Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Neuropharmacology ; 258: 110055, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-38950692

ABSTRACT

Sleep disturbances and persistent pain conditions are public health challenges worldwide. Although it is well-known that sleep deficit increases pain sensitivity, the underlying mechanisms remain elusive. We have recently demonstrated the involvement of nucleus accumbens (NAc) and anterior cingulate cortex (ACC) in the pronociceptive effect of sleep restriction. In this study, we found that sleep restriction increases c-Fos expression in NAc and ACC, suggesting hyperactivation of these regions during prolonged wakefulness in male Wistar rats. Blocking adenosine A2A receptors in the NAc or GABAA receptors in the ventral tegmental area (VTA), dorsal raphe nucleus (DRN), or locus coeruleus (LC) effectively mitigated the pronociceptive effect of sleep restriction. In contrast, the blockade of GABAA receptors in each of these nuclei only transiently reduced carrageenan-induced hyperalgesia. Pharmacological activation of dopamine D2, serotonin 5-HT1A and noradrenaline alpha-2 receptors within the ACC also prevented the pronociceptive effect of sleep restriction. While pharmacological inhibition of these same monoaminergic receptors in the ACC restored the pronociceptive effect which had been prevented by the GABAergic disinhibition of the of the VTA, DRN or LC. Overall, these findings suggest that the pronociceptive effect of sleep restriction relies on increased adenosinergic activity on NAc, heightened GABAergic activity in VTA, DRN, and LC, and reduced inhibitory monoaminergic activity on ACC. These findings advance our understanding of the interplay between sleep and pain, shedding light on potential NAc-brainstem-ACC mechanisms that could mediate increased pain sensitivity under conditions of sleep impairment.


Subject(s)
Nucleus Accumbens , Rats, Wistar , Sleep Deprivation , Ventral Tegmental Area , Animals , Male , Sleep Deprivation/metabolism , Sleep Deprivation/physiopathology , Rats , Ventral Tegmental Area/metabolism , Ventral Tegmental Area/drug effects , Nucleus Accumbens/metabolism , Nucleus Accumbens/drug effects , Receptor, Adenosine A2A/metabolism , Hyperalgesia/metabolism , Dorsal Raphe Nucleus/metabolism , Dorsal Raphe Nucleus/drug effects , Gyrus Cinguli/metabolism , Gyrus Cinguli/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Brain Stem/metabolism , Brain Stem/drug effects , Locus Coeruleus/metabolism , Locus Coeruleus/drug effects , Carrageenan , Receptors, GABA-A/metabolism , Receptors, Dopamine D2/metabolism , Adenosine A2 Receptor Antagonists/pharmacology
2.
Cells ; 13(10)2024 May 16.
Article in English | MEDLINE | ID: mdl-38786068

ABSTRACT

Induction of the adenosine receptor A2B (A2BAR) expression in diabetic glomeruli correlates with an increased abundance of its endogenous ligand adenosine and the progression of kidney dysfunction. Remarkably, A2BAR antagonism protects from proteinuria in experimental diabetic nephropathy. We found that A2BAR antagonism preserves the arrangement of podocytes on the glomerular filtration barrier, reduces diabetes-induced focal adhesion kinase (FAK) activation, and attenuates podocyte foot processes effacement. In spreading assays using human podocytes in vitro, adenosine enhanced the rate of cell body expansion on laminin-coated glass and promoted peripheral pY397-FAK subcellular distribution, while selective A2BAR antagonism impeded these effects and attenuated the migratory capability of podocytes. Increased phosphorylation of the Myosin2A light chain accompanied the effects of adenosine. Furthermore, when the A2BAR was stimulated, the cells expanded more broadly and more staining of pS19 myosin was detected which co-localized with actin cables, suggesting increased contractility potential in cells planted onto a matrix with a stiffness similar to of the glomerular basement membrane. We conclude that A2BAR is involved in adhesion dynamics and contractile actin bundle formation, leading to podocyte foot processes effacement. The antagonism of this receptor may be an alternative to the intervention of glomerular barrier deterioration and proteinuria in the diabetic kidney disease.


Subject(s)
Cell Adhesion , Diabetes Mellitus, Experimental , Focal Adhesion Protein-Tyrosine Kinases , Podocytes , Proteinuria , Receptor, Adenosine A2B , Animals , Humans , Male , Rats , Adenosine/metabolism , Adenosine/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Cell Adhesion/drug effects , Cell Movement/drug effects , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Diabetic Nephropathies/drug therapy , Focal Adhesion Protein-Tyrosine Kinases/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Myosin Light Chains/metabolism , Phosphorylation/drug effects , Podocytes/metabolism , Podocytes/drug effects , Podocytes/pathology , Proteinuria/metabolism , Receptor, Adenosine A2B/drug effects , Receptor, Adenosine A2B/metabolism
3.
Int J Mol Sci ; 24(13)2023 Jun 29.
Article in English | MEDLINE | ID: mdl-37446007

ABSTRACT

Some chemoattractants and leukocytes such as M1 and M2 macrophages are known to be involved in the development of glomerulosclerosis during diabetic nephropathy (DN). In the course of diabetes, an altered and defective cellular metabolism leads to the increase in adenosine levels, and thus to changes in the polarity (M1/M2) of macrophages. MRS1754, a selective antagonist of the A2B adenosine receptor (A2BAR), attenuated glomerulosclerosis and decreased macrophage-myofibroblast transition in DN rats. Therefore, we aimed to investigate the effect of MRS1754 on the glomerular expression/secretion of chemoattractants, the intraglomerular infiltration of leukocytes, and macrophage polarity in DN rats. Kidneys/glomeruli of non-diabetic, DN, and MRS1754-treated DN rats were processed for transcriptomic analysis, immunohistopathology, ELISA, and in vitro macrophage migration assays. The transcriptomic analysis identified an upregulation of transcripts and pathways related to the immune system in the glomeruli of DN rats, which was attenuated using MRS1754. The antagonism of the A2BAR decreased glomerular expression/secretion of chemoattractants (CCL2, CCL3, CCL6, and CCL21), the infiltration of macrophages, and their polarization to M2 in DN rats. The in vitro macrophages migration induced by conditioned-medium of DN glomeruli was significantly decreased using neutralizing antibodies against CCL2, CCL3, and CCL21. We concluded that the pharmacological blockade of the A2BAR decreases the transcriptional expression of genes/pathways related to the immune response, protein expression/secretion of chemoattractants, as well as the infiltration of macrophages and their polarization toward the M2 phenotype in the glomeruli of DN rats, suggesting a new mechanism implicated in the antifibrotic effect of MRS1754.


Subject(s)
Acetamides , Adenosine A2 Receptor Antagonists , Cell Polarity , Chemotactic Factors , Diabetic Nephropathies , Kidney Glomerulus , Macrophages , Purines , Diabetic Nephropathies/genetics , Diabetic Nephropathies/immunology , Kidney Glomerulus/drug effects , Kidney Glomerulus/metabolism , Chemotactic Factors/antagonists & inhibitors , Chemotactic Factors/genetics , Chemotactic Factors/metabolism , Cell Polarity/drug effects , Cell Polarity/immunology , Macrophages/drug effects , Macrophages/immunology , Adenosine A2 Receptor Antagonists/pharmacology , Receptor, Adenosine A2B , Acetamides/pharmacology , Purines/pharmacology , Animals , Rats , Cell Movement/drug effects , Male , Rats, Sprague-Dawley , Transcription, Genetic/drug effects , Protein Biosynthesis/drug effects , Immunity/drug effects , Immunity/genetics
4.
J Biomol Struct Dyn ; 40(20): 9592-9601, 2022.
Article in English | MEDLINE | ID: mdl-34180379

ABSTRACT

Sickle cell disease (SCD) is a disease resulting from mutation in the globin portion of hemoglobin caused by the replacement of adenine for thymine in the codon of the ß globin gene. In Brazil, SCD affects about 0.3% of the black and Caucasian population. Until now, there is no specific treatment and the available drugs have several serious adverse effects which makes the search for new drugs an emergently need. The use of computational techniques can accelerate the drug development process by prioritization of molecules with affinity against essential targets. Adenosine A2b receptor (rA2b) has been studied in SCD due to its relationship with red blood cells concentration of 2,3-diphosphoglycerate which reduces the hemoglobin affinity for oxygen (O2), facilitating its availability for the tissues. Then, development of rA2b antagonists could be helpful for the treatment of SCD. However, there is still no 3D structure of rA2b and to overcome this limitation, homology modeling should be applied. In this scenario, this study aims to build a suitable 3D model of rA2b by SWISS MODEL and to evaluate the structural aspects of rA2b with known antagonists that may be useful for the identification of new potential antagonists by molecular dynamics on a lipid bilayer environment using GROMACS 5.1.4. The complexes with antagonists ZINC223070016 and ZINC17974526 interacted with key residues by hydrophobic contacts and hydrogen bonds which stabilized them at the rA2b binding site. This intermolecular profile can contribute to the development of more potent rA2b antagonists. Communicated by Ramaswamy H. Sarma.


Subject(s)
Adenosine A2 Receptor Antagonists , Anemia, Sickle Cell , Humans , Adenosine A2 Receptor Antagonists/chemistry , Receptor, Adenosine A2B/chemistry , Anemia, Sickle Cell/drug therapy , Molecular Dynamics Simulation , Hydrogen Bonding
5.
Purinergic Signal ; 17(2): 303-312, 2021 06.
Article in English | MEDLINE | ID: mdl-33860899

ABSTRACT

The role of peripheral adenosine receptors in pain is a controversial issue and seems to be quite different from the roles of spinal and central adenosine receptors. The present study is aimed at clarifying the role of these receptors in peripheral nociception. To clarify this, studies were done on Swiss mice with adenosine receptor agonists and antagonists. Nociceptive behavior was induced by subcutaneous injection of glutamate (10 µmol) into the ventral surface of the hind paw of mice. Statistical analyses were performed by one-way ANOVA followed by the Student-Newman-Keuls post hoc test. Results showed that intraplantar (i.pl.) administration of N6-cyclohexyl-adenosine (CHA), an adenosine A1 receptor agonist, at 1 or 10 µg/paw significantly reduced glutamate-induced nociception (p<0.01 and p<0.001 vs. vehicle, respectively, n=8-10). In contrast, i.pl. injection of hydrochloride hydrate (CGS21680, an adenosine A2A receptor agonist) (1 µg/paw) induced a significant increase in glutamate-induced nociception compared to the vehicle (p<0.05, n=8), while 4-(-2-[7-amino-2-{2-furyl}{1,2,4}triazolo{2,3-a} {1,3,5}triazin-5-yl-amino]ethyl)phenol (ZM241385, an adenosine A2A receptor antagonist) (20 µg/paw) caused a significant reduction (p<0.05, n=7-8). There were no significant effects on i.pl. administration of four additional adenosine receptor drugs-8-cyclopentyl-1,3-dipropylxanthine (DPCPX, an A1 antagonist, 1-10 µg/paw), N(6)-[2-(3,5-dimethoxyphenyl)-2-(2-methylphenyl)-ethyl]adenosine (DPMA, an A2B agonist, 1-100 µg/paw), alloxazine (an A2B antagonist, 0.1-3 µg/paw), and 2-hexyn-1-yl-N(6)-methyladenosine (HEMADO) (an A3 agonist, 1-100 µg/paw) (p>0.05 vs. vehicle for all tests). We also found that prior administration of DPCPX (3 µg/paw) significantly blocked the anti-nociceptive effect of CHA (1 µg/paw) (p<0.05, n=7-9). Similarly, ZM241385 (20 µg/paw) administered prior to CGS21680 (1 µg/paw) significantly blocked CGS21680-induced exacerbation of nociception (p<0.05, n=8). Finally, inosine (10 and 100 µg/paw), a novel endogenous adenosine A1 receptor agonist recently reported by our research group, was also able to reduce glutamate-induced nociception (p<0.001 vs. vehicle, n=7-8). Interestingly, as an A1 adenosine receptor agonist, the inosine effect was significantly blocked by the A1 antagonist DPCPX (3 µg/paw) (p<0.05, n=7-9) but not by the A2A antagonist ZM241385 (10 µg/paw, p>0.05). In summary, these results demonstrate for the first time that i.pl administration of inosine induces an anti-nociceptive effect, similar to that elicited by CHA and possibly mediated by peripheral adenosine A1 receptor activation. Moreover, our results suggest that peripheral adenosine A2A receptor activation presents a pro-nociceptive effect, exacerbating glutamate-induced nociception independent of inosine-induced anti-nociceptive effects.


Subject(s)
Glutamates , Nociception/drug effects , Pain/chemically induced , Pain/psychology , Peripheral Nervous System/drug effects , Receptors, Purinergic P1/drug effects , Adenosine A1 Receptor Agonists/pharmacology , Adenosine A1 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Female , Foot , Glutamates/administration & dosage , Injections , Inosine/pharmacology , Male , Mice , Pain Measurement/drug effects , Receptor, Adenosine A2A/drug effects
6.
Purinergic Signal ; 16(3): 379-387, 2020 09.
Article in English | MEDLINE | ID: mdl-32725400

ABSTRACT

Parkinson's disease (PD) signs and symptoms regularly include tremor. Interestingly, the nucleoside guanosine (GUO) has already proven to be effective in reducing reserpine-induced tremulous jaw movements (TJMs) in rodent models, thus becoming a promising antiparkinsonian drug. Here, we aimed at revealing the mechanism behind GUO antiparkinsonian efficacy by assessing the role of adenosine A1 and A2A receptors (A1R and A2AR) on GUO-mediated anti-tremor effects in the reserpinized mouse model of PD. Reserpinized mice showed elevated reactive oxygen species (ROS) production and cellular membrane damage in striatal slices assessed ex vivo and GUO treatment reversed ROS production. Interestingly, while the simultaneous administration of sub-effective doses of GUO (5 mg/kg) and SCH58261 (0.01 mg/kg), an A2AR antagonist, precluded reserpine-induced TJMs, these were ineffective on reverting ROS production in ex vivo experiments. Importantly, GUO was able to reduce TJM and ROS production in reserpinized mouse lacking the A2AR, thus suggesting an A2AR-independent mechanism of GUO-mediated effects. Conversely, the administration of DPCPX (0.75 mg/kg), an A1R antagonist, completely abolished both GUO-mediated anti-tremor effects and blockade of ROS production. Overall, these results indicated that GUO anti-tremor and antioxidant effects in reserpinized mice were A1R dependent but A2AR independent, thus suggesting a differential participation of adenosine receptors in GUO-mediated effects.


Subject(s)
Guanosine/therapeutic use , Parkinson Disease, Secondary/metabolism , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A2A/metabolism , Tremor/metabolism , Adenosine A1 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Guanosine/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/drug therapy , Reactive Oxygen Species/metabolism , Tremor/chemically induced , Tremor/drug therapy , Xanthines/pharmacology
7.
Cells ; 9(4)2020 04 23.
Article in English | MEDLINE | ID: mdl-32340145

ABSTRACT

Diabetic nephropathy (DN) is considered the main cause of kidney disease in which myofibroblasts lead to renal fibrosis. Macrophages were recently identified as the major source of myofibroblasts in a process known as macrophage-myofibroblast transition (MMT). Adenosine levels increase during DN and in vivo administration of MRS1754, an antagonist of the A2B adenosine receptor (A2BAR), attenuated glomerular fibrosis (glomerulosclerosis). We aimed to investigate the association between A2BAR and MMT in glomerulosclerosis during DN. Kidneys/glomeruli of non-diabetic, diabetic, and MRS1754-treated diabetic (DM+MRS1754) rats were processed for histopathologic, transcriptomic, flow cytometry, and cellular in vitro analyses. Macrophages were used for in vitro cell migration/transmigration assays and MMT studies. In vivo MRS1754 treatment attenuated the clinical and histopathological signs of glomerulosclerosis in DN rats. Transcriptomic analysis demonstrated a decrease in chemokine-chemoattractants/cell-adhesion genes of monocytes/macrophages in DM+MRS1754 glomeruli. The number of intraglomerular infiltrated macrophages and MMT cells increased in diabetic rats. This was reverted by MRS1754 treatment. In vitro cell migration/transmigration decreased in macrophages treated with MRS1754. Human macrophages cultured with adenosine and/or TGF-ß induced MMT, a process which was reduced by MRS1754. We concluded that pharmacologic blockade of A2BAR attenuated some clinical signs of renal dysfunction and glomerulosclerosis, and decreased intraglomerular macrophage infiltration and MMT in DN rats.


Subject(s)
Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Macrophages/pathology , Monocytes/pathology , Myofibroblasts/pathology , Receptor, Adenosine A2B/metabolism , Acetamides/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Biomarkers/metabolism , Cell Adhesion Molecules/metabolism , Chemokines/metabolism , Chemotactic Factors/pharmacology , Fibrosis , Humans , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Macrophages/drug effects , Macrophages/metabolism , Male , Monocytes/drug effects , Monocytes/metabolism , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Purines/pharmacology , Rats, Sprague-Dawley , Transcription, Genetic/drug effects
8.
J Immunol ; 204(4): 1056-1068, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31900336

ABSTRACT

Hypoxia is a condition that together with low pH, high amounts of reactive oxygen species (ROS), and increased adenosine levels characterize tumor microenvironment. Mast cells (MCs) are part of tumor microenvironment, but the effect of hypoxia on the production of MC-derived cytokines has not been fully described. Using the hypoxia marker pimonidazole in vivo, we found that MCs were largely located in the low-oxygen areas within B16-F1 mice melanoma tumors. In vitro, hypoxia promoted ROS production, a ROS-dependent increase of intracellular calcium, and the production of MCP 1 (CCL-2) in murine bone marrow-derived MCs. Hypoxia-induced CCL-2 production was sensitive to the antioxidant trolox and to nifedipine, a blocker of L-type voltage-dependent Ca2+ channels (LVDCCs). Simultaneously with CCL-2 production, hypoxia caused the ROS-dependent glutathionylation and membrane translocation of the α1c subunit of Cav1.2 LVDCCs. Relationship between ROS production, calcium rise, and CCL-2 synthesis was also observed when cells were treated with H2O2 In vivo, high CCL-2 production was detected on hypoxic zones of melanoma tumors (where tryptase-positive MCs were also found). Pimonidazole and CCL-2 positive staining diminished when B16-F1 cell-inoculated animals were treated with trolox, nifedipine, or the adenosine receptor 2A antagonist KW6002. Our results show that MCs are located preferentially in hypoxic zones of melanoma tumors, hypoxia-induced CCL-2 production in MCs requires calcium rise mediated by glutathionylation and membrane translocation of LVDCCs, and this mechanism of CCL-2 synthesis seems to operate in other cells inside melanoma tumors, with the participation of the adenosine receptor 2A.


Subject(s)
Calcium Channels, L-Type/metabolism , Chemokine CCL2/metabolism , Mast Cells/immunology , Melanoma, Experimental/immunology , Tumor Microenvironment/immunology , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Antioxidants/pharmacology , Biopsy , Calcium Channel Blockers/pharmacology , Cell Hypoxia/drug effects , Cell Hypoxia/immunology , Cell Line, Tumor/transplantation , Chemokine CCL2/immunology , Hydrogen Peroxide/pharmacology , Mast Cells/drug effects , Mast Cells/metabolism , Melanoma, Experimental/pathology , Mice , Reactive Oxygen Species/metabolism , Receptor, Adenosine A2A/metabolism , Tumor Microenvironment/drug effects
9.
Neurochem Res ; 44(4): 787-795, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30610653

ABSTRACT

Caffeine is a bioactive compound worldwide consumed with effect into the brain. Part of its action in reducing incidence or delaying Alzheimer's and Parkinson's diseases symptoms in human is credited to the adenosine receptors properties. However, the impact of caffeine consumption during aging on survival of brain cells remains debatable. This work, we investigated the effect of low-dose of caffeine on the ectonucleotidase activities, adenosine receptors content, and paying particular attention to its pro-survival effect during aging. Male young adult and aged Swiss mice drank water or caffeine (0.3 g/L) ad libitum for 4 weeks. The results showed that long-term caffeine treatment did not unchanged ATP, ADP or AMP hydrolysis in hippocampus when compared to the mice drank water. Nevertheless, the ATP/ADP hydrolysis ratio was higher in young adult (3:1) compared to the aged (1:1) animals regardless of treatment. The content of A1 receptors did not change in any groups of mice, but the content of A2A receptors was reduced in hippocampus of mice that consumed caffeine. Moreover, the cell viability results indicated that aged mice not only had increased pyknotic neurons in the hippocampus but also had reduced damage after caffeine treatment. Overall, these findings indicate a potential neuroprotective effect of caffeine during aging through the adenosinergic system.


Subject(s)
Aging/drug effects , Aging/metabolism , Caffeine/administration & dosage , Neuroprotection/drug effects , Receptor, Adenosine A2A/metabolism , Adenosine A2 Receptor Antagonists/administration & dosage , Aging/pathology , Animals , Cell Survival/drug effects , Cell Survival/physiology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Male , Mice , Neuroprotection/physiology
10.
Cytokine ; 118: 71-79, 2019 06.
Article in English | MEDLINE | ID: mdl-30301599

ABSTRACT

In cancer, the adenosinergic pathway participates in the generation of an immunosuppressive microenvironment and in the promotion of tumor growth through the generation of adenosine (Ado). The present study analyzed the participation of Ado, generated through the functional activity of the cervical cancer (CeCa) pathway in CeCa cells, to induce the expression and secretion of TGF-ß1, as well as the participation of this factor to maintain CD73 expression. Ado concentrations greater than 10 µM were necessary to induce an increase of over 50% in the production and expression of TGF-ß1 in CeCa tumor cells. Blockade of A2AR and A2BR with the specific antagonists, ZM241385 and MRS1754, respectively, strongly reversed the production of TGF-ß1. TGF-ß1 produced by CeCa cells was necessary to maintain CD73 expression because the addition of anti-TGF-ß neutralizing antibodies or the inhibition of TGF-ßRI strongly reversed the expression of CD73 in the CeCa cells. These results suggested a feedback loop in CeCa cells that favors immunosuppressive activity through the production of TGF-ß1 and Ado as well as the autocrine activity of TGF-ß1 and expression of CD73.


Subject(s)
5'-Nucleotidase/metabolism , Adenosine/metabolism , Autocrine Communication/physiology , Transforming Growth Factor beta1/metabolism , Uterine Cervical Neoplasms/metabolism , Acetamides/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Cell Line, Tumor , Female , GPI-Linked Proteins/metabolism , HeLa Cells , Humans , Immunosuppression Therapy/methods , Purines/pharmacology , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A2B/metabolism , Triazines/pharmacology , Triazoles/pharmacology , Tumor Microenvironment/drug effects , Uterine Cervical Neoplasms/drug therapy
11.
Metab Brain Dis ; 32(6): 1919-1927, 2017 12.
Article in English | MEDLINE | ID: mdl-28795281

ABSTRACT

The aim of the present study was to investigate the effects of SCH58261, a selective adenosine A2A receptor antagonist, on striatal toxicity induced by 3-nitropropionic acid (3-NP) in rats. The experimental protocol consisted of 10 administrations (once a day) of SCH58261 (0.01 or 0.05 mg/kg/day, intraperitoneal, i.p.). From 7th to 10th day, 3-NP (20 mg/kg/day, i.p.) was injected 1 h after SCH58261 administration. Twenty-four hours after the last 3-NP injection, the body weight gain, locomotor activity (open-field test), motor coordination (rotarod test), striatal succinate dehydrogenase (SDH) activity and parameters linked to striatal oxidative status were evaluated in rats. The marked body weight loss resulting from 3-NP injections in rats was partially protected by SCH 58261 at both doses. SCH 58261 at the highest dose was effective against impairments on motor coordination and locomotor activity induced by 3-NP. SCH 58261 was unable to restore the inhibition of SDH activity caused by 3-NP. In addition, the increase in striatal reactive species (RS) levels, depletion of reduced glutathione (GSH) content and stimulation of glutathione reductase (GR) activity provoked by 3-NP injections were alleviated by both doses of SCH 58261. The highest dose of SCH 58261 was also effective in attenuating the increase of protein carbonyl levels as well as the inhibition of glutathione peroxidase (GPx) activity in rats exposed to 3-NP. Our results revealed that reduction of oxidative stress in rat striatum by adenosine A2A receptor antagonism contributes for alleviating 3-NP-induced toxicity.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Corpus Striatum/drug effects , Neuroprotective Agents/pharmacology , Nitro Compounds/pharmacology , Oxidative Stress/drug effects , Propionates/pharmacology , Pyrimidines/pharmacology , Triazoles/pharmacology , Animals , Corpus Striatum/metabolism , Glutathione/metabolism , Male , Motor Activity/drug effects , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Rotarod Performance Test
12.
Sci Rep ; 7(1): 1857, 2017 05 12.
Article in English | MEDLINE | ID: mdl-28500295

ABSTRACT

Tardive dyskinesia (TD) is a serious motor side effect that may appear after long-term treatment with neuroleptics and mostly mediated by dopamine D2 receptors (D2Rs). Striatal D2R functioning may be finely regulated by either adenosine A2A receptor (A2AR) or angiotensin receptor type 1 (AT1R) through putative receptor heteromers. Here, we examined whether A2AR and AT1R may oligomerize in the striatum to synergistically modulate dopaminergic transmission. First, by using bioluminescence resonance energy transfer, we demonstrated a physical AT1R-A2AR interaction in cultured cells. Interestingly, by protein-protein docking and molecular dynamics simulations, we described that a stable heterotetrameric interaction may exist between AT1R and A2AR bound to antagonists (i.e. losartan and istradefylline, respectively). Accordingly, we subsequently ascertained the existence of AT1R/A2AR heteromers in the striatum by proximity ligation in situ assay. Finally, we took advantage of a TD animal model, namely the reserpine-induced vacuous chewing movement (VCM), to evaluate a novel multimodal pharmacological TD treatment approach based on targeting the AT1R/A2AR complex. Thus, reserpinized mice were co-treated with sub-effective losartan and istradefylline doses, which prompted a synergistic reduction in VCM. Overall, our results demonstrated the existence of striatal AT1R/A2AR oligomers with potential usefulness for the therapeutic management of TD.


Subject(s)
Protein Multimerization , Receptor, Adenosine A2A/metabolism , Receptor, Angiotensin, Type 1/metabolism , Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/pharmacology , Angiotensin II Type 1 Receptor Blockers/chemistry , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Cells, Cultured , HEK293 Cells , Humans , Mice , Models, Molecular , Protein Binding , Protein Conformation , Receptor, Adenosine A2A/chemistry , Receptor, Angiotensin, Type 1/chemistry , Tardive Dyskinesia/drug therapy , Tardive Dyskinesia/metabolism
13.
Eur J Med Chem ; 136: 487-496, 2017 Aug 18.
Article in English | MEDLINE | ID: mdl-28528302

ABSTRACT

Adenosine receptors are considered as potential targets for drug development against several diseases. The discovery of subtype 2B adenosine receptors role in erythrocyte sickling process proved its importance to neglected diseases such as sickle cell anemia, which affects approximately 29.000 people around the world, but whose treatment is restricted to just one FDA approved drug (hydroxyurea). In order to widen the therapeutic arsenal available to treat sickle cell anemia patients, it is imperative to identify new lead compounds that modify the sickling course and not just its symptoms. In order to accomplish this goal, ligand-based pharmacophore models that differentiate true ligands from decoys and enlighten the structure-activity relationship of known RA2B antagonists were employed screen the lead-like subset of the ZINC database. Following a chemical diversity analysis, 18 compounds were selected for biological evaluation. Among them, one molecule Z1139491704 (pEC50 = 7.77 ± 0.17) has shown better anti-sickling activity than MRS1754 (pEC50 = 7.63 ± 0.12), a commercial RA2B antagonist. Moreover, these compounds exhibited no cytotoxic effect at low micromolar range on mammalian cells. In conclusion, the sound development of validated ligand-based pharmacophore models proved essential to identify novel chemical scaffolds that might be useful to develop anti-sickling drugs.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Anemia, Sickle Cell/drug therapy , Receptor, Adenosine A2B/metabolism , Adenosine A2 Receptor Antagonists/chemical synthesis , Adenosine A2 Receptor Antagonists/chemistry , Animals , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Ligands , Macrophages/drug effects , Mice , Molecular Structure , Structure-Activity Relationship
14.
Curr Neuropharmacol ; 15(8): 1107-1116, 2017 Nov 14.
Article in English | MEDLINE | ID: mdl-28067172

ABSTRACT

BACKGROUND: Virtual methodologies have become essential components of the drug discovery pipeline. Specifically, structure-based drug design methodologies exploit the 3D structure of molecular targets to discover new drug candidates through molecular docking. Recently, dual target ligands of the Adenosine A2A Receptor and Monoamine Oxidase B enzyme have been proposed as effective therapies for the treatment of Parkinson's disease. METHODS: In this paper we propose a structure-based methodology, which is extensively validated, for the discovery of dual Adenosine A2A Receptor/Monoamine Oxidase B ligands. This methodology involves molecular docking studies against both receptors and the evaluation of different scoring functions fusion strategies for maximizing the initial virtual screening enrichment of known dual ligands. RESULTS: The developed methodology provides high values of enrichment of known ligands, which outperform that of the individual scoring functions. At the same time, the obtained ensemble can be translated in a sequence of steps that should be followed to maximize the enrichment of dual target Adenosine A2A Receptor antagonists and Monoamine Oxidase B inhibitors. CONCLUSION: Information relative to docking scores to both targets have to be combined for achieving high dual ligands enrichment. Combining the rankings derived from different scoring functions proved to be a valuable strategy for improving the enrichment relative to single scoring function in virtual screening experiments.


Subject(s)
Adenosine A2 Receptor Antagonists/therapeutic use , Molecular Docking Simulation , Monoamine Oxidase Inhibitors/therapeutic use , Monoamine Oxidase/metabolism , Parkinson Disease/drug therapy , Receptor, Adenosine A2A/metabolism , Adenosine A2 Receptor Antagonists/chemistry , Animals , Binding Sites/drug effects , Humans , Ligands , Monoamine Oxidase Inhibitors/chemistry , Protein Binding/drug effects , Structure-Activity Relationship , User-Computer Interface
15.
Exp Neurol ; 287(Pt 2): 93-101, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27079999

ABSTRACT

Daily acute intermittent hypoxia (dAIH) improves breathing capacity after C2 spinal hemisection (C2HS) in rats. Since C2HS disrupts spinal serotonergic innervation below the injury, adenosine-dependent mechanisms underlie dAIH-induced functional recovery 2weeks post-injury. We hypothesized that dAIH-induced functional recovery converts from an adenosine-dependent to a serotonin-dependent, adenosine-constrained mechanism with chronic injury. Eight weeks post-C2HS, rats began dAIH (10, 5-min episodes, 10.5% O2; 5-min intervals; 7days) followed by AIH 3× per week (3×wAIH) for 8 additional weeks with/without systemic A2A receptor inhibition (KW6002) on each AIH exposure day. Tidal volume (VT) and bilateral diaphragm (Dia) and T2 external intercostal motor activity were assessed in unanesthetized rats breathing air and during maximum chemoreflex stimulation (MCS: 7% CO2, 10.5% O2). Nine weeks post-C2HS, dAIH increased VT versus time controls (p<0.05), an effect enhanced by KW6002 (p<0.05). dAIH increased bilateral Dia activity (p<0.05), and KW6002 enhanced this effect in contralateral (p<0.05) and ipsilateral Dia activity (p<0.001), but not T2 inspiratory activity. Functional benefits of combined AIH plus systemic A2A receptor inhibition were maintained for 4weeks. Thus, in rats with chronic injuries: 1) dAIH improves VT and bilateral diaphragm activity; 2) VT recovery is enhanced by A2A receptor inhibition; and 3) functional recovery with A2A receptor inhibition and AIH "reminders" last 4weeks. Combined dAIH and A2A receptor inhibition may be a simple, safe, and effective strategy to accelerate/enhance functional recovery of breathing capacity in patients with respiratory impairment from chronic spinal injury.


Subject(s)
Cervical Vertebrae , Hypoxia , Maximal Voluntary Ventilation/physiology , Receptors, Adenosine A2/metabolism , Recovery of Function/physiology , Respiration Disorders/etiology , Respiration Disorders/therapy , Spinal Cord Injuries/complications , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/therapeutic use , Animals , Diaphragm/drug effects , Disease Models, Animal , Functional Laterality/drug effects , Functional Laterality/physiology , Hypercapnia/physiopathology , Male , Maximal Voluntary Ventilation/drug effects , Motor Activity/drug effects , Muscle Contraction/drug effects , Purines/pharmacology , Purines/therapeutic use , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Time Factors , Vital Capacity/drug effects , Vital Capacity/physiology
16.
PLoS One ; 11(11): e0167236, 2016.
Article in English | MEDLINE | ID: mdl-27893847

ABSTRACT

Chronic sleep restriction induces blood-brain barrier disruption and increases pro-inflammatory mediators in rodents. Those inflammatory mediators may modulate the blood-brain barrier and constitute a link between sleep loss and blood-brain barrier physiology. We propose that adenosine action on its A2A receptor may be modulating the blood-brain barrier dynamics in sleep-restricted rats. We administrated a selective A2A adenosine receptor antagonist (SCH58261) in sleep-restricted rats at the 10th day of sleep restriction and evaluated the blood-brain barrier permeability to dextrans coupled to fluorescein (FITC-dextrans) and Evans blue. In addition, we evaluated by western blot the expression of tight junction proteins (claudin-5, occludin, ZO-1), adherens junction protein (E-cadherin), A2A adenosine receptor, adenosine-synthesizing enzyme (CD73), and neuroinflammatory markers (Iba-1 and GFAP) in the cerebral cortex, hippocampus, basal nuclei and cerebellar vermis. Sleep restriction increased blood-brain barrier permeability to FITC-dextrans and Evans blue, and the effect was reverted by the administration of SCH58261 in almost all brain regions, excluding the cerebellum. Sleep restriction increased the expression of A2A adenosine receptor only in the hippocampus and basal nuclei without changing the expression of CD73 in all brain regions. Sleep restriction reduced the expression of tight junction proteins in all brain regions, except in the cerebellum; and SCH58261 restored the levels of tight junction proteins in the cortex, hippocampus and basal nuclei. Finally, sleep restriction induced GFAP and Iba-1 overexpression that was attenuated with the administration of SCH58261. These data suggest that the action of adenosine on its A2A receptor may have a crucial role in blood-brain barrier dysfunction during sleep loss probably by direct modulation of brain endothelial cell permeability or through a mechanism that involves gliosis with subsequent inflammation and increased blood-brain barrier permeability.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Blood-Brain Barrier/drug effects , Cell Membrane Permeability/drug effects , Receptor, Adenosine A2A/chemistry , Sleep Deprivation/physiopathology , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Male , Rats , Rats, Wistar
17.
Life Sci ; 166: 92-99, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27729268

ABSTRACT

The adenosine A2b receptor is a G-protein coupled receptor. Its activation occurs with high extracellular adenosine concentration, for example in inflammation or hypoxia. These conditions are generated in the tumor environment. Studies show that A2b receptor is overexpressed in various tumor lines and biopsies from patients with different cancers. This suggests that A2b receptor can be used by tumor cells to promote progression. Thus A2b participates in different events, such as angiogenesis and metastasis, besides exerting immunomodulatory effects that protect tumor cells. Therefore, adenosine A2b receptor appears as an interesting therapeutic target for cancer treatment.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Neoplasms/pathology , Receptor, Adenosine A2B/genetics , Up-Regulation , Adenosine/analysis , Adenosine/genetics , Adenosine/immunology , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/therapeutic use , Animals , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immune Tolerance/drug effects , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Neoplasms/immunology , Receptor, Adenosine A2B/analysis , Receptor, Adenosine A2B/immunology , Tumor Microenvironment/drug effects , Up-Regulation/drug effects
18.
Acta Cir Bras ; 31(2): 133-7, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26959623

ABSTRACT

PURPOSE: To investigate the role of adenosine A2A receptors on 6-OHDA-induced motor disorder in rat. METHODS: In order to induce experimental model of Parkinson's disease, 6-hydoxydopamine (8 µg/rat) was injected unilaterally into the SNc. After three weeks as a recovery period, 6-OHDA-induced bradykinesia and balance disturbances were assessed by using beam traversal test 10, 30 and 60 minutes after intraperitoneal injections of the drugs (caffeine, SCH58261). RESULTS: The results showed that 6-OHDA (8 µg/rat, Intra-SNc) induced motor disorders of Parkinson's disease and increased elapsed time in the beam test (p<0.001). Injection of caffeine (30 mg/kg, i.p.) and SCH58261 (2 mg/kg, i.p.) attenuated elapsed time on beam (p<0.01 and p<0.001). We showed that acute administration of caffeine and SCH 58261 can improve the 6-OHDA-induced bradykinesia and motor disturbance. CONCLUSION: Adenosine A2AR antagonists improve 6-OHDA-motor deficit and this effect seems to be mediated by the inhibition of A2A presynaptic receptors in substantia nigra pars compacta.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Caffeine/pharmacology , Oxidopamine/adverse effects , Parkinson Disease, Secondary/chemically induced , Purinergic P1 Receptor Antagonists/pharmacology , Animals , Disease Models, Animal , Hypokinesia/chemically induced , Male , Motor Activity/drug effects , Motor Disorders/chemically induced , Rats, Wistar , Time and Motion Studies
19.
Acta cir. bras. ; 31(2): 133-137, fev. 2016. graf
Article in English | VETINDEX | ID: vti-20440

ABSTRACT

PURPOSE:To investigate the role of adenosine A2A receptors on 6-OHDA-induced motor disorder in rat.METHODS:In order to induce experimental model of Parkinson's disease, 6-hydoxydopamine (8 μg/rat) was injected unilaterally into the SNc. After three weeks as a recovery period, 6-OHDA-induced bradykinesia and balance disturbances were assessed by using beam traversal test 10, 30 and 60 minutes after intraperitoneal injections of the drugs (caffeine, SCH58261).RESULTS:The results showed that 6-OHDA (8 μg/rat, Intra-SNc) induced motor disorders of Parkinson's disease and increased elapsed time in the beam test (p<0.001). Injection of caffeine (30 mg/kg, i.p.) and SCH58261 (2 mg/kg, i.p.) attenuated elapsed time on beam (p<0.01 and p<0.001). We showed that acute administration of caffeine and SCH 58261 can improve the 6-OHDA-induced bradykinesia and motor disturbance.CONCLUSION:Adenosine A2AR antagonists improve 6-OHDA-motor deficit and this effect seems to be mediated by the inhibition of A2A presynaptic receptors in substantia nigra pars compacta.(AU)


Subject(s)
Animals , Rats , Parkinson Disease/veterinary , Oxidopamine/administration & dosage , Adenosine A2 Receptor Antagonists/analysis , Motor Disorders
20.
Acta cir. bras ; Acta cir. bras;31(2): 133-137, Feb. 2016. graf
Article in English | LILACS | ID: lil-775559

ABSTRACT

PURPOSE: To investigate the role of adenosine A2A receptors on 6-OHDA-induced motor disorder in rat. METHODS: In order to induce experimental model of Parkinson's disease, 6-hydoxydopamine (8 μg/rat) was injected unilaterally into the SNc. After three weeks as a recovery period, 6-OHDA-induced bradykinesia and balance disturbances were assessed by using beam traversal test 10, 30 and 60 minutes after intraperitoneal injections of the drugs (caffeine, SCH58261). RESULTS: The results showed that 6-OHDA (8 μg/rat, Intra-SNc) induced motor disorders of Parkinson's disease and increased elapsed time in the beam test (p<0.001). Injection of caffeine (30 mg/kg, i.p.) and SCH58261 (2 mg/kg, i.p.) attenuated elapsed time on beam (p<0.01 and p<0.001). We showed that acute administration of caffeine and SCH 58261 can improve the 6-OHDA-induced bradykinesia and motor disturbance. CONCLUSION: Adenosine A2AR antagonists improve 6-OHDA-motor deficit and this effect seems to be mediated by the inhibition of A2A presynaptic receptors in substantia nigra pars compacta.


Subject(s)
Animals , Male , Parkinson Disease, Secondary/chemically induced , Caffeine/pharmacology , Oxidopamine/adverse effects , Purinergic P1 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Time and Motion Studies , Rats, Wistar , Hypokinesia/chemically induced , Disease Models, Animal , Motor Disorders/chemically induced , Motor Activity/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL