Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
Add more filters

Publication year range
1.
Purinergic Signal ; 20(1): 35-45, 2024 Feb.
Article in English | MEDLINE | ID: mdl-36918461

ABSTRACT

Interest has been focused in recent years on the analgesic effects exerted by adenosine and its receptors, A1, A2A, A2B, and A3 adenosine receptor (AR) subtypes, in different in vivo models of chronic pain. In particular, it was demonstrated that selective A3AR agonists reduced pro-nociceptive N-type Ca2+ channels in dorsal root ganglion (DRG) neurons isolated from rats and, by this mechanism, inhibit post inflammatory visceral hypersensitivity. In the present study, we investigate the effect of a previously reported irreversibly binding A3AR agonist, ICBM, on Ca2+ currents (ICa) in rat DRG neurons. Present data demonstrate that ICBM, an isothiocyanate derivative designed for covalent binding to the receptor, concentration-dependently inhibits ICa. This effect is irreversible, since it persists after drug removal, differently from the prototypical A3AR agonist, Cl-IB-MECA. ICBM pre-exposure inhibits the effect of a subsequent Cl-IB-MECA application. Thus, covalent A3AR agonists such as ICBM may represent an innovative, beneficial, and longer-lasting strategy to achieve efficacious chronic pain control versus commonly used, reversible, A3AR agonists. However, the possible limitations of this drug and other covalent drugs may be, for example, a characteristic adverse effect profile, suggesting that more pre-clinical studies are needed.


Subject(s)
Chronic Pain , Ganglia, Spinal , Rats , Animals , Ganglia, Spinal/metabolism , Chronic Pain/metabolism , Neurons/metabolism , Adenosine/metabolism , Receptors, Purinergic P1/metabolism , Receptor, Adenosine A3/metabolism , Adenosine A3 Receptor Agonists/pharmacology
2.
Int J Mol Sci ; 25(11)2024 May 25.
Article in English | MEDLINE | ID: mdl-38891948

ABSTRACT

Cardiovascular diseases (CVDs), particularly heart failure, are major contributors to early mortality globally. Heart failure poses a significant public health problem, with persistently poor long-term outcomes and an overall unsatisfactory prognosis for patients. Conventionally, treatments for heart failure have focused on lowering blood pressure; however, the development of more potent therapies targeting hemodynamic parameters presents challenges, including tolerability and safety risks, which could potentially restrict their clinical effectiveness. Adenosine has emerged as a key mediator in CVDs, acting as a retaliatory metabolite produced during cellular stress via ATP metabolism, and works as a signaling molecule regulating various physiological processes. Adenosine functions by interacting with different adenosine receptor (AR) subtypes expressed in cardiac cells, including A1AR, A2AAR, A2BAR, and A3AR. In addition to A1AR, A3AR has a multifaceted role in the cardiovascular system, since its activation contributes to reducing the damage to the heart in various pathological states, particularly ischemic heart disease, heart failure, and hypertension, although its role is not as well documented compared to other AR subtypes. Research on A3AR signaling has focused on identifying the intricate molecular mechanisms involved in CVDs through various pathways, including Gi or Gq protein-dependent signaling, ATP-sensitive potassium channels, MAPKs, and G protein-independent signaling. Several A3AR-specific agonists, such as piclidenoson and namodenoson, exert cardioprotective impacts during ischemia in the diverse animal models of heart disease. Thus, modulating A3ARs serves as a potential therapeutic approach, fueling considerable interest in developing compounds that target A3ARs as potential treatments for heart diseases.


Subject(s)
Heart Diseases , Receptor, Adenosine A3 , Signal Transduction , Humans , Animals , Signal Transduction/drug effects , Receptor, Adenosine A3/metabolism , Heart Diseases/metabolism , Heart Diseases/drug therapy , Adenosine A3 Receptor Agonists/therapeutic use , Adenosine A3 Receptor Agonists/pharmacology , Adenosine/metabolism
3.
FASEB J ; 35(4): e21211, 2021 04.
Article in English | MEDLINE | ID: mdl-33710641

ABSTRACT

Organization of G protein-coupled receptors at the plasma membrane has been the focus of much recent attention. Advanced microscopy techniques have shown that these receptors can be localized to discrete microdomains and reorganization upon ligand activation is crucial in orchestrating their signaling. Here, we have compared the membrane organization and downstream signaling of a mutant (R108A, R3.50A) of the adenosine A3 receptor (A3 AR) to that of the wild-type receptor. Fluorescence Correlation Spectroscopy (FCS) studies with a fluorescent agonist (ABEA-X-BY630) demonstrated that both wild-type and mutant receptors bind agonist with high affinity but in subsequent downstream signaling assays the R108A mutation abolished agonist-mediated inhibition of cAMP production and ERK phosphorylation. In further FCS studies, both A3 AR and A3 AR R108A underwent similar agonist-induced increases in receptor density and molecular brightness which were accompanied by a decrease in membrane diffusion after agonist treatment. Using bimolecular fluorescence complementation, experiments showed that the R108A mutant retained the ability to recruit ß-arrestin and these receptor/arrestin complexes displayed similar membrane diffusion and organization to that observed with wild-type receptors. These data demonstrate that effective G protein signaling is not a prerequisite for agonist-stimulated ß-arrestin recruitment and membrane reorganization of the A3 AR.


Subject(s)
Adenosine A3 Receptor Agonists/pharmacology , Adenosine/analogs & derivatives , Boron Compounds/pharmacology , GTP-Binding Proteins/metabolism , Receptor, Adenosine A3/metabolism , Adenosine/pharmacology , Animals , Arrestin/metabolism , CHO Cells , Cricetulus , Gene Expression Regulation/drug effects , Mutation , Protein Binding , Receptor, Adenosine A3/genetics
4.
Molecules ; 27(6)2022 03 15.
Article in English | MEDLINE | ID: mdl-35335254

ABSTRACT

Ligands of the Gi protein-coupled adenosine A3 receptor (A3R) are receiving increasing interest as attractive therapeutic tools for the treatment of a number of pathological conditions of the central and peripheral nervous systems (CNS and PNS, respectively). Their safe pharmacological profiles emerging from clinical trials on different pathologies (e.g., rheumatoid arthritis, psoriasis and fatty liver diseases) confer a realistic translational potential to these compounds, thus encouraging the investigation of highly selective agonists and antagonists of A3R. The present review summarizes information on the effect of latest-generation A3R ligands, not yet available in commerce, obtained by using different in vitro and in vivo models of various PNS- or CNS-related disorders. This review places particular focus on brain ischemia insults and colitis, where the prototypical A3R agonist, Cl-IB-MECA, and antagonist, MRS1523, have been used in research studies as reference compounds to explore the effects of latest-generation ligands on this receptor. The advantages and weaknesses of these compounds in terms of therapeutic potential are discussed.


Subject(s)
Adenosine A3 Receptor Agonists , Arthritis, Rheumatoid , Adenosine A3 Receptor Agonists/pharmacology , Adenosine A3 Receptor Agonists/therapeutic use , Arthritis, Rheumatoid/drug therapy , Humans , Ligands , Peripheral Nervous System , Receptors, Purinergic P1
5.
Molecules ; 27(12)2022 Jun 08.
Article in English | MEDLINE | ID: mdl-35744805

ABSTRACT

The A3 adenosine receptor (A3AR) is overexpressed in pathological human cells. Piclidenoson and namodenoson are A3AR agonists with high affinity and selectivity to A3AR. Both induce apoptosis of cancer and inflammatory cells via a molecular mechanism entailing deregulation of the Wnt and the NF-κB signaling pathways. Our company conducted phase I studies showing the safety of these 2 molecules. In the phase II studies in psoriasis patients, piclidenoson was safe and demonstrated efficacy manifested in significant improvements in skin lesions. Namodenoson is currently being developed to treat liver cancer, where prolonged overall survival was observed in patients with advanced liver disease and a Child-Pugh B score of 7. A pivotal phase III study in this patient population has been approved by the FDA and the EMA and is currently underway. Namodenoson is also being developed to treat non-alcoholic steatohepatitis (NASH). A Phase IIa study has been successfully concluded and showed that namodenoson has anti-inflammatory, anti-fibrosis, and anti-steatosis effects. A phase IIb study in NASH is currently enrolling patients. In conclusion, A3AR agonists are promising drug candidates in advanced stages of clinical development and demonstrate safety and efficacy in their targeted indications.


Subject(s)
Non-alcoholic Fatty Liver Disease , Adenosine A3 Receptor Agonists/pharmacology , Adenosine A3 Receptor Agonists/therapeutic use , Anti-Inflammatory Agents/pharmacology , Clinical Trials, Phase II as Topic , Humans , NF-kappa B/metabolism , Receptor, Adenosine A3/metabolism , Signal Transduction
6.
Pharmacol Res ; 170: 105731, 2021 08.
Article in English | MEDLINE | ID: mdl-34157422

ABSTRACT

Psoriasis is a chronic and relapsing inflammatory skin disease lacking a cure that affects approximately 2% of the population. Defective keratinocyte proliferation and differentiation, and aberrant immune responses are major factors in its pathogenesis. Available treatments for moderate to severe psoriasis are directed to immune system causing systemic immunosuppression over time, and thus concomitant serious side effects (i.e. infections and cancer) may appear. In recent years, the Gi protein-coupled A3 receptor (A3R) for adenosine has been suggested as a novel and very promising therapeutic target for psoriasis. Accordingly, selective, and high affinity A3R agonists are known to induce robust anti-inflammatory effects in animal models of autoimmune inflammatory diseases. Here, we demonstrated the efficacy of a selective A3R agonist, namely MRS5698, in preventing the psoriatic-like phenotype in the IL-23 mouse model of psoriasis. Subsequently, we photocaged this molecule with a coumarin moiety to yield the first photosensitive A3R agonist, MRS7344, which in photopharmacological experiments prevented the psoriatic-like phenotype in the IL-23 animal model. Thus, we have demonstrated the feasibility of using a non-invasive, site-specific, light-directed approach to psoriasis treatment.


Subject(s)
Adenosine A3 Receptor Agonists/pharmacology , Adenosine/analogs & derivatives , Photochemotherapy , Psoriasis/prevention & control , Receptor, Adenosine A3/drug effects , Skin/drug effects , Adenosine/pharmacology , Animals , Disease Models, Animal , Interleukin-23 , Ligands , Psoriasis/immunology , Psoriasis/metabolism , Psoriasis/pathology , Receptor, Adenosine A3/metabolism , Signal Transduction , Skin/immunology , Skin/metabolism , Skin/pathology
7.
Int J Mol Sci ; 22(11)2021 May 26.
Article in English | MEDLINE | ID: mdl-34073488

ABSTRACT

Kidney fibrosis is the final outcome of chronic kidney disease (CKD). Adenosine plays a significant role in protection against cellular damage by activating four subtypes of adenosine receptors (ARs), A1AR, A2AAR, A2BAR, and A3AR. A2AAR agonists protect against inflammation, and A3AR antagonists effectively inhibit the formation of fibrosis. Here, we showed for the first time that LJ-4459, a newly synthesized dual-acting ligand that is an A2AAR agonist and an A3AR antagonist, prevents the progression of tubulointerstitial fibrosis. Unilateral ureteral obstruction (UUO) surgery was performed on 6-week-old male C57BL/6 mice. LJ-4459 (1 and 10 mg/kg) was orally administered for 7 days, started at 1 day before UUO surgery. Pretreatment with LJ-4459 improved kidney morphology and prevented the progression of tubular injury as shown by decreases in urinary kidney injury molecular-1 (KIM-1) and neutrophil gelatinase-associated lipocalin (NGAL) excretion. Obstruction-induced tubulointerstitial fibrosis was attenuated by LJ-4459, as shown by a decrease in fibrotic protein expression in the kidney. LJ-4459 also inhibited inflammation and oxidative stress in the obstructed kidney, with reduced macrophage infiltration, reduced levels of pro-inflammatory cytokines, as well as reduced levels of reactive oxygen species (ROS). These data demonstrate that LJ-4459 has potential as a therapeutic agent against the progression of tubulointerstitial fibrosis.


Subject(s)
Adenosine A3 Receptor Agonists/pharmacology , Kidney Diseases/drug therapy , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A3/metabolism , Ureteral Obstruction/drug therapy , Adenosine A3 Receptor Agonists/chemical synthesis , Adenosine A3 Receptor Agonists/chemistry , Animals , Fibrosis , Kidney Diseases/etiology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Ligands , Male , Mice , Ureteral Obstruction/complications , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology
8.
J Neurosci ; 39(36): 7206-7217, 2019 09 04.
Article in English | MEDLINE | ID: mdl-31315945

ABSTRACT

Adenosine 2A receptor (A2AR)-containing indirect medium spiny neurons (iMSNs) in the dorsomedial striatum (DMS) contribute to reward-seeking behaviors. However, those roles for ethanol-seeking behaviors remain unknown. To investigate ethanol-seeking behaviors, we used an ethanol-containing reward (10% ethanol and 10% sucrose solution; 10E10S). Upon conditioning with 10E10S, mice that initially only preferred 10% sucrose, not 10E10S, showed a stronger preference for 10E10S. Then, we investigated whether the manipulation of the DMS-external globus pallidus (GPe) iMSNs circuit alters the ethanol-containing reward (10E10S) seeking behaviors using the combination of pharmacologic and optogenetic approaches. DMS A2AR activation dampened operant conditioning-induced ethanol-containing reward, whereas A2AR antagonist abolished the effects of the A2AR agonist and restored ethanol-containing reward-seeking. Moreover, pre-ethanol exposure potentiated the A2AR-dependent reward-seeking. Interestingly, mice exhibiting ethanol-containing reward-seeking showed the reduction of the DMS iMSNs activity, suggesting that disinhibiting iMSNs decreases reward-seeking behaviors. In addition, we found that A2AR activation reversed iMSNs neural activity in the DMS. Similarly, optogenetic stimulation of the DMS-GPe iMSNs reduced ethanol-containing reward-seeking, whereas optogenetic inhibition of the DMS-GPe iMSNs reversed this change. Together, our study demonstrates that DMS A2AR and iMSNs regulate ethanol-containing reward-seeking behaviors.SIGNIFICANCE STATEMENT Our findings highlight the mechanisms of how operant conditioning develops the preference of ethanol-containing conditioned reward. Mice exhibiting ethanol-containing reward-seeking showed a reduction of the indirect medium spiny neuronal activity in the dorsomedial striatum. Pharmacological activation of adenosine A2A receptor (A2AR) or optogenetic activation of indirect medium spiny neurons dampened operant conditioned ethanol-containing reward-seeking, whereas inhibiting this neuronal activity restored ethanol-containing reward-seeking. Furthermore, repeated intermittent ethanol exposure potentiated A2AR-dependent reward-seeking. Therefore, our finding suggests that A2AR-containing indirect medium spiny neuronal activation reduces ethanol-containing reward-seeking, which may provide a potential therapeutic target for alcohol use disorder.


Subject(s)
Alcoholism/physiopathology , Globus Pallidus/physiopathology , Neurons/physiology , Reward , Adenosine A3 Receptor Agonists/pharmacology , Adenosine A3 Receptor Antagonists/pharmacology , Animals , Conditioning, Operant , Ethanol/pharmacology , Globus Pallidus/cytology , Globus Pallidus/physiology , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Receptors, Adenosine A2/metabolism
9.
J Cell Physiol ; 235(3): 2441-2451, 2020 03.
Article in English | MEDLINE | ID: mdl-31556103

ABSTRACT

Acupuncture has many advantages in the treatment of certain diseases as opposed to drug therapy. Besides, adenosine has been revealed to affect cellular progression including proliferation. Therefore, this study aimed at exploring the mechanism involving acupuncture stress and adenosine in fibroblast proliferation. The fibroblasts from fascia tissues of the acupoint area (Zusanli) were stimulated by different levels of stress, different concentrations of adenosine, and agonist or antagonist of A3 receptor (A3 R) to investigate the effect of stress stimulation, adenosine, and adenosine-A3 R inhibition on fibroblasts. Then, the fibroblasts were treated with stress stimulation of 200 kPa or/and mitogen-activated protein kinase (MAPK) blocker. We revealed that stress stimulation and the binding of adenosine and A3 R promoted fibroblast proliferation in the fascial tissue, increased the expression of immune-related factors, adenosine and A3 R, and activated the MAPK signaling pathway. MAPK signaling pathway also directly affected the expression of adenosine, A3 R, and immune-related factors. Stress stimulation and adenosine treatment upregulated A3 R expression, and then activated the MAPK signaling pathway, which could in turn upregulate expression of adenosine, A3 R and immune-related factors, and promote cell proliferation. Adenosine is shown to form a positive feedback loop with the MAPK signaling pathway. Collectively, stress stimulation in vitro induces the increase of adenosine in fibroblasts through the energy metabolism and activation of the MAPK signaling pathway through A3 R, ultimately promoting fibroblast proliferation.


Subject(s)
Acupuncture/methods , Adenosine/genetics , Energy Metabolism/genetics , Receptor, Adenosine A3/genetics , Acupuncture Points , Adenosine A3 Receptor Agonists/pharmacology , Animals , Cell Proliferation/genetics , Fibroblasts/metabolism , Humans , MAP Kinase Signaling System/genetics , Microscopy, Confocal , Primary Cell Culture , Rats , Signal Transduction/drug effects
10.
Int J Mol Sci ; 21(19)2020 Sep 30.
Article in English | MEDLINE | ID: mdl-33007835

ABSTRACT

Glaucoma is a progressive chronic retinal degenerative disease and a leading cause of global irreversible blindness, characterized by optic nerve damage and retinal ganglion cell (RGC) death. Elevated intraocular pressure (IOP) is a main risk factor of glaucoma. Neuroinflammation plays an important role in glaucoma. We have been demonstrating that elevated pressure triggers microglia reactivity that contribute to the loss of RGCs. Adenosine, acting on adenosine receptors, is a crucial modulator of microglia phenotype. Microglia express all adenosine receptors. Previously, we demonstrated that the activation of adenosine A3 receptor (A3R) affords protection to the retina, including RGCs, unveiling the possibility for a new strategy for glaucoma treatment. Since microglial cells express A3R, we now studied the ability of a selective A3R agonist (2-Cl-IB-MECA) in controlling microglia reactivity induced by elevated hydrostatic pressure (EHP), used to mimic elevated IOP. The activation of A3R reduced EHP-induced inducible nitric oxide synthase (iNOS) expression, microglia migration and phagocytosis in BV-2 cells. In retinal microglia, proliferation and phagocytosis elicited by EHP were also decreased by A3R activation. This work demonstrates that 2-Cl-IB-MECA, the selective agonist of A3R, is able to hinder microglia reactivity, suggesting that A3R agonists could afford protection against glaucomatous degeneration through the control of neuroinflammation.


Subject(s)
Adenosine A3 Receptor Agonists/pharmacology , Adenosine/analogs & derivatives , Glaucoma/drug therapy , Receptor, Adenosine A3/genetics , Adenosine/genetics , Adenosine/pharmacology , Animals , Cell Death/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Glaucoma/genetics , Glaucoma/pathology , Humans , Intraocular Pressure/drug effects , Microglia/drug effects , Optic Nerve/drug effects , Optic Nerve/pathology , Optic Nerve Injuries/drug therapy , Optic Nerve Injuries/genetics , Optic Nerve Injuries/pathology , Phagocytosis/drug effects , Rats , Retinal Degeneration/drug therapy , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology
11.
Mol Pharmacol ; 96(2): 180-192, 2019 08.
Article in English | MEDLINE | ID: mdl-31127007

ABSTRACT

P-glycoprotein (P-gp) is a multidrug transporter that is expressed on the luminal surface of epithelial cells in the kidney, intestine, bile-canalicular membrane in the liver, blood-brain barrier, and adrenal gland. This transporter uses energy of ATP hydrolysis to efflux from cells a variety of structurally dissimilar hydrophobic and amphipathic compounds, including anticancer drugs. In this regard, understanding the interaction with P-gp of drug entities in development is important and highly recommended in current US Food and Drug Administration guidelines. Here we tested the P-gp interaction of some A3 adenosine receptor agonists that are being developed for the treatment of chronic diseases, including rheumatoid arthritis, psoriasis, chronic pain, and hepatocellular carcinoma. Biochemical assays of the ATPase activity of P-gp and by photolabeling P-gp with its transport substrate [125I]-iodoarylazidoprazosin led to the identification of rigidified (N)-methanocarba nucleosides (i.e., compound 3 as a stimulator and compound 8 as a partial inhibitor of P-gp ATPase activity). Compound 8 significantly inhibited boron-dipyrromethene (BODIPY)-verapamil transport mediated by human P-gp (IC50 2.4 ± 0.6 µM); however, the BODIPY-conjugated derivative of 8 (compound 24) was not transported by P-gp. In silico docking of compounds 3 and 8 was performed using the recently solved atomic structure of paclitaxel (Taxol)-bound human P-gp. Molecular modeling studies revealed that both compounds 3 and 8 bind in the same region of the drug-binding pocket as Taxol. Thus, this study indicates that nucleoside derivatives can exhibit varied modulatory effects on P-gp activity, depending on structural functionalization. SIGNIFICANCE STATEMENT: Certain A3 adenosine receptor agonists are being developed for the treatment of chronic diseases. The goal of this study was to test the interaction of these agonists with the human multidrug resistance-linked transporter P-glycoprotein (P-gp). ATPase and photolabeling assays demonstrated that compounds with rigidified (N)-methanocarba nucleosides inhibit the activity of P-gp; however, a fluorescent derivative of one of the compounds was not transported by P-gp. Furthermore, molecular docking studies revealed that the binding site for these compounds overlaps with the site for paclitaxel in the drug-binding pocket. These results suggest that nucleoside derivatives, depending on structural functionalization, can modulate the function of P-gp.


Subject(s)
Adenosine A3 Receptor Agonists/pharmacology , ATP Binding Cassette Transporter, Subfamily B/chemistry , ATP Binding Cassette Transporter, Subfamily B/metabolism , Adenosine A3 Receptor Agonists/chemistry , Azides/metabolism , Binding Sites , HeLa Cells , Humans , Models, Molecular , Molecular Docking Simulation , Paclitaxel/chemistry , Paclitaxel/pharmacology , Prazosin/analogs & derivatives , Prazosin/metabolism , Structure-Activity Relationship , Verapamil/chemistry , Verapamil/pharmacology
12.
Bioorg Med Chem ; 27(15): 3328-3333, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31230970

ABSTRACT

In recent years, special attention has been paid to the A3 adenosine receptor (A3AR) as a possible pharmacological target to treat intestinal inflammation. In this work, it was set up a novel method to quantify the concentration of a promising anti-inflammatory agent inside and outside of intestinal barrier using the everted gut sac technique. The compound chosen for the present study is one of the most potent and selective A3AR agonist reported so far, named AR 170 (N6-methyl-2-phenylethynyl-5'-N-methylcarboxamidoadenosine). In order to evaluate the intestinal absorption of AR 170 the radioligand binding assay in comparison with HPLC-DAD was used. Results showed that the compound is absorbed via passive diffusion by paracellular pathway. The concentrations determined in the serosal (inside the sac) fluid by radioligand binding assay are in good agreement with those obtained through the widely used HPLC/MS protocol, demonstrating the reliability of the method. It is worthwhile to note that the radioligand binding assay allows detecting very low concentrations of analyte, thus offering an excellent tool to measure the intestinal absorption of receptor ligands. Moreover, the AR 170 quantity outside the gut sac and the interaction with A3AR could presuppose good topical anti-inflammatory effects of this compound.


Subject(s)
Adenosine A3 Receptor Agonists/pharmacology , Adenosine/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Intestinal Absorption/drug effects , Intestinal Mucosa/drug effects , Receptor, Adenosine A3/metabolism , Adenosine/analysis , Adenosine/chemistry , Adenosine A3 Receptor Agonists/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , CHO Cells , Cells, Cultured , Cricetulus , Dose-Response Relationship, Drug , Intestinal Mucosa/metabolism , Ligands , Male , Molecular Structure , Rats , Rats, Wistar , Structure-Activity Relationship
13.
Med Res Rev ; 38(4): 1031-1072, 2018 07.
Article in English | MEDLINE | ID: mdl-28682469

ABSTRACT

The A3 adenosine receptor (A3 AR) subtype is a novel, promising therapeutic target for inflammatory diseases, such as rheumatoid arthritis (RA) and psoriasis, as well as liver cancer. A3 AR is coupled to inhibition of adenylyl cyclase and regulation of mitogen-activated protein kinase (MAPK) pathways, leading to modulation of transcription. Furthermore, A3 AR affects functions of almost all immune cells and the proliferation of cancer cells. Numerous A3 AR agonists, partial agonists, antagonists, and allosteric modulators have been reported, and their structure-activity relationships (SARs) have been studied culminating in the development of potent and selective molecules with drug-like characteristics. The efficacy of nucleoside agonists may be suppressed to produce antagonists, by structural modification of the ribose moiety. Diverse classes of heterocycles have been discovered as selective A3 AR blockers, although with large species differences. Thus, as a result of intense basic research efforts, the outlook for development of A3 AR modulators for human therapeutics is encouraging. Two prototypical selective agonists, N6-(3-Iodobenzyl)adenosine-5'-N-methyluronamide (IB-MECA; CF101) and 2-chloro-N6-(3-iodobenzyl)-adenosine-5'-N-methyluronamide (Cl-IB-MECA; CF102), have progressed to advanced clinical trials. They were found safe and well tolerated in all preclinical and human clinical studies and showed promising results, particularly in psoriasis and RA, where the A3 AR is both a promising therapeutic target and a biologically predictive marker, suggesting a personalized medicine approach. Targeting the A3 AR may pave the way for safe and efficacious treatments for patient populations affected by inflammatory diseases, cancer, and other conditions.


Subject(s)
Adenosine A3 Receptor Agonists/pharmacology , Arthritis, Rheumatoid/drug therapy , Carcinoma, Hepatocellular/drug therapy , Inflammation/drug therapy , Liver Neoplasms/drug therapy , Psoriasis/drug therapy , Receptor, Adenosine A3/metabolism , Allosteric Site , Animals , Clinical Trials as Topic , Crystallography, X-Ray , Humans , Immune System , Mice , Molecular Dynamics Simulation , Rats , Structure-Activity Relationship
14.
J Cell Physiol ; 233(3): 2032-2057, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28233320

ABSTRACT

Tumor cells overcome anti-tumor responses in part through immunosuppressive mechanisms. There are several immune modulatory mechanisms. Among them, adenosine is an important factor which is generated by both cancer and immune cells in tumor microenvironment to suppress anti-tumor responses. Two cell surface expressed molecules including CD73 and CD39 catalyze the generation of adenosine from adenosine triphosphate (ATP). The generation of adenosine can be enhanced under metabolic stress like tumor hypoxic conditions. Adenosine exerts its immune regulatory functions through four different adenosine receptors (ARs) including A1, A2A, A2B, and A3 which are expressed on various immune cells. Several studies have indicated the overexpression of adenosine generating enzymes and ARs in various cancers which was correlated with tumor progression. Since the signaling of ARs enhances tumor progression, their manipulation can be promising therapeutic approach in cancer therapy. Accordingly, several agonists and antagonists against ARs have been designed for cancer therapy. In this review, we will try to clarify the role of different ARs in the immunopathogenesis, as well as their role in the treatment of cancer.


Subject(s)
Adenosine/biosynthesis , Neoplasms/immunology , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A3/metabolism , Receptors, Adenosine A2/metabolism , Tumor Escape/immunology , 5'-Nucleotidase/metabolism , Adenosine A1 Receptor Agonists/pharmacology , Adenosine A1 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine A3 Receptor Agonists/pharmacology , Adenosine A3 Receptor Antagonists/pharmacology , Adenosine Triphosphate/metabolism , Animals , Antigens, CD/metabolism , Apyrase/metabolism , GPI-Linked Proteins/metabolism , Humans , Mice , Signal Transduction/immunology
15.
J Neurosci Res ; 96(6): 1066-1079, 2018 06.
Article in English | MEDLINE | ID: mdl-29436006

ABSTRACT

At the mouse neuromuscular junction, adenosine triphosphate (ATP) is co-released with the neurotransmitter acetylcholine (ACh), and once in the synaptic cleft, it is hydrolyzed to adenosine. Both ATP/adenosine diphosphate (ADP) and adenosine modulate ACh secretion by activating presynaptic P2Y13 and A1 , A2A , and A3 receptors, respectively. To elucidate the action of endogenous purines on K+ -dependent ACh release, we studied the effect of purinergic receptor antagonists on miniature end-plate potential (MEPP) frequency in phrenic diaphragm preparations. At 10 mM K+ , the P2Y13 antagonist N-[2-(methylthio)ethyl]-2-[3,3,3-trifluoropropyl]thio-5'-adenylic acid, monoanhydride with (dichloromethylene)bis[phosphonic acid], tetrasodium salt (AR-C69931MX) increased asynchronous ACh secretion while the A1 , A3 , and A2A antagonists 8-cyclopentyl-1,3-dipropylxanthine (DPCPX), (3-Ethyl-5-benzyl-2-methyl-4-phenylethynyl-6-phenyl-1, 4-(±)-dihydropyridine-3,5-, dicarboxylate (MRS-1191), and 2-(2-Furanyl)-7-(2-phenylethyl)-7H-pyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidin-5-amine (SCH-58261) did not modify neurosecretion. The inhibition of equilibrative adenosine transporters by S-(p-nitrobenzyl)-6-thioinosine provoked a reduction of 10 mM K+ -evoked ACh release, suggesting that the adenosine generated from ATP is being removed from the synaptic space by the transporters. At 15 and 20 mM K+ , endogenous ATP/ADP and adenosine bind to inhibitory P2Y13 and A1 and A3 receptors since AR-C69931MX, DPCPX, and MRS-1191 increased MEPP frequency. Similar results were obtained when the generation of adenosine was prevented by using the ecto-5'-nucleotidase inhibitor α,ß-methyleneadenosine 5'-diphosphate sodium salt. SCH-58261 only reduced neurosecretion at 20 mM K+ , suggesting that more adenosine is needed to activate excitatory A2A receptors. At high K+ concentration, the equilibrative transporters appear to be saturated allowing the accumulation of adenosine in the synaptic cleft. In conclusion, when motor nerve terminals are depolarized by increasing K+ concentrations, the ATP/ADP and adenosine endogenously generated are able to modulate ACh secretion by sequential activation of different purinergic receptors.


Subject(s)
Acetylcholine/metabolism , Miniature Postsynaptic Potentials/drug effects , Neuromuscular Junction/drug effects , Neuromuscular Junction/metabolism , Potassium/pharmacology , Purinergic P1 Receptor Antagonists/pharmacology , Purines/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A3 Receptor Agonists/pharmacology , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Animals , Female , Male , Mice , Phenethylamines/pharmacology , Pyrimidines/pharmacology , Receptors, Purinergic P1/metabolism , Thionucleotides/pharmacology , Triazoles/pharmacology
16.
Exp Eye Res ; 170: 160-168, 2018 05.
Article in English | MEDLINE | ID: mdl-29486164

ABSTRACT

Among candidate neuroprotective agents, adenosine is thought to be a possible treatment for central nervous system disorders. Adenosine elicits biological effects through four G protein-coupled receptors (A1, A2A, A2B, and A3). The A2A and A2B receptors stimulate adenylyl cyclase (AC) and increase cyclic adenosine monophosphate (cAMP) levels, whereas A1 and A3 receptors inhibit AC and decrease cAMP levels. Several studies have investigated the effects of adenosine receptors (AdoRs) in glaucoma, because modulation of A1, A2A, or A3 receptor regulates intraocular pressure. In addition, AdoR-related phenomena may induce neuroprotective effects in retinal neurons. Notably, A1, A2A, and A3 receptor agonists reportedly inhibit retinal ganglion cell (RGC) death in in vitro and in vivo glaucoma models. However, there is limited knowledge of the effects of AdoR activation on neurite outgrowth or the regeneration of RGCs. In this report, we described the role of an AdoR subtype in neurite outgrowth and RGC axonal regeneration. The distribution of AdoRs in the retina was evaluated by immunohistochemical analysis. Using primary cultured rat RGCs in vitro and an optic nerve crush model in vivo, neurite elongation was evaluated after stimulation by the following AdoR agonists: CHA, an A1 receptor agonist; CGS21680, an A2A receptor agonist; BAY60-6583, an A2B receptor agonist; and 2-Cl-IB-MECA, an A3 receptor agonist. To determine the mechanism of neurite promotion, the candidate molecules of signal transduction associated with the neurite elongation of AdoRs were evaluated by enzyme-linked immunosorbent assay (ELISA) and Western blot analysis, respectively. All four AdoRs (A1, A2A, A2B, and A3) were present in the inner retinal layers. Among the agonists for AdoR, only 2-Cl-IB-MECA significantly promoted neurite outgrowth in primary cultured RGCs. Signaling pathway analyses showed that 2-Cl-IB-MECA caused upregulated phosphorylation of Akt in cultured RGCs. Additionally, LY294002, an inhibitor of Akt, suppressed the neurite-promoting effects of the A3 receptor agonist in RGCs. Moreover, 2-Cl-IB-MECA increased the number of regenerating axons in the optic nerve crush model. Taken together, these data indicate that activation of the A3 receptor, not the A1 or A2 receptors, promotes in vitro and in vivo neurite outgrowth during the regeneration of rat RGCs, which is caused by the activation of an Akt-dependent signaling pathway. Therefore, AdoR activation may be a promising candidate for the development of novel regenerative modalities for glaucoma and other optic neuropathies.


Subject(s)
Nerve Regeneration/physiology , Neuronal Outgrowth/physiology , Receptor, Adenosine A3/metabolism , Retinal Ganglion Cells/metabolism , Adenosine A1 Receptor Agonists/pharmacology , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A3 Receptor Agonists/pharmacology , Animals , Axons/physiology , Blotting, Western , Cells, Cultured , Cyclic AMP/metabolism , Enzyme-Linked Immunosorbent Assay , Phosphorylation , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A1/metabolism , Receptors, Adenosine A2/metabolism , Retina/metabolism , Retinal Ganglion Cells/drug effects , Signal Transduction
17.
Purinergic Signal ; 14(1): 59-71, 2018 03.
Article in English | MEDLINE | ID: mdl-29170977

ABSTRACT

Activity of the A3 adenosine receptor (AR) allosteric modulators LUF6000 (2-cyclohexyl-N-(3,4-dichlorophenyl)-1H-imidazo [4,5-c]quinolin-4-amine) and LUF6096 (N-{2-[(3,4-dichlorophenyl)amino]quinolin-4-yl}cyclohexanecarbox-amide) was compared at four A3AR species homologs used in preclinical drug development. In guanosine 5'-[γ-[35S]thio]triphosphate ([35S]GTPγS) binding assays with cell membranes isolated from human embryonic kidney cells stably expressing recombinant A3ARs, both modulators substantially enhanced agonist efficacy at human, dog, and rabbit A3ARs but provided only weak activity at mouse A3ARs. For human, dog, and rabbit, both modulators increased the maximal efficacy of the A3AR agonist 2-chloro-N 6-(3-iodobenzyl)adenosine-5'-N-methylcarboxamide as well as adenosine > 2-fold, while slightly reducing potency in human and dog. Based on results from N 6-(4-amino-3-[125I]iodobenzyl)adenosine-5'-N-methylcarboxamide ([125I]I-AB-MECA) binding assays, we hypothesize that potency reduction is explained by an allosterically induced slowing in orthosteric ligand binding kinetics that reduces the rate of formation of ligand-receptor complexes. Mutation of four amino acid residues of the human A3AR to the murine sequence identified the extracellular loop 1 (EL1) region as being important in selectively controlling the allosteric actions of LUF6096 on [125I]I-AB-MECA binding kinetics. Homology modeling suggested interaction between species-variable EL1 and agonist-contacting EL2. These results indicate that A3AR allostery is species-dependent and provide mechanistic insights into this therapeutically promising class of agents.


Subject(s)
Adenosine A3 Receptor Agonists/chemistry , Adenosine A3 Receptor Agonists/pharmacology , Receptor, Adenosine A3/drug effects , Receptor, Adenosine A3/metabolism , Aminoquinolines/chemistry , Aminoquinolines/pharmacology , Animals , Dogs , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Mice , Molecular Docking Simulation , Rabbits , Species Specificity
18.
J Cell Biochem ; 118(9): 2909-2920, 2017 09.
Article in English | MEDLINE | ID: mdl-28230290

ABSTRACT

Numerous studies have demonstrated the role of A3 adenosine receptor (A3AR) and signaling pathways in the multiple aspects of the tumor. However, there is a little study about the function of A3AR in the biological processes of cancer stem cells (CSCs). CSCs have a critical role in the maintenance and survival of breast cancer. The aim of current study was to investigate the effect of A3AR agonist on breast cancer stem cells (BCSCs). XTT assay showed antiproliferative effect of A3AR agonist (Cl-IB-MECA) on BCSCs. Our results also demonstrated that A3AR agonist reduces mammosphere formation in a dose-dependent manner. Flow cytometry analysis showed that A3AR agonist induces G1 cell cycle arrest and apoptosis in BCSCs. Western blot assay showed that A3AR agonist inhibits the expression of cell cycle and apoptotic regulatory proteins as well as the expression of ERK1/2 and GLI-1 proteins. Finally, these findings propose that A3AR agonist induces cell cycle arrest and apoptosis in BCSCs by inhibition of ERK1/2 and GLI-1 cascade. J. Cell. Biochem. 118: 2909-2920, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Adenosine A3 Receptor Agonists/pharmacology , Breast Neoplasms/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Proteins/metabolism , Receptor, Adenosine A3/metabolism , Zinc Finger Protein GLI1/metabolism , Breast Neoplasms/pathology , Cell Survival/drug effects , Female , Humans , MCF-7 Cells , Neoplasm Proteins/agonists , Neoplasm Proteins/genetics , Receptor, Adenosine A3/genetics
19.
Vis Neurosci ; 34: E001, 2017 01.
Article in English | MEDLINE | ID: mdl-28304243

ABSTRACT

Adenosine is a neuromodulator present in various areas of the central nervous system, including the retina. Adenosine may serve a neuroprotective role in the retina, based on electroretinogram (ERG) recordings from the rat retina. Our purpose was to assess the role of A2A and A3 adenosine receptors in the generation and modulation of the rat ERG. The flash ERG was recorded with corneal electrodes from Sprague Dawley rats. Agonists and antagonists for A2A and A3 receptors, and adenosine were injected (5 µl) into the vitreous. The effects on the components of the single flash scotopic and photopic ERGs were examined, and ERG flicker. Adenosine (0.5 mM) increased the mean amplitudes of the scotopic ERG a-waves (68 ± 8 to 97 ± 14 µV, P = 0.042), and b-waves (236 ± 38 µV to 305 ± 42 µV). A2A agonist CGS21680 (2 mM) reduced the mean amplitude of the ERG b-wave, from 298 ± 21 µV in response to the brightest stimulus to 212 ± 19 µV (P = 0.005), and mean scotopic oscillatory potentials (OPs) from 100 ± 9 µV to 47 ± 11 µV (P = 0.023). ZM241385 [4 mM], an A2A antagonist, decreased the scotopic b-wave of the ERG. A3 agonist 2-CI-IB-MECA (0.5 mM) increased the a-wave, while decreasing the scotopic and photopic ERG b-waves, and the scotopic OPs. A3 antagonist VUF5574 (1 mM) increased the mean amplitude of the scotopic a-wave (66 ± 8 to 140 ± 29 µV, P = 0.046) and b-wave (224 ± 20 to 312 ± 39 µV, P = 0.0037). No significant effects on ERG flicker were found. We conclude that retinal neurons containing A2A and/or A3 adenosine receptors contribute to the generation of the ERG a- and b-waves and OPs.


Subject(s)
Receptor, Adenosine A2A/physiology , Receptor, Adenosine A3/physiology , Retina/physiology , Adenosine/pharmacology , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine A3 Receptor Agonists/pharmacology , Adenosine A3 Receptor Antagonists/pharmacology , Animals , Dark Adaptation , Electroretinography/drug effects , Female , Intravitreal Injections , Photic Stimulation , Rats , Rats, Sprague-Dawley
20.
Clin Exp Nephrol ; 21(2): 203-211, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27278932

ABSTRACT

BACKGROUND: ADPKD is a renal pathology caused by mutations of PKD1 and PKD2 genes, which encode for polycystin-1 (PC1) and polycystin-2 (PC2), respectively. PC1 plays an important role regulating several signal transducers, including cAMP and mTOR, which are involved in abnormal cell proliferation of ADPKD cells leading to the development and expansion of kidney cysts that are a typical hallmark of this disease. Therefore, the inhibition of both pathways could potentiate the reduction of cell proliferation enhancing benefits for ADPKD patients. METHODS: The inhibition of cAMP- and mTOR-related signalling was performed by Cl-IB-MECA, an agonist of A3 receptors, and rapamycin, respectively. Protein kinase activity was evaluated by immunoblot and cell growth was analyzed by direct cell counting. RESULTS: The activation of A3AR by the specific agonist Cl-IB-MECA causes a marked reduction of CREB, mTOR, and ERK phosphorylation in kidney tissues of Pkd1 flox/-: Ksp-Cre polycystic mice and reduces cell growth in ADPKD cell lines, but not affects the kidney weight. The combined sequential treatment with rapamycin and Cl-IB-MECA in ADPKD cells potentiates the reduction of cell proliferation compared with the individual compound by the inhibition of CREB, mTOR, and ERK kinase activity. Conversely, the simultaneous application of these drugs counteracts their effect on cell growth, because the inhibition of ERK kinase activity is lost. CONCLUSION: The double treatment with rapamycin and Cl-IB-MECA may have synergistic effects on the inhibition of cell proliferation in ADPKD cells suggesting that combined therapies could improve renal function in ADPKD patients.


Subject(s)
Adenosine A3 Receptor Agonists/pharmacology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Cell Proliferation/drug effects , Cyclic AMP/antagonists & inhibitors , Kidney/drug effects , Polycystic Kidney, Autosomal Dominant/drug therapy , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , CREB-Binding Protein/metabolism , Cell Line , Cyclic AMP/metabolism , Disease Models, Animal , Drug Synergism , Drug Therapy, Combination , Extracellular Signal-Regulated MAP Kinases/metabolism , Genetic Predisposition to Disease , Humans , Kidney/metabolism , Kidney/pathology , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Phosphorylation , Polycystic Kidney, Autosomal Dominant/enzymology , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/pathology , TOR Serine-Threonine Kinases/metabolism , TRPP Cation Channels/deficiency , TRPP Cation Channels/genetics , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL