Abstract
Agonists and antagonists of various subtypes of G protein coupled adenosine receptors (ARs), P2Y receptors (P2YRs), and ATP-gated P2X receptor ion channels (P2XRs) are under consideration as agents for the treatment of ocular diseases, including glaucoma and dry eye. Numerous nucleoside and nonnucleoside modulators of the receptors are available as research tools and potential therapeutic molecules. Three of the 4 subtypes of ARs have been exploited with clinical candidate molecules for treatment of the eye: A1, A2A, and A3. An A1AR agonist is in clinical trials for glaucoma, A2AAR reduces neuroinflammation, A3AR protects retinal ganglion cells from apoptosis, and both A3AR agonists and antagonists had been reported to lower intraocular pressure (IOP). Extracellular concentrations of endogenous nucleotides, including dinucleoside polyphosphates, are increased in pathological states, activating P2Y and P2XRs throughout the eye. P2YR agonists, including P2Y2 and P2Y6, lower IOP. Antagonists of the P2X7R prevent the ATP-induced neuronal apoptosis in the retina. Thus, modulators of the purinome in the eye might be a source of new therapies for ocular diseases.
Keywords: : adenosine receptor, P2Y receptor, P2X receptor, ATP
Introduction
Adenosine and various adenine and uracil nucleotides act as extracellular autocrine or paracrine signals by activating cell surface receptors.1,2 The adenosine receptors (ARs) represent 4 subtypes of G protein-coupled receptors (GPCRs), and nucleotides also activate distinct GPCRs (8 subtypes of P2Y receptors, P2YRs) and/or ligand-gated ion channels (7 subunits forming trimeric P2X receptors, P2XRs).
Numerous synthetic agonists and antagonists for exogenous application are available as tools to study these receptors (Figs. 1–5), and some of these molecules have progressed into clinical trials.3–5 These ligands are largely orthosteric, that is, they bind to the same site on the receptor as the native agonist, but some allosteric modulators of the ARs, P2YRs, and P2XRs have been reported.6 The allosteric modulators can either enhance the action of the native agonist, that is, act as a positive allosteric modulator, or antagonize it as a negative allosteric modulator. Recently, monoclonal antibodies were also reported to modulate P2XR activity.7
FIG. 1.
Structures of AR agonist ligands. AR, adenosine receptor.
FIG. 2.
Structures of AR antagonist ligands.
FIG. 3.
Structures of P2YR and P2XR agonists.
FIG. 4.
Structures of P2YR antagonists.
FIG. 5.
Structures of P2XR antagonists.
Table 1 lists receptors associated with the biological actions of extracellular purines and pyrimidines, which collectively could be considered a purinome and representative ligands. All ARs, P2XRs, and P2YRs are well expressed in all ocular tissues (Fig. 6), as indicated by the PLIER values generated by the Affymetrix algorithm.8 There is a generally higher expression level for A2B, P2Y14, and P2X2Rs in various ocular tissues. However, the level of expression of these receptors can vary greatly in pathological states; thus, use of purine receptor agonists and antagonists in disease models must take receptor regulation into account.4
Table 1.
Receptor Subtypes Involved in the Action of Extracellular Purines and Pyrimidines (pKa Values at Human Receptors for Representative Agonist and Antagonist Ligands Are Given in Parentheses).1,3,5,6,106,121
| Subtype | Gene name | Chromosome | Native and synthetic agonist(s) | Representative antagonist(s) |
|---|---|---|---|---|
| Adenosine receptors (GPCRs) | ||||
| A1 | ADORA1 | 1q32.1 | Adenosine1 | PSB36 28, SLV320 29 |
| A2A | ADORA2A | 22q11.23 | Adenosine1 | SCH442416 34 |
| A2B | ADORA2B | 17p12 | Adenosine1 | MRS1754 36 |
| A3 | ADORA3 | 1p13.2 | Adenosine1, inosine2 | MRS1191 39, MRS1523 42 |
| P2Y receptors for nucleotides (GPCRs) | ||||
| P2Y1 | P2RY1 | 3q25.2 | ADP47 (5.1), 2-MeSADP 48 (8.2), MRS2365 51 (9.4) | MRS2500 73 (9.0) |
| P2Y2 | P2RY2 | 11q13.4 | UTP53 (8.1), ATP49 (7.1), PSB1114 61 (6.9) | AR-C1189251XX 74 (7.4) |
| P2Y4 | P2RY4 | Xq13 | UTP53 (6.3) | - |
| P2Y6 | P2RY6 | 11q13.4 | UDP64 (7.9), MR2693 65 (7.8) | MRS2578 75 (7.4) |
| P2Y11 | P2RY11 | 19p13.2 | ATP49 (4.8), NF546 69 (6.3) | NF340 76 (7.1) |
| P2Y12 | P2RY12 | 3q25.1 | ADP47 (7.2), 2-MeSADP 48 (8.3) | Cangrelor 77 (9.4), Ticagrelor 78 (7.9), AZD1283 79 (8.0) |
| P2Y13 | P2RY13 | 3q24 | ADP47 (7.9) | MRS2211 82 (6.0), |
| P2Y14 | P2RY14 | 3q21-q25 | UDP64 (6.8), UDP-glucose70 (6.5) | PPTN 83 (8.7) |
| P2X receptors for ATP (in channels) | ||||
| P2X1 | P2RX1 | 17p13.3 | ATP49 (7.3) | NF449 85 (10.7), MRS2159 86 (8.0) |
| P2X2 | P2RX2 | 12q24.33 | ATP49 (6.0), Bz-ATP 52 (6.4) | PSB-1011 87 (7.1) |
| P2X3 | P2RX3 | 11q12 | ATP49 (6.5) | RO-3 88 (7.0) |
| P2X4 | P2RX4 | 12q24.32 | ATP49 (6.3) | 5-BDBD 89 (6.3), PSB-12054 90 (6.7) |
| P2X5 | P2RX5 | 17p13.3 | ATP49 (5.4) | BBG (6.3) |
| P2X6 | P2RX6 | 22q11.21 | ATP49 (6.3) | TNP-ATP (6.1) |
| P2X7 | P2RX7 | 12q24 | ATP49 (2.2), Bz-ATP 52 (4.3) | A438079 92 (6.9), JNJ 47965567 96 (8.3) |
ARs, adenosine receptors; BBG, Brilliant Blue G; GPCR, G protein-coupled receptor.
FIG. 6.
Occurrence of purinergic receptors (A, ARs; B, P2YRs; C, P2XRs;) determined in exon-level expression profiling of ocular tissues.8
Furthermore, a set of enzymes and transporters control the levels of extracellular purines and pyrimidines and, therefore, indirectly modulate the degree of activation of the various receptors.9 The numerous members of the ectonucleotidase families (such as CD39 and CD73) catalyze the hydrolysis of phosphate groups of the nucleotide ligands of P2Y and P2XRs. Thus, inhibition of these enzymes would reduce the amount of adenosine 1 available to activate ARs. There are both equilibrative (ENTs) and concentrative (CNTs) transporters for adenosine. For example, the approved vasodilator dipyridamole (Persantine, structure not shown) inhibits the equilibrative transporter ENT1 and other ENTs to increase the amount of extracellular adenosine.10 The therapeutic modulation of enzymes and transporters associated with the extracellular concentrations of purines and pyrimidines is also being considered for clinical development.11
Adenosine receptor modulators as drug targets in the eye
The ARs are coupled to G proteins that cause either increases (A2A and A2B) or decreases (A1 and A3) of intracellular cyclic AMP. However, a range of other G protein-dependent and independent signaling mechanisms are associated with activation of ARs.1 Nucleoside derivatives that activate ARs (1–20) are shown in Fig. 1, and AR antagonists (both nucleosides and nonnucleosides, 25–49) are shown in Fig. 2. Compounds 21–24 indirectly modulate the activity at ARs as inhibitors of the removal of adenosine by phosphorylation (21 and 22, by adenosine kinase) or through allosteric enhancement of the A1 23 or A3AR 24. There are potent agonists and antagonists associated with each of the 4 AR subtypes, but caution must be used when applying them to pharmacological experiments due to selectivities that are not completely definitive for the AR subtypes. For example, the “A1AR agonist” R-PIA 3 causes reductions in heart rate, activity, and body temperature in mice that are dependent on the presence of the A3AR; genetic knockout of the A3AR prevents some of these effects.12 Various agonists were defined as A1AR selective before the discovery of the A3AR. For example, CPA 4 and CHA 8 are more potent at the A3AR than at the A2AAR.3 The A1AR agonist CCPA 5 is more selective than CPA, but it has lower than full efficacy at the A3AR, that is, it can act as a partial agonist or an antagonist.13 However, among the more selective agonists are: 5′-Cl-5′-d-ENBA (Cl-ENBA) 10 at the A1AR and MRS5698 18 at the A3AR,14,15 which have been shown to be selective in vivo using A1AR and A3AR knockout mice.16 Agonists of the A1AR 9 (moderately selective) and the A3AR 19 (highly selective) do not cross biological membranes because of a charged sulfonate group that has been introduced.
The generally suppressant role of the A2AAR and A2BAR in the immune system has been explored.17 Potent A2AAR agonist UK432,097 12 failed clinical trials for COPD, but displays extended lung retention.18 Lexiscan 13a is a short acting A2AAR agonist that is approved for dilating coronary vessels during stress testing and was recently demonstrated to open the blood–brain barrier transiently, with a decreased P-glycoprotein expression, and to ameliorate sickle cell disease.19–21 Its A2AAR affinity and selectivity are only moderate (Ki ∼1.3 μM, >10-fold selective versus A1AR). More potent and selective A2AAR agonist ATL-313 13b and other A2AAR agonists are being considered for treatment of inflammatory pain and sepsis.3,22,23 The potential of using A2AAR antagonists, for example, 30–35, to boost the immunotherapy of cancer is based on blocking the effects of elevated adenosine levels in the tumor microenvironment that suppress the immune response, and many pharma companies are actively pursuing this concept.24 Nonnucleoside A2BAR agonist BAY 60-6583 14 has been noted to act as partial agonist or even antagonist at that receptor in different models.25
Suggested antagonists for pharmacological experiments are provided in Table 1. A relatively nonselective but potent pan-AR antagonist is xanthine amine congener (XAC, structure not shown). One should also be cognizant of species differences in the affinity of a given ligand.26 For example, the A3AR antagonist MRS1191 39 has a Ki value of 31 nM at the human A3AR, 1.4 μM at the rat A3AR, and >10 μM at the mouse A3AR.27,28 A3AR antagonist OT-7999 46 has a Ki value of 0.61 nM at the human A3AR and >10 μM at the rat A3AR.27 For that reason, its preclinical validation for glaucoma had to be performed in primate species.29 In some cases, such as human A3AR antagonist MRS1220 41, the selectivity can be inverted, depending on species. The A3AR affinities of nucleoside-derived antagonists, such as MRS1292 43, LJ1251 44, and LJ979 45 tend to be more constant across species than the nonnucleotide antagonists. Thus, the ligand tools for the ARs must be used with all appropriate controls and blocking with antagonists or, preferably, genetic knockout. There are also species differences in the biology of a given purine receptor subtype, for example, the A3AR in mast cells.15
A rise in the concentrations of adenosine 1 and inosine 2 in aqueous humor accompanies intraocular hypertension and correlates with the magnitude of the pressure.30 Inosine is formed from adenosine by adenosine deaminase, a ubiquitous enzyme, and at high concentrations can activate various ARs.31 The increase in adenosine is thought to arise from release of ATP from both the nonpigmented ciliary epithelial cells responsible for aqueous humor inflow and from trabecular meshwork cells of the outflow tract. The ATP is subsequently converted to adenosine by ectoenzymes of those cells.32–36 It might be expected that solvent drag would preclude a contribution of adenosine by the trabecular meshwork cells in a direction against the flow of aqueous humor from the anterior chamber. However, even proteins, having a higher frictional interaction with water can proceed against net flow. As an example, net diffusion of proteins from posterior to anterior chambers occurs across the iris root in rabbits,37 monkeys,38 and human.39 This net diffusion proceeds against the net flow.40,41 Adenosine concentration in the rabbit eye can be increased by application of dipyridamole.42 Modulation of ARs has been explored for several decades for the treatment of glaucoma. Three of the 4 subtypes of ARs have been exploited with clinical candidate molecules for treatment of the eye: A1, A2A, and A3.43–45 Clinical trials for glaucoma have ensued.46
The effects of AR modulation on specific tissues in the eye and on intraocular pressure (IOP) have been extensively probed by multiple techniques. For example, activation of A1AR or A2AAR in juxtacanalicular tissue cells increased the concentrations of Cl− and K+ and the cell volume measured by electron probe X-ray microanalysis.47 The intracellular ionic composition of cells in the inner wall of Schlemm's canal was similarly affected by activating the A2AAR, but not the A1AR. The differential effects of AR agonists to alter the resistance to outflow of aqueous humor have also been studied. Topically applied A1AR agonists lower resistance, thereby enhancing aqueous humor outflow and, consequently, reducing IOP.48,49 The reduction in resistance appears mediated by release of matrix metalloproteinases (MMP-2 and MMP-9) from trabecular meshwork cells of the aqueous outflow tract.32,48 However, the time course of the effects of MMPs in the perfused bovine anterior segments48 was much faster than the effects of MMPs on human eyes in organ culture.50 Whether the different time courses reflected species or experimental differences remains unknown. Topically applied ATP also stimulated release of MMPs, but this release is dependent on conversion of the nucleotide to adenosine.45 The finding that A1AR agonists can lower IOP has led to clinical trials of INO-8875 (Trabodenoson) 6 for glaucoma.49 In the anesthetized rat, INO-8875 displayed a longer duration of action in lowering atrioventricular (AV)-nodal conduction, reflective of activation of the A1AR compared to another A1AR agonist CVT-510 (Tecadenoson) 7.51 A single topical dose of INO-8875 6 reduced IOP in glaucoma patients in a Phase1/2 clinical trial.52 A Phase 3 study of topically applied 6 in adults with ocular hypertension or primary open-angle glaucoma (MATrX-1) is underway (NCT02565173).46
A2AAR agonists and antagonists have also been undergoing study to treat glaucoma.53 The A2AAR agonist, OPA-6566 (structure not disclosed), underwent Phase1/2 clinical trials (NCT01410188) by Acucela, Inc. and Otsuka Pharmaceutical Co., Ltd. for the management of open-angle glaucoma or ocular hypertension.46 The trials have been completed, but no study results have yet been posted. Santen Pharmaceuticals has also been exploring the potential use of A2AAR agonist ATL-313 13b for lowering IOP.54 The promise of A2AAR agonists is uncertain since they have exerted both unfavorable and favorable effects in preclinical studies. Activation of the A2AAR was found to be neuroprotective against traumatic optic neuropathy by attenuating the inflammatory response to optic nerve trauma.55 Low levels of traumatic optic neuropathy that would otherwise induce only a minimal neuroinflammatory response in wild-type mice induced severe inflammation in A2AAR−/− mice. In addition, A2AAR had an anti-inflammatory effect on retinal ganglion cells subjected to elevated pressure.56 Given that inflammation plays a role in the pathogenesis of glaucoma,57–59 A2AAR agonists might be helpful. However, A2AR agonists also act to reduce vascular resistance and increase blood flow to the retina and optic nerve head.43 Antagonists, but not agonists, of the A2AAR can improve recovery of retinal function after ischemia reperfusion.43 Furthermore, a selective A2AAR antagonist (SCH58261, 33) prevented ischemia-reperfusion death of retinal ganglion cells arising from transient elevation of IOP.60 The neuroprotection was thought to have been mediated by attenuating the microglial-mediated neuroinflammatory response.55,60 Another caveat is that the effect of A2AAR agonists on IOP is expected to be unfavorable since the A2AAR antagonist ZM241385 31 lowered pressure in the mouse,33 while activation of A2AARs transiently increased pressure in rabbit and cynomolgus monkey eyes.61 In short, A2AAR agonists and antagonists have multiple effects, and current published evidence that A2AAR agonists would be helpful in glaucoma is not yet compelling.
As an alternative to the use of side effect-prone AR agonists, adenosine kinase inhibitor ABT-702 21 was administered to mice to increase the level of endogenous adenosine and, thereby, protect against traumatic optic neuropathy-induced retinal injury and reduce pro-inflammatory cytokines.62 Nevertheless, adenosine kinase inhibitors have their own side effects, led to the discontinuation of past clinical trials of centrally-acting inhibitors.63
At the A3AR, there is evidence that topically applied selective antagonists reduce IOP, and topically applied selective agonists increase IOP.33,64–66 The mechanistic basis for this action is that the A3AR is coupled to a chloride channel in the nonpigmented ciliary epithelial layer, such that its activation causes inflow leading to a rise in IOP.67 This concept was supported by studies of A3AR knockout mice, in which the absence of the A3AR lowered IOP.68 In addition, the A3AR is more highly expressed in the nonpigmented ciliary epithelium in glaucoma associated with pseudoexfoliation syndrome.69 The finding of A3AR as a regulator of chloride transport in the nonpigmented ciliary epithelium led to the development of selective A3AR antagonists for the treatment of glaucoma. A nucleoside-based antagonist of the A3AR, LJ1251 44, was found to lower IOP.66 Acorn Biomedical has proposed using ACN-1052 (structure not disclosed) for glaucoma treatment.70 In addition, a “first-in-human” Phase I trial (NCT02639975) of Palobiofarma's orally administered A3AR antagonist PBF-677 (structure not disclosed) intended for treatment of glaucoma is expected to begin enrolling healthy volunteers to test safety and tolerability, as it has received the approval of the Spanish Regulatory Agency.46,71
Interestingly, agonists of the A3AR improved aqueous humor outflow in an animal model and in preliminary human clinical data.72,73 OphthaliX is a daughter company of Can-Fite Biopharma, which is already sponsoring clinical trials of A3AR agonists, originally reported by the NIDDK laboratory,74,75 for autoimmune inflammatory diseases (rheumatoid arthritis and psoriasis) and hepatocellular carcinoma. During a previous unsuccessful trial of the A3AR agonist CF101 (IB-MECA, Piclodenoson 15) for dry eye disease, a modest reduction of IOP was noted in patients receiving the drug.72 The reported reduction of IOP was 0.92 mm Hg at a dose of 1 mg CF101 twice daily. This prompted a separate clinical path sponsored by OphthaliX for the same A3AR agonist in glaucoma treatment. However, in a phase 2 trial of systemic 15 (1 or 2 mg oral dose, twice daily) for 16 weeks in patients with elevated IOP, no statistically significant differences were found between the drug-treated group and placebo group (NCT01033422).45,76
The apparent contradiction that both agonists and antagonists of the A3AR might be useful in treating the same conditions, other than glaucoma, still lacks a mechanistic explanation. The agonist-stimulated downregulation of the A3AR in cell systems has been reported to occur in ∼20 min,77,78 but in animals a sustained action of A3AR activation in chronic pain has been noted.79 The limitation of using adenosine agonists in glaucoma due to the desensitization of ARs has been noted.80 Nucleoside prodrugs of A3AR agonists and antagonists have been explored for their action on IOP.65,66,81,82
Elevated IOP causes degeneration and eventually apoptotic death of retinal ganglion cells (RGCs). ARs are known to be present in the retina8,83 and their activation provides protection against this apoptosis. Specifically, A3AR agonists such as Cl-IB-MECA 16 and MRS3558 17 protect RGCs from apoptosis induced by ATP and other nucleotides that activate the P2X7R, such as Bz-ATP 52. A mixed A1/A3 agonist MRS3630 20 was also effective. A3AR agonist IB-MECA 15 also induced an anti-inflammatory effect in experimental autoimmune uveitis induced by retinal antigen interphotoreceptor retinoid-binding protein.84
The pathophysiology of retinal ARs and dysregulated levels of adenosine have been explored with the goal of correcting the excitotoxicity and inflammation associated with diabetic neuropathy.85 Exposure of rat retinal cells to sustained (12 week) high glucose upregulated the A1AR, and the retinal A2AAR is upregulated in diabetic animal and cellular models, while the A3AR displayed only a transient increase. However, conflicting results do not provide a clear path forward to clinical approaches using AR ligands.
P2YR modulators as drug targets in the eye
Nucleotide derivatives that activate P2YRs and P2XRs (47–71) are shown in Fig. 3 and P2YR antagonists (both nucleotides and nonnucleotides, 72–84) are shown in Fig. 4. ATP is the common agonist used for all of the P2XRs, but several subtypes (e.g., P2X1R and P2X7R) are activated more potently by Bz-ATP 52. We still lack potent agonists and antagonists for various subtypes of P2YRs and P2XRs.3 Nevertheless, potent (∼nM) antagonists for the P2Y1R, P2Y12R, and P2Y14R and for the P2X1R, P2X3R, and P2X7R are available (Fig. 5). P2Y2R ligand AR-C1189251XX 74 has been used effectively as a selective antagonist for this subtype.86 Four antagonists of the P2Y12R receptor are approved as antithrombotic agents. Two of them (thienopyridines Clopidogrel 80 and Prasugrel 81) are prodrugs that must be first activated enzymatically in vivo; the other 2 are nucleotide Cangrelor 77 and nucleoside Ticagrelor 78, both of which are competitive at the receptor.87 Applications of P2YR agonists in the eye and elsewhere are envisioned,3 and consequently, selective agonists for P2Y1R, P2Y2R, P2Y4R, P2Y6R, P2Y11R, and P2Y14R have been reported.
P2YRs are important in many physiological and pathophysiological ocular functions and have been immunolocalized in the eye.45,88,89 The P2Y1R is localized in the cornea, ciliary processes, and trabecular meshwork. Activation of the P2Y1R has contradictory effects in the retina. It affects volume control and worsens reactive gliosis in Müller cells (astrocytes) in the retina, but it reduces ischemia-induced apoptosis of cells in all retinal layers.65,90 The P2Y2R, P2Y4R, and P2Y6R are expressed in the cornea and ciliary processes, while the retinal pigmented epithelium expresses P2Y2R. The P2Y4R and P2Y6R are also found in the photoreceptors, outer plexiform layer, and ganglion cell layer. The retinal pigmented epithelium also expressed the P2Y11R. P2Y2R, P2Y4R, P2Y11R, and P2Y13R are present on lachrymal glands, where UTP causes myoepithelial cell contraction.91
P2YRs have multiple actions in the eye: they control tear production, corneal wound healing, aqueous humor dynamics, and retinal physiology. Nucleotides can affect both the volume and composition of tears, while regulating processes necessary for corneal wound healing. In the lens, osmotic stress activates TRPV4 channels, which leads to ATP release through connexin and pannexin hemichannels.45 The released ATP activates a P2YR to help maintain lens transparency by activating a Na/K ATPase pump on the epithelium. Both UTP 53 and Ap4A 58 applied topically to the rabbit cornea stimulate a MAPK-dependent increase in epithelial cell migration to accelerate wound healing.92 Where P2Y2R activation accelerates, P2Y6R activation impedes the migration rate of corneal epithelial cells in rabbit primary cell cultures.93 Nucleotides cause an increase in the content of secreted mucins, lysozyme, and other tear proteins. The complicated distribution of P2Y2R, P2Y4R, and P2Y6R suggested some redundancy in the role of receptors for uracil nucleotides in the eye, for example, in tear production and IOP regulation. However, the use of P2YR knockout mice to clarify their role in the eye has been limited.90 siRNA knockdown was used to show that the P2Y2R promotes cell migration in epithelial scratch wound repair.40 P2YR agonists, including those of P2Y2R and P2Y6R, lower IOP. The potential role of P2Y14R is particularly puzzling. Exon-level profiling has indicated that P2Y14R is more highly expressed than any other purine receptor8 (Fig. 6). Furthermore, the gene is expressed far higher in the trabecular meshwork than in any of the 9 other ocular tissues studied (Fig. 6B). One might speculate that the P2Y14R protein product could play a role in the regulation of outflow resistance, which is regulated by the trabecular meshwork. Indeed, Podos presented results in abstract form, suggesting that topical application of UDP-glucose to activate P2Y14R did lower IOP in glaucomatous monkey eyes.94 However, no peer-reviewed publication has since verified those early promising observations.
Naturally occurring diadenosine polyphosphates, such as Ap4A 58, activate P2YRs to regulate tear secretion and other functions.45,95–97 These dinucleotides are typically found in higher concentrations in eye diseases, such as Sjögren and non-Sjögren dry eye disease and glaucoma. It was suggested that lowering IOP by Ap4A is mediated by the P2X2R, causing acetylcholine release from nerve terminals that regulate ciliary processes.45 In addition to the adenine dinucleotides, the concentration of Ip4I 60 was shown to be elevated in glaucoma. Ip4I 60 reduced IOP by activating P2YRs, although the subtype(s) involved were not clearly delineated.
The role of the P2Y2R in the eye has been reviewed.98 The dinucleotide INS365 59b (Diquafosol, also known as Diquas®) is a mixed agonist of the P2Y2R and P2Y4R that has been shown to improve the corneal barrier function.99 In a rat dry eye model, this agonist increased both tear fluid secretion and corneal epithelial resistance and induced the release of glycoprotein-containing moieties from goblet cells. These successful preclinical results led to the introduction of Diquafosol in Japan and South Korea for the treatment of dry eye syndrome.100 As a topical ophthalmic solution (3%, 6-times daily), it stimulated secretion of tears and mucin in patients with dry eye disease, and its effectiveness was maintained for 12 months.101 In a large randomized, double-blind trial, it improved the fluorescein staining score in dry eye disease comparably to 0.1% sodium hyaluronate and improved the rose bengal subjective symptom scores significantly better than hyaluronate.102 The encouraging clinical results with Diquafosol in dry eye disease were recently reviewed.103 It was shown to be effective in various types of dry eye disease, such as aqueous deficient, short tear film break-up time, and obstructive meibomian glandular. It is also useful in treating dry eye disease resulting from surgery (for in situ keratomileusis and cataracts) and from use of contact lenses and visual display terminals. Although this compound is a GPCR agonist, which is prone to inducing desensitization of the receptor, its effect was maintained over the long term (3% solution, 6-times daily, for 6 months or 52 weeks).103 Eye irritation was reported as an adverse event in only 6.3% of the cases in a trial of Diquafosol and there were no serious adverse effects.104 Diquafosol was applied topically because the P2Y2R occurs on the ocular surface, and systemic absorption was not detectable and without effect. The enzyme mainly responsible for hydrolysis of the drug in airway epithelial cells was found to be alkaline phosphodiesterase 1 (PDE1), which is membrane bound and has a broad substrate specificity. Nucleotides are also rapidly metabolized when in contact with the corneal cells.105
P2Y6R agonists have also been explored for reducing IOP.106–108 Topically applied UDP 64 reduced rabbit IOP by a maximal 17% compared to control, corresponding to an EC50 value of 27 nM. A potent agonist of the P2Y6R, TG46 (67, not to be confused with the JAK2 inhibitor TG-46), was administered topically to reduce IOP in rabbits by 45%. P2Y6R is also important in the retina. Müller cells in the retina release UTP, which is hydrolyzed rapidly to UDP, to act at the P2Y6R in neighboring RGCs.109 P2Y6R activation induced neurite outgrowth, which would be beneficial in glaucoma. In glaucoma, both the expression level of the P2Y6R and its endogenous agonists might be downregulated.109 However, in glaucomatous mice, topical application of Ap4A lowered the expression of mRNA for P2Y2R and P2Y6R that was elevated due to the disease state.110
P2XR modulators as drug targets in the eye
P2XRs also have important roles in the eye and their modulation has been explored for therapeutic purposes. The pharmacology of P2XRs is complicated by the presence of heterotrimers, which have distinct ligand profiles compared to the homotrimeric channels. In theory, there is a therapeutic opportunity due to unique heterotrimer pharmacology, but most of the known combinations of subunits are not yet associated with specific biological effects and do not yet have selective ligands.2,3,5 In addition, splice variants have been shown to display different pharmacological properties.111 Neuronal P2XRs in the inner and outer retina contribute to visual processing, as well as cell death in retinal ganglia. P2X2R is present in the trigeminal ganglion sensory neurons and ciliary body.112 In chronic glaucoma, ATP is released through pannexin hemichannels in astrocytes of the optic nerve and activates P2X7Rs that can increase cell death in the retina. The ATP release from astrocytes can result from chronic mechanical strain (stretch) as it occurs in glaucoma through the upregulation of pannexins.113 The P2X7R in lacrimal glands interacts with M3 muscarinic acetylcholine receptors and α1D-adrenergic receptors to regulate tear secretion.45 P2X7R plays an essential role in corneal epithelial cell migration and stromal organization during healing from abrasion wounds.114 The P2X1R mediates ATP release in trabecular meshwork and ciliary epithelial cells.45,115 P2X2, P2X3, and P2X7Rs colocalize with GABA in amacrine cells, including neurons postsynaptic to cone bipolar cells (all 3 subtypes) and rod bipolar cells (only P2X3 and P2X7Rs).116 Although all P2XRs are present in the rat lacrimal gland, ATP acting at P2X3R and P2X7R increases protein secretion by raising intracellular [Ca2+].117 ATP acts at P2X7R to modulate both M3 muscarinic cholinergic and α1D-adrenergic signaling in the lacrimal gland.45
The retinal P2X7R is overactivated in diabetes, leading to pathological gliosis.118 Furthermore, both degeneration of the retinal pigment epithelium and choroidal neovascularization associated with different types of age-related macular degeneration are dependent on the P2X7R.118 Thus, P2X7R antagonists might prove useful in treating these conditions.
Nonnucleotide P2X3R antagonists are proceeding toward the clinic for treatment of cough,119 and P2X4R antagonist (structure not disclosed) is scheduled to enter clinical trials for chronic neuropathic pain.120 P2X7R antagonists that have been in clinical trials for conditions other than in the eye include AZD9056 93 and CE-224,535 94 (rheumatoid arthritis) and GSK1482160 95 (inflammatory pain).4,121 The trials for rheumatoid arthritis failed to show efficacy, but the trial of GSK1482160 95 showed a reduction of IL1β following exposure to LPS.122 Antagonists of the P2X7R prevent neuronal apoptosis in the retina induced by ATP or by hypoxia.118,123–126,127,128 Such antagonists include Brilliant Blue G (BBG, structure not shown) and MRS 2540 91. JNJ-47965567 96 is a CNS penetrant, selective P2X7R antagonist, while JNJ-54173717 97 was recently reported as a PET ligand for imaging the P2X7R.129
Summary
In summary, the current review has focused broadly upon the gene and protein expression of the 19 purine receptors so far identified in human ocular tissues and their physiological role. The current puzzlements and apparent inconsistencies in the results thus far have also been highlighted, as well as promising leads for future probes. In many cases, there are beneficial effects of both the nucleotides that are released locally to activate P2XRs and P2YRs and the adenosine that is produced enzymatically to activate ARs. The enormous spectrum of functions and the early utilization of probes in clinical studies have been addressed. Synthetic ligands that are selective for various purine receptors have promise for ocular therapeutics.
Acknowledgment
K.A.J. thanks the NIDDK, NIH Intramural Research Program for support.
Author Disclosure Statement
No competing financial interests exist.
References
- 1.Fredholm B.B., IJzerman A.P., Jacobson K.A., Linden J., and Müller C. Nomenclature and classification of adenosine receptors–An update. Pharmacol. Rev. 63:1–34, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Ralevic V., and Burnstock G. Receptors for purines and pyrimidines. Pharmacol. Rev. 50:413–492, 1998 [PubMed] [Google Scholar]
- 3.Jacobson K.A., and Müller C.E. Medicinal chemistry of adenosine, P2Y and P2X receptors. Neuropharmacology. 104:31–49, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Ochoa-Cortes F., Liñán-Rico A., Jacobson K.A., and Christofi F.L. Potential for developing purinergic drugs for gastrointestinal diseases. Inflamm. Bowel Dis. 20:1259–1287, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Coddou C., Yan Z., Obsil T., Huidobro-Toro J.P., and Stojilkovic S.S. Activation and regulation of purinergic P2X receptor channels. Pharmacol. Rev. 63:641–683, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Jacobson K.A., and Gao Z.G. Allosteric modulators of adenosine, P2Y and P2X receptors. Chapter in Allosterism in Drug Discovery (RSC), Doller D., Ed., in press
- 7.Shcherbatko A., Foletti D., Poulsen K., Strop P., Zhu G., Hasa-Moreno A., Melton Witt J., Loo C., Krimm S., Pios A., Yu J., Brown C., Lee J.K., Stroud R., Rajpal A., and Shelton D. Modulation of P2X3 and P2X2/3 receptors by monoclonal antibodies. J. Biol. Chem. 291:12254–12270, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Wagner A.H., Anand V.N., Wang W.H., Chatterton J.E., Sun D., Shepard A.R., Jacobson N., Pang I.H., Deluca A.P., Casavant T.L., Scheetz T.E., Mullins R.F., Braun T.A., and Clark A.F. Exon-level expression profiling of ocular tissues. Exp. Eye Res. 111:105–111, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Deaglio S., and Robson S.C. Ectonucleotidases as regulators of purinergic signaling in thrombosis, inflammation, and immunity. Adv. Pharmacol. 61:301–332, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Wang C., Lin W., Playa H., Sun S., Cameron K., and Buolamwini J. Dipyridamole analogues as pharmacological inhibitors of equilibrative nucleoside transporters. Identification of novel potent and selective inhibitors of the adenosine transporter function of human equilibrative nucleoside transporter 4 (hENT4). Biochem. Pharmacol. 86:1531–1540, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Al-Rashida M., and Iqbal J. Therapeutic potentials of ecto-nucleoside triphosphate diphosphohydrolase, ecto-nucleotide pyrophosphatase/phosphodiesterase, ecto-5′-nucleotidase, and alkaline phosphatase inhibitors. Med. Res. Rev. 34:703–743, 2014 [DOI] [PubMed] [Google Scholar]
- 12.Yang J.N., Wang Y., Garcia-Roves P.M, Björnholm M., and Fredholm B.B. Adenosine A3 receptors regulate heart rate, motor activity and body temperature. Acta Physiol. (Oxf). 199:221–230, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Gao Z.G., and Jacobson K.A. 2-Chloro-N6-cyclopentyladenosine, adenosine A1 receptor agonist, antagonizes the adenosine A3 receptor. Eur. J. Pharmacol. 443:39–42, 2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Franchetti P., Cappellacci L., Vita P., Petrelli R., Lavecchia A., Kachler S., Klotz K.-N., Marabese I., Luongo L., Maione S., and Grifantini M. N6-Cycloalkyl- and N6-bicycloalkyl-C5′(C2′)-modified adenosine derivatives as high-affinity and selective agonists at the human a1 adenosine receptor with antinociceptive effects in mice. J. Med. Chem. 52:2393–2406, 2009 [DOI] [PubMed] [Google Scholar]
- 15.Carlin J.L., Tosh D.K., Xiao C., Piñol R.A., Chen Z., Salvemini D., Gavrilova O., Jacobson K.A., and Reitman M.L. Peripheral adenosine A3 receptor activation causes regulated hypothermia in mice that is dependent on central histamine H1 receptors. J. Pharmacol. Exp. Therap. 356:474–482, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Carlin J.L., Tosh D.K., Panyutin A., Piñol R.A., Jacobson K.A., Gavrilova O., and Reitman M.L. A1 and A3 adenosine receptors mediate hypothermia via distinct mechanisms. Soc. for Neuroscience Abstract 285.01, Oct. 19, 2015, Chicago, IL [Google Scholar]
- 17.Cekic C., and Linden J. Purinergic regulation of the immune system. Nat. Rev. Immunol. 16:177–192, 2016 [DOI] [PubMed] [Google Scholar]
- 18.Åstrand A.B.M., et al. The discovery of a selective and potent A2A agonist with extended lung retention. Pharma. Res. Per. 3:e00134, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Gao Z., Li Z., Baker S.P., et al. Novel short-acting A2A adenosine receptor agonists for coronary vasodilation: inverse relationship between affinity and duration of action of A2A agonists. J. Pharmacol. Exp. Ther. 298:209–218, 2001 [PubMed] [Google Scholar]
- 20.Kim D.G., and Bynoe M.S. A2A adenosine receptor modulates drug efflux transporter P-glycoprotein at the blood-brain barrier. J. Clin. Invest. 126:1717–1733, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Field J.J., Lin G., Okam M.M., Majerus E., Keefer J., Onyekwere O., Ross A., Campigotto F., Neuberg D., Linden J., and Nathan D.G. Sickle cell vaso-occlusion causes activation of iNKT cells that is decreased by the adenosine A2A receptor agonist regadenoson. Blood. 121:3329–3334, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Montes G.C., Hammes N., Rocha M.D., Montagnoli T.L., Fraga C.A., Barreiro E.J., Sudo R.T., and Zapata-Sudo G. Treatment with adenosine receptor agonist ameliorates pain induced by acute and chronic inflammation. J. Pharmacol. Exp. Ther. 358:315–323, 2016 [DOI] [PubMed] [Google Scholar]
- 23.Sivak K.V., Vasina A.V., Egorova V.V., Tsevtkova V.B., Kuzmicha N.N., Savinaa V.A., and Kiselev O.I. Adenosine A2A receptor as a drug target for treatment of sepsis. Mol. Biol. 50:200–212, 2016 [DOI] [PubMed] [Google Scholar]
- 24.Jazayeri A., Andrews S.P., and Marshall F.H. Structurally enabled discovery of adenosine A2A receptor antagonists. Chem. Rev. 2016; DOI: 10.1021/acs.chemrev.6b00119 [DOI] [PubMed] [Google Scholar]
- 25.Gao Z.G., Balasubramanian R., Kiselev E., Wei Q., and Jacobson K.A. Probing biased/partial agonism at the G protein-coupled A2B adenosine receptor. Biochem. Pharmacol. 90:297–306, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Alnouri M.W., Jepards S., Casari A., Schiedel A.C., Hinz S., and Müller C.E. Selectivity is species-dependent: characterization of standard agonists and antagonists at human, rat, and mouse adenosine receptors. Purinergic Signal. 11:389–407, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Yang H., Avila M.Y., Peterson-Yantorno K., Coca-Prados M., Stone R.A., Jacobson K.A., and Civan M.M. The cross-species A3 adenosine-receptor antagonist MRS 1292 inhibits adenosine-triggered human nonpigmented ciliary epithelial cell fluid release and reduces mouse intraocular pressure. Curr. Eye Res. 30:747–754, 2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Wan T.C., Kreckler L.M., Van Orman J., and Auchampach J.A. Pharmacological characterization of recombinant mouse adenosine receptors expressed in HEK 293 cells. Poster, 4th International Symposium of Nucleosides and Nucleotides, Chapel Hill, NC, June9–11th, 2004 [Google Scholar]
- 29.Okamura T., Kurogi Y., Hashimoto K., Sato S., Nishikawa H., Kiryu K., and Nagao Y. Structure-activity relationships of adenosine A3 receptor ligands: new potential therapy for the treatment of glaucoma. Bioorg. Med. Chem. Lett. 14:3775–3779, 2004 [DOI] [PubMed] [Google Scholar]
- 30.Daines B.S., Kent A.R., McAleer M.S., and Crosson C.E. Intraocular adenosine levels in normal and ocular-hypertensive patients. J. Ocul. Pharmacol. Ther. 19:113, 2003 [DOI] [PubMed] [Google Scholar]
- 31.Welihinda A.A., Kaur M., Greene K., Zhai Y., and Amento E.P. The adenosine metabolite inosine is a functional agonist of the adenosine A2A receptor with a unique signaling bias. Cell Signal. 28:552–560, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Li A., Leung C.T., Peterson-Yantorno K., Stamer W.D., and Civan M.M. Cytoskeletal dependence of adenosinetriphosphate release by human trabecular meshwork cells. Invest. Ophthalmol. Vis. Sci. 52:7996–8005, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Avila M.Y., Stone R.A., and Civan M.M. A1-, A2- and A3-Subtype adenosine receptors modulate intraocular pressure in the mouse. Br. J. Pharmacol. 134:241–245, 2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Yegutkin G.G. Nucleotide- and nucleoside-converting ectoenzymes: important modulators of purinergic signalling cascade. Biochim. Biophys. Acta. 1783:673–694, 2008 [DOI] [PubMed] [Google Scholar]
- 35.Li A., Banerjee J., Peterson-Yantorno K., Stamer W.D., Leung C.T., and Civan M.M. Effects of cardiotonic steroids on trabecular meshwork cells: search for mediator of ouabain-enhanced outflow facility. Exp. Eye Res. 96:4–12, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Fleischhauer J.C., Mitchell C.H., Stamer W.D., Karl M.O., Peterson-Yantorno K., and Civan M.M. Common actions of adenosine receptor agonists in modulating human trabecular meshwork cell transport. J. Membr. Biol. 193:121–136, 2003 [DOI] [PubMed] [Google Scholar]
- 37.Freddo T.F., Bartels S.P., Barsotti M.F., and Kamm R.D. The source of proteins in the aqueous humor of the normal rabbit. Invest. Ophthalmol. Vis. Sci. 31:125–137, 1990 [PubMed] [Google Scholar]
- 38.Barsotti M.F., Bartels S.P., Freddo T.F., and Kamm R.D. The source of protein in the aqueous humor of the normal monkey eye. Invest. Ophthalmol. Vis. Sci. 33:581–595, 1992 [PubMed] [Google Scholar]
- 39.Bert R.J., Caruthers S.D., Jara H., Krejza J., Melhem E.R., Kolodny N.H., Patz S., and Freddo T.F. Demonstration of an anterior diffusional pathway for solutes in the normal human eye with high spatial resolution contrast-enhanced dynamic MR imaging. Invest. Ophthalmol. Vis. Sci. 47:5153–5162, 2006 [DOI] [PubMed] [Google Scholar]
- 40.Civan M.M. Formation of the aqueous humor: transport components and their integration. In: Civan M.M., ed. Eye's Aqueous Humor: From Secretion to Glaucoma, 2nd ed. San Diego: Academic Press; Curr. Topics Membr. 62:1–45, 2008 [Google Scholar]
- 41.Bill A. The role of ciliary blood flow and ultrafiltration in aqueous humor formation. Exp. Eye Res. 16:287–298, 1973 [DOI] [PubMed] [Google Scholar]
- 42.Podos S.M. Effect of dipyridamole on prostaglandin-induced ocular hypertension in rabbits. Invest. Ophthalmol. Vis. Sci. 18:646–648, 1979 [PubMed] [Google Scholar]
- 43.Zhong Y., Yang Z., Huang W.C., and Luo X. Adenosine, adenosine receptors and glaucoma: an updated overview. Biochim. Biophys. Acta 1830:2882–2890, 2013 [DOI] [PubMed] [Google Scholar]
- 44.Donegan R.K., Raquel L., and Lieberman R.L. Discovery of molecular therapeutics for glaucoma: challenges, successes, and promising directions. J. Med. Chem. 59:788–809, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Sanderson J., Dartt D.A., Trinkaus-Randall V., Pintor J., Civan M.M., Delamere N.A., Fletcher E.L., Salt T.E., Grosche A., and Mitchell C.H. Purines in the eye: recent evidence for the physiological and pathological role of purines in the RPE, retinal neurons, astrocytes, Müller cells, lens, trabecular meshwork, cornea and lacrimal gland. Exp. Eye Res. 127:270–279, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.https://clinicaltrials.gov (accessed June8, 2016)
- 47.McLaughlin C.W., Karl M.O., Zellhuber-McMillan S., Wang Z., Do C.W., Leung C.T., Li A., Stone R.A., Macknight A.D., and Civan M.M. Electron probe X-ray microanalysis of intact pathway for human aqueous humor outflow. Am. J. Physiol. Cell Physiol. 295:C1083–C1091, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Crosson C.E., Sloan C.F., and Yates P.W. Modulation of conventional outflow facility by the adenosine A1 agonist N6-cyclohexyladenosine. Invest. Ophthalmol. Vis. Sci. 46:3795–3799, 2005 [DOI] [PubMed] [Google Scholar]
- 49.Kim N., Crosson C., Supuran C., McCauley T., Southan G., Baumgartner R., and McVicar W. INO-8875, an adenosine A1 agonist, in development for open-angle glaucoma reduces IOP in three rabbit models. Invest. Ophthalmol. Vis. Sci. 50:4061, 2009 [Google Scholar]
- 50.Bradley J.M., Vranka J., Colvis C.M., Conger D.M., Alexander J.P., Fisk A.S., Samples J.R., and Acott T.S. Effect of matrix metalloproteinases activity on outflow in perfused human organ culture. Invest. Ophthalmol. Vis. Sci. 39:2649–2658, 1998 [PubMed] [Google Scholar]
- 51.Mor M., Shalev A., Dror S., Pikovsky O., Beharier O., Moran A., Katz A., and Etzion Y. INO-8875, a highly selective A1 adenosine receptor agonist: evaluation of chronotropic, dromotropic, and hemodynamic effects in rats. J. Pharmacol. Exp. Ther. 344:59–67, 2013 [DOI] [PubMed] [Google Scholar]
- 52.Kaufman P.L., and Rasmussen C.A. Advances in glaucoma treatment and management: outflow drugs. Invest. Ophthalmol. Vis. Sci. 53:2495–2500, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Konno T., Murakami A., Uchibori T., Nagai A., Kogi K., and Nakahata N. Involvement of adenosine A2A receptor in intraocular pressure decrease induced by 2-(1-octyn-1-yl)adenosine or 2-(6-cyano-1-hexyn-1-yl)adenosine. J. Pharmacol. Sci. 97:501–509, 2005 [DOI] [PubMed] [Google Scholar]
- 54.Clinical Data, Inc. licenses adenosine A2A agonist to Santen for development of ophthalmic treatments. Santen Pharmaceuticals, 2010. www.santen.com/en/news/20100430.pdf (accessed March21, 2016)
- 55.Ahmad S., Fatteh N., El-Sherbiny N.M., Naime M., Ibrahim A.S., El-Sherbini A.M., El-Shafey S.A., Khan S., Fulzele S., Gonzales J., and Liou G.I. Potential role of A2A adenosine receptor in traumatic optic neuropathy. J. Neuroimmunol. 264:54–64, 2013 [DOI] [PubMed] [Google Scholar]
- 56.Madeira M.H., Elvas F., Boia R., Gonçalves F.Q., Cunha R.A., Ambrósio A.F., and Santiago A.R. Adenosine A2AR blockade prevents neuroinflammation-induced death of retinal ganglion cells caused by elevated pressure. J. Neuroinflamm. 12:115, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Mac Nair C.E., and Nickells R.W. Neuroinflammation in glaucoma and optic nerve damage. Prog. Mol. Biol. Transl. Sci. 134:343–363, 2015 [DOI] [PubMed] [Google Scholar]
- 58.Levkovitch-Verbin H. Retinal ganglion cell apoptotic pathway in glaucoma: initiating and downstream mechanisms. Prog. Brain Res. 220:37–57, 2015 [DOI] [PubMed] [Google Scholar]
- 59.Xu H., and Chen M. Targeting the complement system for the management of retinal inflammatory and degenerative diseases. Eur. J. Pharmacol. 2016. [Epub ahead of print]; DOI: 10.1016/j.ejphar.2016.03.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Madeira M.H., Boia R., Elvas F., Martins T., Cunha R.A., Ambrósio A.F., and Santiago A.R. Selective A2A receptor antagonist prevents microglia-mediated neuroinflammation and protects retinal ganglion cells from high intraocular pressure-induced transient ischemic injury. Transl. Res. 169:112–128, 2016 [DOI] [PubMed] [Google Scholar]
- 61.Tian B., Gabelt B.T., Crosson C.E., and Kaufman P.L. Effects of adenosine agonists on intraocular pressure and aqueous humor dynamics in cynomolgus monkeys. Exp. Eye Res. 64:979–989, 1997 [DOI] [PubMed] [Google Scholar]
- 62.Ahmad S., Elsherbiny N.M., Bhatia K., Elsherbini A.M., Fulzele S., and Liou G.I. Inhibition of adenosine kinase attenuates inflammation and neurotoxicity in traumatic optic neuropathy. J. Neuroimmunol. 277:96–104, 2014 [DOI] [PubMed] [Google Scholar]
- 63.Toti K., Osborne D., Ciancetta A., Boison D., and Jacobson K.A. South (S)- and North (N)-methanocarba-7-deazaadenosine analogues as inhibitors of human adenosine kinase. J. Med. Chem. 59:6860-6877, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Civan M.M., and Macknight A.D. The ins and outs of aqueous humour secretion. Exp. Eye Res. 78:625–631, 2004 [DOI] [PubMed] [Google Scholar]
- 65.Wang Z., Do C.W., Avila M.Y., Stone R.A., Jacobson K.A., and Civan M.M. Barrier qualities of the mouse eye to topically applied drugs. Exp. Eye Res. 85:105–112, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Wang Z., Do C.W., Avila M.Y., Peterson-Yatorno K., Stone R.A., Gao Z.G., Joshi B.V., Besada P., Jeong L.S., Jacobson K.A., and Civan M.M. Nucleoside-derived antagonists to A3 adenosine receptors lower mouse intraocular pressure and act across species. Exp. Eye Res. 90:146–154, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Li A., Banerjee J., Leung C.T., Peterson-Yantorno K., Stamer W.D., and Civan M.M. Mechanisms of ATP release, the enabling step in purinergic dynamics. Cell. Physiol. Biochem. 28:1135–1144, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Avila M.Y., Stone R.A., and Civan M.M. Knockout of A3 adenosine receptors reduces mouse intraocular pressure. Invest. Ophthalmol. Vis. Sci. 43:3021–3026, 2002 [PubMed] [Google Scholar]
- 69.Schlotzer-Schrehardt U., Zenkel M., Decking U., Haubs D., Kruse F.E., Jünemann A., Coca-Prados M., and Naumann G.O. Selective upregulation of the A3 adenosine receptor in eyes with pseudoexfoliation syndrome and glaucoma. Invest. Ophthalmol. Vis. Sci. 46:2023–2034, 2005 [DOI] [PubMed] [Google Scholar]
- 70.www.acornbio.com/pipeline/acn1052.html (accessed March21, 2016)
- 71.www.palobiofarma.com (accessed March21, 2016)
- 72.Avni I., Garzozi H.J., Barequet I.S., Segev F., Varssano D., Sartani G., Chetrit N., Bakshi E., Zadok D., Tomkins O., Litvin G., Jacobson K.A., Fishman S., Harpaz Z., Farbstein M., Bar-Yehuda S., Silverman M.H., Kerns W.D., Cohn I., and Fishman P. Treatment of dry eye syndrome with orally-administered CF101: data from a Phase 2 clinical trial. Ophthalmology. 117:1287–1293, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Fishman P., Cohen S., and Bar-Yehuda S. Targeting the A3 adenosine receptor for glaucoma treatment (Review). Mol. Med. Rep. 7:1723–1725, 2013 [DOI] [PubMed] [Google Scholar]
- 74.Gallo-Rodriguez C., Ji X.-D., Melman N., Siegman B.D., Sanders L.H., Orlina J., Fischer B., Pu Q.-L., Olah M.E., van Galen P.J.M., Stiles G.L., and Jacobson K.A. Structure-activity relationships of N6-benzyladenosine-5′-uronamides as A3-selective adenosine agonists. J. Med. Chem. 37:636–646, 1994 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kim H.O., Ji X.D., Siddiqi S.M., Olah M.E., Stiles G.L., and Jacobson K.A. 2-Substitution of N6-benzyladenosine-5′-uronamides enhances selectivity for A3-adenosine receptors. J. Med. Chem. 37:3614–3621, 1994 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.http://ir.canfite.com/press-releases/detail/761 (accessed July7, 2016)
- 77.Trincavelli M.L., Tuscano D., Marroni M., Falleni A., Gremigni V., Ceruti S., Abbracchio M.P., Jacobson K.A., Cattabeni F., and Martini C. A3 adenosine receptors in human astrocytoma cells: agonist-mediated desensitization, internalization and down-regulation. Mol. Pharmacol. 65:1373–1384, 2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Rudich N., Dekel O., and Sagi-Eisenberg R. Down-regulation of the A3 adenosine receptor in human mast cells upregulates mediators of angiogenesis and remodeling. Mol. Immunol. 65:25–33, 2015 [DOI] [PubMed] [Google Scholar]
- 79.Little J.W., Ford A., Symons-Liguori A.M., Chen Z., Janes K., Doyle T., Xie J., Luongo L., Tosh D.K., Maione S., Bannister K., Dickenson A., Vanderah T.W., Porreca F., Jacobson K.A., and Salvemini D. Endogenous adenosine A3 receptor activation selectively alleviates persistent pain states. Brain. 138:28–35, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Zhang K., Zhang L., and Weinreb R.N. Ophthalmic drug discovery: novel targets and mechanisms for retinal diseases and glaucoma. Nat. Rev. Drug Disc. 11:541–559, 2012 [DOI] [PubMed] [Google Scholar]
- 81.Besada P., Mamedova L.K., Palaniappan K.K., Gao Z.G., Joshi B.V., Jeong L.S., Civan M.M., and Jacobson K.A. Nucleoside prodrugs of A3 adenosine receptor agonists and antagonists. Coll. Czech. Chem. Comm. 71:912–928, 2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Zhang M., Hu H., Zhang X., Lu W., Lim J., Eysteinsson T., Jacobson K.A., Laties A.M., and Mitchell C.H. The A3 adenosine receptor attenuates the calcium rise triggered by NMDA receptors in retinal ganglion cells. Neurochem. Int. 56:35–41, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Crosson C.E., Debenedetto R., and Gidday J.M. Functional evidence for retinal adenosine receptors. J. Ocul. Pharmacol. Therap. 10:499–507, 1994 [DOI] [PubMed] [Google Scholar]
- 84.Bar-Yehuda S., Luger D., Ochaion A., Cohen S., Patokaa R., Zozulya G., Silver P.B, de Morales J.M., Caspi R.R., and Fishman P. Inhibition of experimental auto-immune uveitis by the A3 adenosine receptor agonist CF101. Int. J. Mol. Med. 28:727–731, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Vindeirinho J., Santiago A.R., Cavadas C., Ambrósio A.F., and Santos P.F. The adenosinergic system in diabetic retinopathy. J. Diabetes Res. 2016:4270301, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Brunschweiger A., Christa E., and Müller C.E. P2 Receptors activated by uracil nucleotides—an update. Curr. Med. Chem. 13:289–312, 2006 [DOI] [PubMed] [Google Scholar]
- 87.Hoffmann K., Lutz D.A., Straßburger J., Baqi Y., Müller C.E., and von Kügelgen I. Competitive mode and site of interaction of ticagrelor at the human platelet P2Y12-receptor. J. Thromb. Haemost. 12:1898–1905, 2014 [DOI] [PubMed] [Google Scholar]
- 88.Pintor J., Sánchez-Nogueiro J., Irazu M., Mediero A., Peláez T., and Peral A. Immunolocalisation of P2Y receptors in the rat eye. Purinergic Signal. 1:83–90, 2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Guzman-Aranguez A., Santano C., Martin-Gil A., Fonseca B., and Pintor J. Nucleotides in the eye: focus on functional aspects and therapeutic perspectives. J. Pharmacol. Exp. Ther. 345:331–341, 2013 [DOI] [PubMed] [Google Scholar]
- 90.Pannicke T., Frommherz I., Biedermann B., Wagner L., Sauer K., Ulbricht E., et al. Differential effects of P2Y1 deletion on glial activation and survival of photoreceptors and amacrine cells in the ischemic mouse retina. Cell Death Dis. 5:e1353, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Ohtomo K., Hodges R., Shatos M., and Dartt D. Location and function of P2Y receptors in lacrimal gland myoepithelial cells. Invest. Ophthalmol. Vis. Sci. 52:3704, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Pintor J., Bautista A., Carracedo G., and Peral A. UTP and diadenosine tetraphosphate accelerate wound healing in the rabbit cornea. Ophthal. Physiol. Opt. 24:186–193, 2004 [DOI] [PubMed] [Google Scholar]
- 93.Mediero A., Peral A., and Pintor J. Dual roles of diadenosine polyphosphates in corneal epithelial cell migration. Invest. Ophthalmol. Vis. Sci. 47:4500–4506, 2006 [DOI] [PubMed] [Google Scholar]
- 94.Podos S. M., Wang R., Boyer J. L., Yerxa B. R. and Serle J. B. Effect of the P2Y14 receptor agonist, UDP-glucose, on intraocular pressure (IOP) in glaucomatous monkey eyes. Invest. Ophthalmol. Vis. Sci. 44: ARVO E-Abstract 4404; 2003 [Google Scholar]
- 95.Castany M., Pintor J., et al. Glaucoma patients present increased levels of diadenosine tetraphosphate, Ap4A, in the aqueous humour. Exp. Eye Res. 92:221–226, 2011 [DOI] [PubMed] [Google Scholar]
- 96.Loma P., Guzman-Aranguez A., Pérez de Lara M.J., and Pintor J. Diadenosine tetraphosphate induces tight junction disassembly thus increasing corneal epithelial permeability. Br. J. Pharmacol. 172:1045–1058, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Loma P., Guzman-Aranguez A., Perez de Lara M.J., and Pintor J. Diadenosine tetraphosphate improves adrenergic anti-glaucomatous drug delivery and efficiency. Exp. Eye Res. 134:141–147, 2015 [DOI] [PubMed] [Google Scholar]
- 98.Guzman-Aranguez A., and Pintor J. Focus on molecules: purinergic P2Y2 receptor. Exp. Eye Res. 91:83–84, 2012 [DOI] [PubMed] [Google Scholar]
- 99.Fujihara T., Murakami T., Fujita H., Nakamura M., and Nakata K. Improvement of corneal barrier function by the P2Y2 agonist INS365 in a rat dry eye model. Invest. Ophthalmol. Vis. Sci. 42:96–100, 2001 [PubMed] [Google Scholar]
- 100.Nakamura M., Imanaka T., and Sakamoto A. Diquafosol ophthalmic solution for dry eye treatment. Adv. Ther. 29:579–589, 2012 [DOI] [PubMed] [Google Scholar]
- 101.Yokoi N., Sonomura Y., Kato H., Komuro A., and Kinoshita S. Three percent diquafosol ophthalmic solution as an additional therapy to existing artificial tears with steroids for dry-eye patients with Sjögren's syndrome. Eye. 29:1204–1212, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Gong L., Sun X., Ma Z., Wang Q., Xu X., Chen X., Shao Y., Yao K., Tang L., Gu Y., et al. A randomised, parallel-group comparison study of diquafosol ophthalmic solution in patients with dry eye in China and Singapore. Br. J. Ophthalmol. 99:903–908, 2015 [DOI] [PubMed] [Google Scholar]
- 103.Keating G.M. Diquafosol ophthalmic solution 3%: a review of its use in dry eye. Drugs. 75:911–922, 2015 [DOI] [PubMed] [Google Scholar]
- 104.Bremond-Gignac D., Gicquel J.J., and Chiambaretta F. Pharmacokinetic evaluation of diquafosol tetrasodium for the treatment of Sjögren's syndrome. Expert Opin. Drug Metab. Toxicol. 10:905–913, 2014 [DOI] [PubMed] [Google Scholar]
- 105.Gendaszewska-Darmach E., and Kucharska M. Nucleotide receptors as targets in the pharmacological enhancement of dermal wound healing. Purinergic Signal. 7:193–206, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Ginsburg-Shmuel T., Haas M., Grbic D., Arguin G., Nadel Y., Gendron F.P., Reiser G., and Fischer B. UDP made a highly promising stable, potent, and selective P2Y6-receptor agonist upon introduction of a boranophosphate moiety. Bioorg. Med. Chem. 20:5483–5495, 2012 [DOI] [PubMed] [Google Scholar]
- 107.Markovskaya A., Crooke A., Guzman-Aranguez A.I., Peral A., Ziganshin A.U., and Pintor J. Hypotensive effect of UDP on intraocular pressure in rabbits. Eur. J. Pharmacol. 579:93–97, 2008 [DOI] [PubMed] [Google Scholar]
- 108.Haas M., Ginsburg-Shmuel T., Fischer B., Reiser G. 5-OMe-uridine-5′-O-(α-boranodiphosphate), a novel nucleotide derivative highly active at the human P2Y6 receptor protects against death-receptor mediated glial apoptosis. Neurosci. Lett. 88:384–392, 2014 [DOI] [PubMed] [Google Scholar]
- 109.Taguchi M., Shinozaki Y., Kashiwagi K., Shigetomi E., Robaye B., and Koizumi S. Müller cell-mediated neurite outgrowth of the retinal ganglion cells via P2Y6 receptor signals. J. Neurochem. 136:741–751, 2016 [DOI] [PubMed] [Google Scholar]
- 110.Fonseca B., Martínez-Águilam A., Pérez de Lara M.J., Crooke A., and Pintor J. Participation of P2Y2 and P2Y6 purinergic receptors in the physiopathology of glaucoma. Invest. Ophthalmol. Vis. Sci. 55:2425, 2014 [Google Scholar]
- 111.Liang X., Samways D.S.K., Wolf K., Bowles E.A., Richards J.P., Bruno J., Dutertre S., DiPaolo R.J., and Egan T.M. (2015). Quantifying Ca2+ current and permeability in ATP-gated P2X7 receptors. J. Biol. Chem. 290:7930–7942, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Guzman-Aranguez A., Gasull X., Diebold Y., and Pintor J. Purinergic receptors in ocular inflammation. Mediators Inflamm. 2014:320906, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Beckel J.M., Argall A.J., Lim J.C., Xia J., Lu W., Coffey E.E., et al. Mechanosensitive release of ATP through pannexin channels and mechanosensitive upregulation of pannexin channels in optic nerve head astrocytes: a mechanism for purinergic involvement in chronic strain. Glia. 62:1486–1501, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Mayo C., Ren R., Rich C., Stepp M.A., and Trinkaus-Randall V. Regulation by P2X7: epithelial migration and stromal organization in the cornea. Invest. Ophthalmol. Vis. Sci. 49:4384–4391, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Li A., Leung C.T., Peterson-Yantorno K., Stamer W.D., Mitchell C.H., and Civan M.M. Mechanisms of ATP release by human trabecular meshwork cells, the enabling step in purinergic regulation of aqueous humor outflow. J. Cell. Physiol. 227:172–182, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Puthussery T., and Fletcher E.L. P2X2 receptors on ganglion and amacrine cells in cone pathways of the rat retina. J. Comp. Neurol. 496:595e609, 2006 [DOI] [PubMed] [Google Scholar]
- 117.Hodges R.R., Vrouvlianis J., Scott R., and Dartt D.A. Identification of P2X3 and P2X7 purinergic receptors activated by ATP in rat lacrimal gland. Invest. Ophthalmol. Vis. Sci. 52:3254–3263, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Reichenbach A., and Bringmann A. Purinergic signaling in retinal degeneration and regeneration. Neuropharmacology. 104:194–211, 2016 [DOI] [PubMed] [Google Scholar]
- 119.www.businesswire.com/news/home/20160609006459/en/Merck-Acquire-Afferent-Pharmaceuticals (accessed June30, 2016)
- 120.www.chemiphar.co.jp/english/ir/doc/2016/20160617_e.pdf (accessed June30, 2016)
- 121.North R.A., and Jarvis M.F. P2X receptors as drug targets. Mol. Pharmacol. 83:759–769, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Ali Z., Laurijssens B., Ostenfeld T., McHugh S., Stylianou A., Scott-Stevens P., Hosking L., Dewit O., Jill C., Richardson J.C., and Chen C. Pharmacokinetic and pharmacodynamics profiling of a P2X7 receptor allosteric modulator GSK1482160 in healthy human subjects. Br. J. Clin. Pharmacol. 75:197–207, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Hu H., Lu W., Zhang M., Xhang X., Argall A.J., Patel S., Lee G.E., Kim Y.C., Jacobson K.A., Laties A.M., and Mitchell C.H. Stimulation of the P2X7 receptor kills rat retinal ganglion cells in vivo. Exp. Eye Res. 91:425–432, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Sugiyama T., Oku H., Shibata M., Fukuhara M., Yoshida H., and Ikeda T. Involvement of P2X7 Receptors in the hypoxia-induced death of rat retinal neurons. Invest. Ophthalmol. Vis. Sci. 51:3236–3243, 2010 [DOI] [PubMed] [Google Scholar]
- 125.Sugiyama T., Lee S.Y., Horie T., Oku H., Takai S., Tanioka H., Kuriki Y., Kojima S., and Ikeda T. P2X7 receptor activation may be involved in neuronal loss in the retinal ganglion cell layer after acute elevation of intraocular pressure in rats. Mol. Vis. 19:2080–2091, 2013 [PMC free article] [PubMed] [Google Scholar]
- 126.Sugiyama T. Role of P2X7 receptors in the development of diabetic retinopathy. World J. Diabetes. 5.2:141, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Mac Nair C.E., Schlamp C.L., Montgomery A.D., Shestopalov V.I., and Nickells R.W. Retinal glial responses to optic nerve crush are attenuated in Bax-deficient mice and modulated by purinergic signaling pathways. J. Neuroinflamm. 13:93, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Sakamoto K., Endo K., Suzuki T., Fujimura K., Kurauchi Y., Mori A., Nakahara T., and Ishii K. P2X7 receptor antagonists protect against N-methyl-D-aspartic acid-induced neuronal injury in the rat retina. Eur. J. Pharmacol. 756:52–58, 2015 [DOI] [PubMed] [Google Scholar]
- 129.Ory D., Celen S., Gijsbers R., Van Den Haute C., Postnov A., Koole M., Vandeputte C., Andrés J.I., De Angelis M., Langlois X., and Bhattacharya A. Preclinical evaluation of a P2X7 receptor selective radiotracer: PET studies in a rat model with local overexpression of the human P2X7 receptor and in non-human primates. J. Nucl. Med. 2016; DOI: 10.2967/jnumed.115.169995 [DOI] [PubMed] [Google Scholar]






