in wild type macrophages, but had no effect on that in A2AR null cells. Our data indicate that A2AR signaling regulates both basal and LPS-induced DUSP1 levels in macrophages via activating the adenylate cyclase pathway.


Subject(s)
Dual Specificity Phosphatase 1/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Receptor, Adenosine A2A/metabolism , Signal Transduction/drug effects , Adenosine A2 Receptor Agonists/pharmacology , Adenylyl Cyclases/metabolism , Animals , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Dual Specificity Phosphatase 1/genetics , Genotype , Inflammation Mediators/immunology , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophages/enzymology , Macrophages/immunology , Mice, Knockout , NF-kappa B p50 Subunit/metabolism , Phenotype , Phosphorylation , RNA Interference , Receptor, Adenosine A2A/deficiency , Receptor, Adenosine A2A/drug effects , Receptor, Adenosine A2A/genetics , Time Factors , Transcription Factor RelA/metabolism , Transfection
5.
Acta Pharmacol Sin ; 38(4): 469-476, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28112177

ABSTRACT

Prostaglandin D2 (PGD2) is one of the most potent endogenous sleep promoting substances. PGD2 activates the PGD2 receptor (DPR) and increases the extracellular level of adenosine in wild-type (WT) mice but not DPR knockout (KO) mice, suggesting that PGD2-induced sleep is DPR-dependent, and adenosine may be the signaling molecule that mediates the somnogenic effect of PGD2. The aim of this study was to determine the involvement of the adenosine A2A receptor (A2AR) in PGD2-induced sleep. We infused PGD2 into the lateral ventricle of WT and A2AR KO mice between 20:00 and 2:00 for 6 h, and electroencephalograms and electromyograms were simultaneously recorded. In WT mice, PGD2 infusion dose-dependently increased non-rapid eye movement (non-REM, NREM) sleep, which was 139.1%, 145.0% and 202.7% as large as that of vehicle-treated mice at doses of 10, 20 and 50 pmol/min, respectively. PGD2 infusion at doses of 20 and 50 pmol/min also increased REM sleep during the 6-h PGD2 infusion and 4-h post-dosing periods in WT mice to 148.9% and 166.7%, respectively. In A2AR KO mice, however, PGD2 infusion at 10 pmol/min did not change the sleep profile, whereas higher doses at 20 and 50 pmol/min increased the NREM sleep during the 6-h PGD2 infusion to 117.5% and 155.6%, respectively, but did not change the sleep in the post-dosing period. Moreover, PGD2 infusion at 50 pmol/min significantly increased the episode number in both genotypes but only enhanced the episode duration in WT mice. The results demonstrate that PGD2-induced sleep in mice is mediated by both adenosine A2AR-dependent and -independent systems.


Subject(s)
Prostaglandin D2/pharmacology , Receptor, Adenosine A2A/deficiency , Sleep/drug effects , Animals , Infusions, Intraventricular , Male , Mice, Knockout , Prostaglandin D2/administration & dosage , Receptor, Adenosine A2A/metabolism , Wakefulness/drug effects
6.
Chembiochem ; 17(20): 1951-1960, 2016 10 17.
Article in English | MEDLINE | ID: mdl-27432157

ABSTRACT

The adenosine A2A receptor (A2A R) is expressed in immune cells, as well as brain and heart tissue, and has been intensively studied as a therapeutic target for multiple disease indications. Inhibitors of the A2A R have the potential for stimulating immune response, which could be valuable for cancer immune surveillance and mounting a response against pathogens. One well-established potent and selective small molecule A2A R antagonist, ZM-241385 (ZM), has a short pharmacokinetic half-life and the potential for systemic toxicity due to A2A R effects in the brain and the heart. In this study, we designed an analogue of ZM and tethered it to the Fc domain of the immunoglobulin IgG3 by using expressed protein ligation. The resulting protein-small molecule conjugate, Fc-ZM, retained high affinity for two Fc receptors: FcγRI and the neonatal Fc receptor, FcRn. In addition, Fc-ZM was a potent A2A R antagonist, as measured by a cell-based cAMP assay. Cell-based assays also revealed that Fc-ZM could stimulate interferon γ production in splenocytes in a fashion that was dependent on the presence of A2A R. We found that Fc-ZM, compared with the small molecule ZM, was a superior A2A R antagonist in mice, consistent with the possibility that Fc attachment can improve pharmacokinetic and/or pharmacodynamic properties of the small molecule.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Immunoglobulin Fab Fragments/pharmacology , Receptor, Adenosine A2A/metabolism , Triazines/pharmacology , Triazoles/pharmacology , Adenosine A2 Receptor Antagonists/chemical synthesis , Adenosine A2 Receptor Antagonists/chemistry , Animals , Female , Humans , Immunoglobulin Fab Fragments/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Molecular Structure , Receptor, Adenosine A2A/deficiency , Respiratory Tract Infections/drug therapy , Triazines/chemical synthesis , Triazines/chemistry , Triazoles/chemical synthesis , Triazoles/chemistry , Vaccinia virus/isolation & purification
7.
J Cardiovasc Pharmacol ; 66(6): 569-75, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26647014

ABSTRACT

Numerous evidence suggests that RhoA/Rho kinase (ROCK) signaling pathway plays an important role in the pathogenesis of pulmonary arterial hypertension (PAH), but little is known about its effects on the development of PAH in mice with absence of the adenosine A2A receptor (A2AR). Eight A2AR knockout (KO) and 8 wild-type mice were used. Morphometric analysis of pulmonary arterioles included right ventricle/left ventricle plus ventricular septum (Fulton index), vessel wall thickness/total vascular diameter (WT%), and vessel wall area/total vascular area (WA%). The expression of RhoA and ROCK1 mRNA was determined by real-time polymerase chain reaction. The expression of RhoA, ROCK1, and phosphorylation of myosin phosphatase target subunit 1 proteins in pulmonary tissue was tested by Western blot. The position of ROCK1 protein was evaluated by immunohistochemistry. Compared with wild-type mice, A2AR KO mice displayed (1) increased Fulton index, WT%, and WA% (P < 0.01); (2) increased mRNA expression of RhoA and ROCK1 (each P < 0.05); (3) increased protein expression of RhoA, ROCK1, and phosphorylation of myosin phosphatase target subunit 1 (each P < 0.01); (4) increased location of ROCK1 protein in endothelial and smooth muscle cells of pulmonary artery, bronchial, and alveolar epithelial cells. Activation of RhoA/ROCK signaling pathway may cause pulmonary vascular constriction, pulmonary artery remodeling, and PAH in adenosine A2A receptor KO mice.


Subject(s)
Hypertension, Pulmonary/metabolism , Receptor, Adenosine A2A/deficiency , Signal Transduction/physiology , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism , Animals , Hypertension, Pulmonary/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , rhoA GTP-Binding Protein
8.
J Neurosci ; 33(47): 18492-502, 2013 Nov 20.
Article in English | MEDLINE | ID: mdl-24259572

ABSTRACT

Astrocytic glutamate transporter-1 (GLT-I) is critical to control the bulk of glutamate uptake and, thus, to regulate synaptic plasticity and excitotoxicity. GLT-I glutamate uptake is driven by the sodium gradient implemented by Na(+)/K(+)-ATPases (NKAs) and the α2 subunit of NKA (NKA-α2) is actually linked to GLT-I to regulate astrocytic glutamate transport. We recently found that adenosine A2A receptors (A2ARs), which control synaptic plasticity and neurodegeneration, regulate glutamate uptake through unknown mechanisms. Here we report that A2AR activation decreases NKA activity selectively in astrocytes to inhibit glutamate uptake. Furthermore, we found a physical association of A2ARs with NKA-α2s in astrocytes, as gauged by coimmunoprecipitation and in situ proximity ligation assays, in the cerebral cortex and striatum, two brain regions where A2ARs inhibit the astrocytic glutamate uptake. Moreover, the selective deletion of A2ARs in astrocytes (using Gfa2-A2AR-KO mice) leads to a concurrent increase of both astrocytic glutamate uptake and NKA-α2 levels and activity in the striatum and cortex. This coupling of astrocytic A2ARs to the regulation of glutamate transport through modulation of NKA-α2 activity provides a novel mechanism linking neuronal activity to ion homeostasis controlling glutamatergic activity, all of which are processes intricately associated with the etiology of several brain diseases.


Subject(s)
Astrocytes/drug effects , Glutamic Acid/metabolism , Receptor, Adenosine A2A/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Analysis of Variance , Animals , Aspartic Acid/metabolism , Astrocytes/ultrastructure , Glial Fibrillary Acidic Protein/genetics , Immunoprecipitation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ouabain/pharmacology , Phenethylamines/pharmacology , Pyrimidines/pharmacology , Receptor, Adenosine A2A/deficiency , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Triazoles/pharmacology , Tritium/metabolism
9.
Am J Physiol Gastrointest Liver Physiol ; 307(3): G338-46, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24875104

ABSTRACT

Adenosine is a purine metabolite that can mediate anti-inflammatory responses in the digestive tract through the A(2A) adenosine receptor (A(2A)AR). We examined the role of this receptor in the control of inflammation in the adoptive transfer model of colitis. Infection of A(2A)AR(-/-) mice with Helicobacter hepaticus increased colonic inflammation scores compared with uninfected A(2A)AR controls. Comparison of T cell subsets in wild-type and A(2A)AR(-/-) mice revealed differences in markers associated with activated helper T (Th) cells and regulatory T (Treg) cells. Previous studies showed that expression of A(2A)AR on CD45RB(HI) and CD45RB(LO) Th cells is essential for the proper regulation of colonic inflammation. Adoptive transfer of CD45RB(HI) with CD45RB(LO) from wild-type mice into RAG1(-/-)/A(2A)AR(-/-) mice induced severe disease within 3 wk, although transfer of the same subsets into RAG1(-/-) mice does not induce colitis. This suggests that the presence of A(2A)AR on recipient cells is also important for controlling colitis. To investigate the role of A(2A)AR in myeloid cells, chimeric recipients were generated by injection of bone marrow from RAG1(-/-) or RAG1(-/-)/A(2A)AR(-/-) mice into irradiated RAG1(-/-) mice. After adoptive transfer, these recipients did not develop colitis, regardless of A(2A)AR expression by the donor. Together, our results suggest that the control of inflammation in vivo is dependent on A(2A)AR signaling through multiple cell types that collaborate in the regulation of colitis by responding to extracellular adenosine.


Subject(s)
Adenosine/metabolism , Colitis/prevention & control , Colon/metabolism , Lymph Nodes/metabolism , T-Lymphocyte Subsets/metabolism , Adoptive Transfer , Animals , Biomarkers/metabolism , Colitis/immunology , Colitis/metabolism , Colitis/microbiology , Colon/immunology , Colon/microbiology , Cytokines/metabolism , Disease Models, Animal , Female , Helicobacter hepaticus/pathogenicity , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Inflammation Mediators/metabolism , Leukocyte Common Antigens/metabolism , Lymph Nodes/immunology , Lymph Nodes/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Adenosine A2A/deficiency , Receptor, Adenosine A2A/genetics , Severity of Illness Index , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/microbiology , T-Lymphocyte Subsets/transplantation , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Time Factors
10.
Brain Behav Immun ; 41: 218-31, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24907587

ABSTRACT

Anxiety is one of the most commonly reported psychiatric conditions, but its pathogenesis is poorly understood. Ailments associated with activation of the innate immune system, however, are increasingly linked to anxiety disorders. In adult male mice, we found that adenosine doubled caspase-1 activity in brain by a pathway reliant on ATP-sensitive potassium (KATP) channels, protein kinase A (PKA) and the A2A adenosine receptor (AR). In addition, adenosine-dependent activation of caspase-1 increased interleukin (IL)-1ß in the brain by 2-fold. Peripheral administration of adenosine in wild-type (WT) mice led to a 2.3-fold increase in caspase-1 activity in the amygdala and to a 33% and 42% reduction in spontaneous locomotor activity and food intake, respectively, that were not observed in caspase-1 knockout (KO), IL-1 receptor type 1 (IL-1R1) KO and A2A AR KO mice or in mice administered a caspase-1 inhibitor centrally. Finally, adenosine administration increased anxiety-like behaviors in WT mice by 28% in the open field test and by 55% in the elevated zero-maze. Caspase-1 KO mice, IL-1R1 KO mice, A2A AR KO mice and WT mice treated with the KATP channel blocker, glyburide, were resistant to adenosine-induced anxiety-like behaviors. Thus, our results indicate that adenosine can act as an anxiogenic by activating caspase-1 and increasing IL-1ß in the brain.


Subject(s)
Adenosine/toxicity , Anxiety/chemically induced , Brain/metabolism , Caspase 1/physiology , Interleukin-1beta/biosynthesis , Nerve Tissue Proteins/physiology , Receptor, Adenosine A2A/physiology , Adenosine/pharmacology , Amygdala/metabolism , Animals , Anxiety/physiopathology , Carbazoles/pharmacology , Caspase 1/deficiency , Cyclic AMP-Dependent Protein Kinases/physiology , Enzyme Activation/drug effects , Exploratory Behavior/drug effects , Feeding Behavior/drug effects , Glyburide/pharmacology , Interleukin-1beta/genetics , Interleukin-1beta/physiology , Ion Transport/drug effects , KATP Channels/physiology , Locomotion/drug effects , Maze Learning/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/deficiency , Potassium/metabolism , Pyrroles/pharmacology , Receptor, Adenosine A2A/deficiency , Receptor, Adenosine A2A/drug effects , Receptors, Interleukin-1 Type I/deficiency , Receptors, Interleukin-1 Type I/physiology
11.
J Immunol ; 188(11): 5713-22, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22529293

ABSTRACT

Extracellular adenosine has an important role in regulating the severity of inflammation during an immune response. Although there are four adenosine receptor (AR) subtypes, the A2AAR is both highly expressed on lymphocytes and known as a prime mediator of adenosine's anti-inflammatory effects. To define the importance of A2AAR signaling during neuroinflammatory disease progression, we used the experimental autoimmune encephalomyelitis (EAE) animal model for multiple sclerosis. In EAE induction experiments, A2AAR antagonist treatment protected mice from disease development and its associated CNS lymphocyte infiltration. However, A2AAR(-/-) mice developed a more severe acute EAE phenotype characterized by more proinflammatory lymphocytes and activated microglia/macrophages. Interestingly, very high levels of A2AAR were expressed on the choroid plexus, a well-established CNS lymphocyte entry point. To determine the contribution of A2AAR signaling in lymphocytes and the CNS during EAE, we used bone marrow chimeric mice. Remarkably, A2AAR(-/-) donor hematopoietic cells potentiated severe EAE, whereas lack of A2AAR expression on nonhematopoietic cells protected against disease development. Although no defect in the suppressive ability of A2AAR(-/-) regulatory T cells was observed, A2AAR(-/-) lymphocytes were shown to proliferate more and produced more IFN-γ following stimulation. Despite this more proinflammatory phenotype, A2AAR antagonist treatment still protected against EAE when A2AAR(-/-) lymphocytes were adoptively transferred to T cell-deficient A2AAR(+/+) mice. These results indicate that A2AAR expression on nonimmune cells (likely in the CNS) is required for efficient EAE development, while A2AAR lymphocyte expression is essential for limiting the severity of the inflammatory response.


Subject(s)
Brain/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammation Mediators/physiology , Lymphocytes/immunology , Receptor, Adenosine A2A/physiology , Signal Transduction/immunology , Spinal Cord/immunology , Up-Regulation/immunology , Animals , Brain/metabolism , Brain/pathology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Lymphocytes/metabolism , Lymphocytes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptor, Adenosine A2A/deficiency , Receptor, Adenosine A2A/metabolism , Severity of Illness Index , Signal Transduction/genetics , Spinal Cord/metabolism , Spinal Cord/pathology , Up-Regulation/genetics
12.
Ann Neurol ; 71(2): 278-82, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22367999

ABSTRACT

To investigate the putative interaction between chronic exposure to adenosine receptor antagonist caffeine and genetic influences on Parkinson's disease (PD), we determined whether deletion of the adenosine A(2A) receptor in knockout (KO) mice protects against dopaminergic neuron degeneration induced by a mutant human α-synuclein (hm(2)-αSYN) transgene containing both A53T and A30P. The A(2A) KO completely prevented loss of dopamine and dopaminergic neurons caused by the mutant α-synuclein transgene without altering levels of its expression. The adenosine A(2A) receptor appears required for neurotoxicity in a mutant α-synuclein model of PD. Together with prior studies the present findings indirectly support the neuroprotective potential of caffeine and more specific A(2A) antagonists.


Subject(s)
Gene Deletion , Parkinson Disease/genetics , Parkinson Disease/metabolism , Receptor, Adenosine A2A/deficiency , Receptor, Adenosine A2A/genetics , alpha-Synuclein/metabolism , Adenosine A2 Receptor Antagonists/therapeutic use , Animals , Caffeine/therapeutic use , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Receptor, Adenosine A2A/physiology
13.
J Am Soc Nephrol ; 23(9): 1528-37, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22835488

ABSTRACT

Regulatory T cells (Tregs) suppress the innate inflammation associated with kidney ischemia-reperfusion injury (IRI), but the mechanism is not well understood. Tregs express CD73, the final enzyme involved in the production of extracellular adenosine, and activation of the adenosine 2A receptor (A(2A)R) on immune cells suppresses inflammation and preserves kidney function after IRI. We hypothesized that Treg-generated adenosine is required to block innate immune responses in kidney IRI and that the Treg-generated adenosine would signal through A(2A)Rs on inflammatory cells and, in an autocrine manner, on Tregs themselves. We found that adoptively transferred wild-type Tregs protected wild-type mice from kidney IRI, but the absence of adenosine generation (CD73-deficient Tregs) or adenosine responsiveness (A(2A)R-deficient Tregs) led to inhibition of Treg function. Pharmacologic stimulation of A(2A)R before adoptive transfer augmented the ability of wild-type and CD73-deficient Tregs to suppress kidney IRI. Microarray analysis and flow cytometry revealed that A(2A)R activation enhanced surface PD-1 expression on Tregs in the absence of any other activation signal. Treatment of Tregs with a PD-1 blocking antibody before adoptive transfer reversed their protective effects, even if pretreated with an A(2A)R agonist. Taken together, these results demonstrate that the simultaneous ability to generate and respond to adenosine is required for Tregs to suppress innate immune responses in IRI through a PD-1-dependent mechanism.


Subject(s)
Adenosine/physiology , Autocrine Communication/physiology , Kidney/blood supply , Reperfusion Injury/prevention & control , T-Lymphocytes, Regulatory/physiology , 5'-Nucleotidase/deficiency , 5'-Nucleotidase/genetics , 5'-Nucleotidase/physiology , Animals , Immunity, Innate/physiology , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Programmed Cell Death 1 Receptor/physiology , Receptor, Adenosine A2A/deficiency , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/physiology , Reperfusion Injury/pathology , Signal Transduction/physiology , T-Lymphocytes, Regulatory/pathology
14.
J Neurosci ; 31(27): 10067-75, 2011 Jul 06.
Article in English | MEDLINE | ID: mdl-21734299

ABSTRACT

Caffeine, the most widely used psychoactive compound, is an adenosine receptor antagonist. It promotes wakefulness by blocking adenosine A(2A) receptors (A(2A)Rs) in the brain, but the specific neurons on which caffeine acts to produce arousal have not been identified. Using selective gene deletion strategies based on the Cre/loxP technology in mice and focal RNA interference to silence the expression of A(2A)Rs in rats by local infection with adeno-associated virus carrying short-hairpin RNA, we report that the A(2A)Rs in the shell region of the nucleus accumbens (NAc) are responsible for the effect of caffeine on wakefulness. Caffeine-induced arousal was not affected in rats when A(2A)Rs were focally removed from the NAc core or other A(2A)R-positive areas of the basal ganglia. Our observations suggest that caffeine promotes arousal by activating pathways that traditionally have been associated with motivational and motor responses in the brain.


Subject(s)
Arousal/drug effects , Caffeine/pharmacology , Central Nervous System Stimulants/pharmacology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Receptor, Adenosine A2A/metabolism , Analysis of Variance , Animals , Basal Ganglia/drug effects , Basal Ganglia/metabolism , Cell Line, Transformed , Choline O-Acetyltransferase/metabolism , Dose-Response Relationship, Drug , Electroencephalography/methods , Electromyography/methods , Green Fluorescent Proteins/genetics , Humans , Locomotion/drug effects , Locomotion/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutagenesis , Mutation/genetics , Phosphopyruvate Hydratase/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A2A/deficiency , Receptor, Adenosine A2A/genetics , Receptors, Dopamine D2/metabolism , Transfection/methods
15.
J Cell Biochem ; 113(5): 1766-75, 2012 May.
Article in English | MEDLINE | ID: mdl-22213163

ABSTRACT

Extracellular adenosine induced apoptosis in HepG2 cells, a human hepatoma cell line, by tuning apoptosis-mediator gene transcription. The present study aimed at identifying the responsible adenosine receptor and clarifying the signaling pathway underlying adenosine-induced HepG2 cell apoptosis. Adenosine and CGS21680, an A(2a) adenosine receptor agonist, induced HepG2 cell apoptosis, and the effect was inhibited by DMPX, an A(2a) adenosine receptor antagonist, or by knocking-down A(2a) adenosine receptors. Adenosine reduced expression of Bcl-X(L) mRNA and protein but otherwise increased expression of the Bid mRNA and protein in HepG2 cells, and those effects were also prevented by knocking-down A(2a) adenosine receptors. Adenosine caused disruption of mitochondrial membrane potentials and stimulated cytochrome c efflux from the mitochondria in HepG2 cells. Adenosine activated caspases-3 and -9 in HepG2 cells, which was significantly inhibited by knocking-down A(2a) adenosine receptors. The results of the present study indicate that extracellular adenosine downregulates Bcl-X(L) expression and upregulates Bid expression, thereby disrupting mitochondrial membrane potentials to allow cytochrome c efflux from the mitochondria, and then causing activation of caspase-9 and the effector caspase-3, as mediated via A(2a) adenosine receptors.


Subject(s)
Apoptosis/physiology , BH3 Interacting Domain Death Agonist Protein/metabolism , Receptor, Adenosine A2A/metabolism , bcl-X Protein/metabolism , Adenosine/pharmacology , Adenosine A2 Receptor Agonists/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , BH3 Interacting Domain Death Agonist Protein/genetics , Base Sequence , Caspases/metabolism , Cytochromes c/metabolism , Down-Regulation , Gene Knockdown Techniques , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Receptor, Adenosine A2A/deficiency , Receptor, Adenosine A2A/genetics , Signal Transduction , Up-Regulation , bcl-X Protein/genetics
16.
J Neurochem ; 123(1): 100-12, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22639925

ABSTRACT

Studies with multiple sclerosis patients and animal models of experimental autoimmune encephalomyelitis (EAE) implicate adenosine and adenosine receptors in modulation of neuroinflammation and brain injury. Although the involvement of the A(1) receptor has been recently demonstrated, the role of the adenosine A(2A) receptor (A(2A)R) in development of EAE pathology is largely unknown. Using mice with genetic inactivation of the A(2A) receptor, we provide direct evidence that loss of the A(2A)R exacerbates EAE pathology in mice. Compared with wild-type mice, A(2A)R knockout mice injected with myelin oligodendroglia glycoprotein peptide had a higher incidence of EAE and exhibited higher neurological deficit scores and greater decrease in body weight. A(2A)R knockout mice displayed increased inflammatory cell infiltration and enhanced microglial cell activation in cortex, brainstem, and spinal cord. In addition, demyelination and axonal damage in brainstem were exacerbated, levels of Th1 cytokines increased, and Th2 cytokines decreased. Collectively, these findings suggest that extracellular adenosine acting at A(2A)Rs triggers an important neuroprotective mechanism. Thus, the A(2A) receptor is a potential target for therapeutic approaches to multiple sclerosis.


Subject(s)
Brain Injuries/etiology , Brain Injuries/pathology , Encephalomyelitis, Autoimmune, Experimental/complications , Gene Expression Regulation/genetics , Microglia/pathology , Receptor, Adenosine A2A/genetics , Adenosine A1 Receptor Antagonists/pharmacokinetics , Animals , Astrocytes/drug effects , Astrocytes/pathology , Axons/pathology , Brain Injuries/complications , Cell Proliferation , Cells, Cultured , Cerebral Cortex/pathology , Cytokines/metabolism , Demyelinating Diseases/etiology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Enzyme-Linked Immunosorbent Assay , Female , Filtration , Flow Cytometry , Freund's Adjuvant/toxicity , Gene Expression Regulation/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/toxicity , Peptide Fragments/toxicity , Protein Binding/drug effects , Protein Binding/genetics , RNA, Messenger/metabolism , Receptor, Adenosine A2A/deficiency , Spinal Cord/pathology , Spleen/cytology , Statistics, Nonparametric , Tritium/pharmacokinetics , Xanthines/pharmacokinetics
17.
J Exp Med ; 203(12): 2639-48, 2006 Nov 27.
Article in English | MEDLINE | ID: mdl-17088433

ABSTRACT

Ischemia reperfusion injury results from tissue damage during ischemia and ongoing inflammation and injury during reperfusion. Liver reperfusion injury is reduced by lymphocyte depletion or activation of adenosine A2A receptors (A2ARs) with the selective agonist 4-{3-[6-amino-9-(5-ethylcarbamoyl-3,4-dihydroxy-tetrahydro-furan-2-yl)-9H-purin-2-yl]- prop-2-ynyl}-cyclohexanecarboxylic acid methyl ester (ATL146e). We show that NKT cells are stimulated to produce interferon (IFN)-gamma by 2 h after the initiation of reperfusion, and the use of antibodies to deplete NK1.1-positive cells (NK and NKT) or to block CD1d-mediated glycolipid presentation to NKT cells replicates, but is not additive to, the protection afforded by ATL146e, as assessed by serum alanine aminotransferase elevation, histological necrosis, neutrophil accumulation, and serum IFN-gamma elevation. Reduced reperfusion injury observed in RAG-1 knockout (KO) mice is restored to the wild-type (WT) level by adoptive transfer of NKT cells purified from WT or A2AR KO mice but not IFN-gamma KO mice. Additionally, animals with transferred A2AR-/- NKT cells are not protected from hepatic reperfusion injury by ATL146e. In vitro, ATL146e potently inhibits both anti-CD3 and alpha-galactosylceramide-triggered production of IFN-gamma by NKT cells. These findings suggest that hepatic reperfusion injury is initiated by the CD1d-dependent activation of NKT cells, and the activation of these cells is inhibited by A2AR activation.


Subject(s)
Antigens, CD1/physiology , Killer Cells, Natural/immunology , Liver/blood supply , Lymphocyte Activation/immunology , Receptor, Adenosine A2A/metabolism , Reperfusion Injury/immunology , Reperfusion Injury/prevention & control , T-Lymphocyte Subsets/immunology , Animals , Antigens, CD1d , Immunosuppression Therapy , Ischemic Preconditioning , Killer Cells, Natural/metabolism , Liver/immunology , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Adenosine A2A/deficiency , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/physiology , Reperfusion Injury/pathology , T-Lymphocyte Subsets/metabolism
18.
Cancer Immunol Immunother ; 61(6): 917-26, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22116345

ABSTRACT

The A(2A) adenosine receptor plays a critical and non-redundant role in suppressing inflammation at sites of hypoxia and tissue damage. The tumor microenvironment has high levels of adenosine as a result of hypoxia and ectopic expression of enzymes responsible for the generation of extracellular adenosine. Thus, we sought to determine the ability of A(2A) receptor null mice to immunologically reject tumors. We observed that mice lacking the A(2A) adenosine receptor showed significantly delayed growth of lymphoma cells when compared to WT mice. Furthermore, when immunized with a low dose of tumor or with an irradiated GM-CSF-secreting tumor vaccine, A(2A) receptor null mice showed significantly enhanced protection from a subsequent high-dose challenge from both immunogenic and poorly immunogenic tumor lines. This increase in protection was accompanied by an increase in the number of tumor-antigen-specific CD8 T cells at the vaccine-site draining lymph node. Finally, we found that A(2A) receptor null mice displayed more robust anti-tumor responses than WT mice when they were treated with a soluble B7-DC/Fc fusion protein designed to antagonize B7-H1-mediated co-inhibition. This combinatorial immunotherapy strategy could also be recapitulated with pharmacological A(2A) receptor blockade paired with B7-DC/Fc administration. In light of these data, we believe that blockade of the A(2A) adenosine receptor is an attractive target for tumor immunotherapy that synergizes with other immunomodulatory approaches currently in clinical trials.


Subject(s)
Immunotherapy/methods , Neoplasms/therapy , Receptor, Adenosine A2A/deficiency , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/immunology , Neoplasms/pathology , Signal Transduction/immunology , Tumor Cells, Cultured , Tumor Microenvironment
19.
Int J Neuropsychopharmacol ; 15(7): 995-1001, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21816123

ABSTRACT

Adenosine A2A receptors and metabotropic glutamate type 5 (mGlu5) receptors are co-localized in the striatum and can functionally interact to regulate drug-seeking. We further explored this interaction using antagonism of mGlu5 receptors with 3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]-pyridine (MTEP) in combination with genetic deletion of A2A receptors. The conditioned rewarding and locomotor-activating properties of cocaine were evaluated via conditioned place preference (CPP). Vehicle-treated mice of both genotypes expressed a CPP to cocaine while MTEP abolished cocaine CPP in wild-type, but not A2A knockout, mice. These results were mirrored when conditioned hyperactivity was assessed. In contrast, MTEP attenuated the acute locomotor-activating properties of cocaine similarly in both genotypes. These data provide evidence for a functional interaction between adenosine A2A and mGlu5 receptors in mediating the conditioned effects of cocaine but not direct cocaine-induced hyperactivity. This functional interaction is supported by modulation of 4-(2-[7-amino-2-[2-furyl][1,2,4]triazolol[2,3-a][1,3,5]triazin-5-yl-amino]ethyl)phenol ([125I]ZM241385) binding to the A2A receptor by MTEP.


Subject(s)
Brain/metabolism , Cocaine/pharmacology , Conditioning, Operant/drug effects , Dopamine Uptake Inhibitors/pharmacology , Receptor, Adenosine A2A/metabolism , Receptors, Metabotropic Glutamate/metabolism , Analysis of Variance , Animals , Autoradiography , Brain/diagnostic imaging , Brain/drug effects , Conditioning, Operant/physiology , Drug Interactions , Excitatory Amino Acid Antagonists/pharmacology , Iodine Isotopes/pharmacokinetics , Locomotion/drug effects , Locomotion/genetics , Male , Mice , Mice, Knockout , Protein Binding/drug effects , Protein Binding/genetics , Pyridines/pharmacology , Radionuclide Imaging , Receptor, Adenosine A2A/deficiency , Receptor, Metabotropic Glutamate 5 , Thiazoles/pharmacology , Time Factors , Triazines/pharmacokinetics , Triazoles/pharmacokinetics
20.
Neurobiol Dis ; 41(2): 570-6, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21062644

ABSTRACT

Huntington's disease (HD) is a progressive neurodegenerative genetic disorder that leads to motor, cognitive, and psychiatric disturbances. The primary neuropathological hallmark is atrophy of the striatum. HD preferentially affects efferent striato-pallidal neurons that express enkephalin as well as dopamine D2 and A(2A) adenosine receptors (A(2A)Rs). Expression and function of A(2A)Rs are altered in HD but, despite being an important modulator of the striato-pallidal function, the subsequent pathophysiological consequence of such changes remains unclear. Whether blockade of A(2A)Rs is of therapeutic interest in HD remains ill-defined. In the present work, we aimed to determine the pathophysiological consequences of genetic deletion of A(2A)Rs in HD by crossing A(2A)R knockout mice with the N171-82Q HD transgenic model. Our data demonstrate that knockout of A(2A)Rs moderately but significantly worsens motor performances and survival of N171-82Q mice and leads to a decrease in striatal enkephalin expression. These results support that early and chronic blockade of A(2A)Rs might not be beneficial in HD.


Subject(s)
Huntington Disease/genetics , Huntington Disease/metabolism , Movement Disorders/genetics , Movement Disorders/metabolism , Receptor, Adenosine A2A/deficiency , Receptor, Adenosine A2A/genetics , Animals , Enkephalins/antagonists & inhibitors , Enkephalins/metabolism , Female , Huntingtin Protein , Huntington Disease/pathology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Movement Disorders/pathology , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neurons/pathology , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL