Open Access
Physiology and Pharmacology  |   June 2018
Myopia-Inhibiting Concentrations of Muscarinic Receptor Antagonists Block Activation of Alpha2A-Adrenoceptors In Vitro
Author Affiliations & Notes
  • Brittany J. Carr
    Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
    Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
    Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
    Department of Neuroscience, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
  • Koichiro Mihara
    Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
    Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
  • Rithwik Ramachandran
    Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
    Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
    Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
  • Mahmoud Saifeddine
    Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
    Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
  • Neil M. Nathanson
    Department of Pharmacology, University of Washington, Seattle, Washington, United States
  • William K. Stell
    Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
    Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
    Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
  • Morley D. Hollenberg
    Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
    Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
    Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
  • Correspondence: William K. Stell, Health Sciences Centre B101, 3330 Hospital Drive NW, Calgary, Alberta, Canada, T2N 4N1; wstell@ucalgary.ca
  • Brittany J. Carr, Eye Care Centre Room 330, 2550 Willow Street, Vancouver, British Columbia, Canada, V5Z 3N9; bcarr@ucalgary.ca
Investigative Ophthalmology & Visual Science June 2018, Vol.59, 2778-2791. doi:https://doi.org/10.1167/iovs.17-22562
  • Views
  • PDF
  • Share
  • Tools
    • Alerts
      ×
      This feature is available to authenticated users only.
      Sign In or Create an Account ×
    • Get Citation

      Brittany J. Carr, Koichiro Mihara, Rithwik Ramachandran, Mahmoud Saifeddine, Neil M. Nathanson, William K. Stell, Morley D. Hollenberg; Myopia-Inhibiting Concentrations of Muscarinic Receptor Antagonists Block Activation of Alpha2A-Adrenoceptors In Vitro. Invest. Ophthalmol. Vis. Sci. 2018;59(7):2778-2791. https://doi.org/10.1167/iovs.17-22562.

      Download citation file:


      © ARVO (1962-2015); The Authors (2016-present)

      ×
  • Supplements
Abstract

Purpose: Myopia is a refractive disorder that degrades vision. It can be treated with atropine, a muscarinic acetylcholine receptor (mAChR) antagonist, but the mechanism is unknown. Atropine may block α-adrenoceptors at concentrations ≥0.1 mM, and another potent myopia-inhibiting ligand, mamba toxin-3 (MT3), binds equally well to human mAChR M4 and α1A- and α2A-adrenoceptors. We hypothesized that mAChR antagonists could inhibit myopia via α2A-adrenoceptors, rather than mAChR M4.

Methods: Human mAChR M4 (M4), chicken mAChR M4 (cM4), or human α2A-adrenergic receptor (hADRA2A) clones were cotransfected with CRE/promoter-luciferase (CRE-Luc; agonist-induced luminescence) and Renilla luciferase (RLuc; normalizing control) into human cells. Inhibition of normalized agonist-induced luminescence by antagonists (ATR: atropine; MT3; HIM: himbacine; PRZ: pirenzepine; TRP: tropicamide; OXY: oxyphenonium; QNB: 3-quinuclidinyl benzilate; DIC: dicyclomine; MEP: mepenzolate) was measured using the Dual-Glo Luciferase Assay System.

Results: Relative inhibitory potencies of mAChR antagonists at mAChR M4/cM4, from most to least potent, were QNB > OXY ≥ ATR > MEP > HIM > DIC > PRZ > TRP. MT3 was 56× less potent at cM4 than at M4. Relative potencies of mAChR antagonists at hADRA2A, from most to least potent, were MT3 > HIM > ATR > OXY > PRZ > TRP > QNB > MEP; DIC did not antagonize.

Conclusions: Muscarinic antagonists block hADRA2A signaling at concentrations comparable to those used to inhibit chick myopia (≥0.1 mM) in vivo. Relative potencies at hADRA2A, but not M4/cM4, correlate with reported abilities to inhibit chick form-deprivation myopia. mAChR antagonists might inhibit myopia via α2-adrenoceptors, instead of through the mAChR M4/cM4 receptor subtype.

Myopia (near- or short-sightedness) is a common eye condition that increases the risk of ocular pathology as it progresses.1,2 As its prevalence continues to rise,3 so does the need for an effective therapy. Topical atropine is effective against myopia in children4; however, treatment requires daily application, and the dosage most commonly prescribed for many years (1%) may induce allergic reactions, as well as muscarinic acetylcholine receptor (mAChR) M3-mediated side effects—mydriasis, photophobia, cycloplegia—and possibly early presbyopia.4,5 Atropine administered topically at concentrations of 1% loses effectiveness over a period of 1 to 2 years; and upon cessation, “rebound” may occur, wherein myopia returns at a faster rate than in untreated eyes6,7—possibly because of the desensitization of target receptors in response to a high concentration of anticholinergic drug. Recent studies have supported the use of 0.01% atropine to treat childhood myopia.7,8 This comparatively lower dose of atropine appears to retain its effectiveness for inhibiting myopia, and results in a reduced severity of side effects such as allergies, loss of accommodation, and rebound. Some of those treated with 0.01% atropine still complain of muscarinic receptor (mAChR) blockade-induced complications, including photophobia and blurred vision; however, those symptoms are not severe enough to prompt a discontinuation of treatment. Therefore, 0.01% atropine is currently the most favored concentration prescribed by clinicians both in Southeast Asia and North America.912 
Atropine is a potent nonselective mAChR antagonist. Thus, it has been widely assumed to inhibit myopia via blockade of mAChRs. Another potent mAChR antagonist, mamba toxin-3 (MT3), is strongly selective for mAChR M4 over other mAChR subtypes in mammals,13 and inhibits form-deprivation myopia (FDM) in the chick at far lower concentrations than those required for atropine (17.5–70 pmol versus 20–2000 nmol per injection, respectively).1416 Therefore, given the ability of MT3 to block myopia development in the chick model, it has been suggested that the mAChR M4 receptor subtype is the target for myopia-controlling action of both atropine and MT3.14,17 It is important to point out, however, that a direct mechanistic link between mAChR systems in the eye and the mAChR antagonist properties of atropine or MT3 for inhibition of myopia has never been demonstrated. 
Moreover, some evidence suggests that the ability of atropine to inhibit myopia development may not involve mAChRs at all.18 For example, most mAChR antagonists, even very potent ones such as 3-quinuclidinyl benzilate (QNB) or dicyclomine, do not inhibit myopia in chicks, and those muscarinic antagonists that do so inhibit myopia require concentrations that can be orders of magnitude above their inhibition constants for blocking mAChRs (Table 1).16,19 Furthermore, ablating ≥90% of choline acetyltransferase (ChAT)-containing amacrine cells—the only known source of retinal acetylcholine (ACh; the natural agonist at mAChRs)—in the chick has no effect on the eyes' ability to achieve emmetropia, nor does elimination of retinal ChAT impair myopia inhibition by atropine.20 Treatment with myopia-inhibiting concentrations of atropine in chick retina–RPE–choroid–sclera preparations causes a massive, nonspecific release of retinal neurotransmitters (including dopamine), and induces spreading depression that affects the entire retina, not just mAChR-related circuits.21 Finally, significant myopia-related changes in eye size in the tree shrew and chick do not result in significant changes in mRNA or protein expression of any mAChR subtype in the retina,22,23 nor does myopia-induced axial elongation result in changes in the concentration of retinal ACh, or its metabolite choline.24 Results such as these have led to arguments that favor a nonretinal mechanism of atropine-mediated myopia inhibition, with mAChR binding in the choroid or sclera suggested as the most likely alternative. One way to determine whether the molecular targets of anti-myopia therapy—that are modified directly by treatment with mAChR antagonists—are located in these tissues, is to look for changes in receptor density upon induction of experimental myopia and during atropine treatment. There is no change, however, in mAChR mRNA expression in either the choroid or sclera, upon induction of experimental myopia in intact eyes of tree shrew and chick.22,23 A second method to determine whether the choroid or sclera is directly responsible for atropine-mediated myopia inhibition is to examine the responses to atropine in these tissues specifically, in in vitro preparations of eyecups lacking retina. In such preparations, the action of atropine to inhibit extracellular matrix production and sulfate incorporation into the glycosaminoglycans of chick scleral cartilage25 and the inhibition of carbachol-induced mouse scleral fibroblast proliferation26 were observed only at high concentrations of atropine (0.5–100 μM in the medium), which are 500- to 10,000-fold higher than those that block mAChRs (Ki = 1–10 nM; Table 1). Finally, while pirenzepine treatment has been shown to result in changes in choroidal thickness when applied to in vitro preparations of chick eyecups comprising the RPE–choroid–sclera, it did so only at high concentration (5 mM); other mAChR-targeted antagonists, such as oxyphenonium (1 mM) and dicyclomine (0.6 mM), had no effect on the choroid in these preparations.27 Unfortunately, the effect of atropine on choroidal thickness in chick eyecups lacking retina was not tested. 
Table 1
 
Binding Data for Muscarinic Antagonists Tested at mAChR M4 and cM4 in Our Assay
Table 1
 
Binding Data for Muscarinic Antagonists Tested at mAChR M4 and cM4 in Our Assay
Of particular interest in the above examples is the requirement of high concentrations of atropine and other mAChR antagonists for myopia-inhibiting effects, relative to the subnanomolar affinities (Ki) of many of these antagonists for the M4 mAChR (Table 1). Indeed, even if a drug binds selectively to its preferred receptors with high affinity (e.g., Ki ≤ 1 nM), it is expected that at concentrations two to three orders of magnitude higher it will bind to, and affect the activity of, other (“off-target”) receptors.21 Thus, atropine, while receptor-selective at nanomolar concentrations, would very likely have off-target effects at other receptors at the micro- to millimolar concentrations used to mitigate myopia. Additionally, while it is true that MT3 can inhibit myopia at much lower concentrations than atropine,14,15 this result cannot be interpreted to mean that it does so through M4 or any other mAChR on this basis alone. Although MT3 is selective for the M4 receptor over other mAChRs,13,28 it has a comparably high inhibitory potency (IC50 = 1–10 nM) at human α1A-, α1D-, and α2A-adrenergic receptors; moderate inhibitory potency (IC50 = 25–50 nM) at α1B- and α2C-adrenergic receptors; and low inhibitory potency (IC50 = 200 nM) at the mAChR M1 receptor.29,13 There are also data to indicate that at high concentrations (1–100 μM) atropine can also have antagonist activity at α-adrenoceptors,30,31 although the specific adrenoceptor subtypes to which it may bind remain unknown. 
Chicks are a popular animal model for myopia drug studies. They have high-quality vision, active accommodation and emmetropization, and large eyes for easy intravitreal drug delivery. Diffusers3234 and negative lenses35,36 induce large changes in axial length and refractive error (−5 to −15 diopter [D]) in a matter of days, and results from chick experiments can usually be replicated in mammals and nonhuman primates.37 Interpretations of results, using mAChR antagonists to prevent FDM in the chick, have generally presumed that these ligands work the same way at avian receptors as they do at human or other mammalian receptors. That said, the chick retinal structure and G-protein receptor sequences differ significantly from those of mammals. Chickens lack an orthologue for the mammalian mAChR M1. Instead, chicken M2 (cM2), the orthologue for the mammalian M2 receptor, has some pharmacologic properties of both mammalian M1 and M2 receptors, such as relatively high affinity for pirenzepine.38 Thus, in the chick, one can conclude that MT3 will target the M4 mAChR and not other mAChR subtypes. Furthermore, there are examples of substantial differences in ligand behavior between species, even for mammals, whereby even minor sequence differences can cause receptor-selective ligands to interact differently, with the same G-protein-coupled receptor (GPCR) orthologue.39,40 We therefore considered it likely that MT3, which is highly selective for the mammalian M4 receptor, might have a significantly altered ability to bind to the chick M4 receptor. 
Given the likelihood that the concentrations of atropine used for treating myopia in humans and chicks can act at receptors other than mAChR M4, we focused on the molecular pharmacology of the chick cM4 and human M4 receptors, and the interactions of these receptors with the mAChR-targeted agents that have been used to evaluate myopia development in the chick model. Furthermore, since both MT3 and atropine can potentially interact with α-adrenoceptors, we also explored the interactions of FDM-mitigating agents with the human α2A-adrenergic receptor ADRA2A (hADRA2A) subtype. To test the hypothesis that non-mAChR mechanisms can mediate the inhibition of myopia development by mAChR antagonists in the chick FDM model, we evaluated (1) whether mAChR antagonists have similar relative inhibitory potencies at chick cM4 compared to human M4, and (2) whether mAChR antagonists can bind to, and inhibit signaling by, hADRA2A. We chose to investigate the binding of mAChR antagonists to α2A-adrenoceptors because MT3 has a high affinity for these adrenergic receptors, because this adrenoceptor is the most common α-adrenoceptor subtype in the central nervous system,41 and because some α2-adrenoceptor agonists have been reported to inhibit FDM in the chick (Carr BJ, Stell WK. IOVS 2016;57:ARVO E-Abstract 4738) and guinea pig.42 
Methods and Materials
Experimental Strategy
Our strategy to explore the interactions of the FDM-related antagonists with either mAChR M4/cM4 or hADRA2A was to express the recombinant receptors in a mammalian Lenti-X HEK 293T cell line, in which the only endogenously expressed mAChR (M3) was eliminated by a CRISPR/Cas9-mediated genomic editing approach (CRISPR-M3 cells: CR-M3). The HEK cell line is well recognized for its ability to couple the signaling of GPCRs from multiple species efficiently to multiple effectors, including Gs, Gi, Gq, and G12/13. In this system, the abundance of expressed muscarinic and adrenergic receptors enables coupling to Gs, with a resulting increase in synthesis of cyclic AMP (cAMP).43,44 In turn, the cAMP drives a cAMP response element (CRE)-luciferase reporter construct, such that increased luciferase activity serves as an index of activation of the transfected muscarinic and adrenergic receptors. Carbachol was used to activate the chick and human mAChRs, and clonidine was used as the agonist to activate hADRA2A. The ability of increasing concentrations of antagonists to inhibit the cholinergic and adrenergic responses was used as an index of their relative potencies for blocking the expressed receptors. The relative inhibition constants of the myopia-inhibiting compounds, for blocking either the muscarinic or α2A-adrenergic receptors, were compared directly with the relative ability of the compounds to mitigate myopia in the chick FDM model. 
DNA and Expression Vectors
All receptor clones were expressed in a pcDNA 3.1(+) vector (Invitrogen, ThermoFisher Scientific, Waltham, MA, USA). The cM4 genomic clone was obtained as previously described,18 and the M4 (MAR0400000) and hADRA2A (AR0A2A0000) receptor clones were purchased from the cDNA Resource Center (Bloomsburg University Foundation, Bloomsburg, PA, USA). The cAMP response element luciferase vector (CRE-Luc; pGL4.29[luc2P/CRE/Hygro]) and the constitutively active Renilla luciferase control vector (RLuc; pRL-RK) were purchased from Promega (Madison, WI, USA). 
CRISPR/Cas9 Knockdown of mAChR M3
CRISPR knockout of mAChR M3 in Lenti-X HEK 293T cells (LX293T; Clontech, Mountain View, CA, USA) was performed to ensure that mAChR ligand-mediated effects on cAMP were the result of activity at transfected, and not endogenous, receptors.45,46 Stable CRISPR M3-knockout LX293T cells are referred to as CR-M3 cells. Cell culture medium, serum, transfection materials, culture flasks, and multiwell plates were purchased from ThermoFisher Scientific unless specified otherwise. The knockout design and procedures used to derive the CR-M3 cells from the wild-type LX293T cells, using the GeCKO CRISPR protocol (available in the public domain, https://www.addgene.org/crispr/libraries/geckov2/), were as described by Sanjana et al.47 and Shalem et al.48 Three sets of genome-specific single guide RNA sequences to mAChR M3 (F to R) were chosen from the GeCKOv2 Human Library, and both strands of oligonucleotides were synthesized for each target sequence (FA: CACCGgtcacaagcgcgcacccgagc; RA: AAACgctcgggtgcgcgcttgtgacC; FB: CACCGgcggtaccaccgatgaccctc; RB: AAACgagggtcatcggtggtaccgcC; FC: CACCGgcgctttcttaacgggcatcc; RC: AAACggatgcccgttaagaaagcgcC); the lowercase sequences are target sequences of the M3 genomic locus, and the uppercase sequences are flanking sequences for cloning. Both strands of the oligonucleotides were annealed and inserted in BsmBl restriction enzyme sites under a U6 promoter in a CRISPR/Cas9 vector (lentiCRISPR v2), a gift from Feng Zhang (Addgene plasmid #52961; Cambridge, MA, USA). The lentiCRISPR v2 plasmids containing three M3-targeting sequences were mixed and transfected using Lipofectamine LTX Reagent. The transfected cells were maintained in the presence of 5 mg/mL puromycin to select for knockout cells. Once established, stable-knockout CR-M3 cells were resuspended in high-glucose Dulbecco's modified Eagle's medium (DMEM) + 10% dimethyl sulfoxide and frozen in liquid nitrogen for future use. 
Cell Culture and Transfection Protocols
CR-M3 cells were grown in high-glucose DMEM supplemented with 10% fetal bovine serum, 1 mM sodium pyruvate, and 5 μg/mL Plasmocin (InvivoGen, San Diego, CA, USA) in a 5% CO2 environment at 37°C. Cells were maintained at <80% confluency and passaged with phosphate-buffered isotonic saline, pH 7.4, containing enzyme-free EDTA (1 mM), with mild trypsinization (90 μL 0.25% trypsin-EDTA/25 cm2 T-flask) to optimize homogeneity during replating. Transfection was performed in 12-well plates at 30% confluency. For each well to be transfected, 4 μL Lipofectamine LTX was diluted in 50 μL Opti-MEM reduced serum medium; vector cDNAs (CRE-Luc [180 ng], RLuc [160 ng], and receptor genomic clones [160 ng]) were mixed in a separate aliquot of 50 μL Opti-MEM. Equal volumes of the LTX and DNA solutions were then combined (1:1 ratio), and the cells were incubated at room temperature for 5 minutes, after which the LTX-DNA complexes were added (100 μL LTX-DNA complex/well) to wells with gentle mixing. The cells were allowed to grow in 5% CO2 at 37°C, and the cell medium was changed to supplemented DMEM without selection after a minimum exposure of 8 hours to the transfection medium. Twenty-four hours after transfection, cells were seeded (7500 cells/well) into white, clear-bottomed, tissue-treated 96-well plates, fed with supplemented DMEM without selection, and allowed to grow overnight. 
CRE-Luc Luminescence Assay and Antagonist IC50 Curves
Forty-eight hours after transfection, the cell medium was aspirated and replaced with various concentrations of antagonist diluted in FluoroBrite DMEM containing a fixed concentration of agonist (50 μL/well); treated cells were incubated for 4 hours at 37°C under 5% CO2 + O2. The fixed agonist concentration (carbachol: 10 μM; or clonidine: 1 μM) was chosen, on the basis of preliminary concentration–response studies, to produce large but submaximal activation of normalized CRE-Luc luminescence (LucN). Submaximal concentrations (78%–90% activation) were used instead of 50% or 100% activation, so as to extend the dynamic range of measurable responses. The choice of antagonists for testing was based on availability and previously published reports of their ability to prevent FDM in the chick (Table 1). All drug stock solutions (10 mM) were made using sterile distilled H2O, with the exception of QNB (5 mM) and himbacine (9 mM), which were dissolved in 100% and 90% methanol, respectively. The maximum concentration of methanol that did not cause cell death, as determined by Robust regression and Outlier removal (ROUT; GraphPad, La Jolla, CA, USA) statistical analysis, was 6%. 
Changes in intracellular cAMP were measured indirectly using the Dual-Glo Luciferase Assay System and protocol (Promega). Drug-treated cells and Dual-Glo assay reagents were equilibrated to room temperature, and then 50 μL Dual-Glo Luciferase Reagent was added to each sample well. The plate was incubated for 10 minutes at room temperature, with moderate shaking to ensure complete cell lysis. After incubation, CRE-Luc levels were measured using a Victor X4 Spectrophotometer (PerkinElmer, Waltham, MA, USA). Once CRE-Luc measurements were complete, 50 μL Dual-Glo Stop-and-Glo Reagent was added to each of the sample wells, the 10-minute incubation with shaking was repeated, and then the levels of RLuc (normalizing control) in the wells were measured in the same order, using the same spectrophotometer. 
Data Analysis
Raw RLuc data were first subjected to outlier detection using ROUT analysis with a 1% false detection rate (GraphPad Prism v 6.07; La Jolla, CA, USA). Flagged data usually represented significant decreases in RLuc expression, due to cell death caused by high-concentration drug or methanol. There were no instances of significant cell death for any of the experiments involving testing at M4 or cM4. At hADRA2A, cell death occurred at dicyclomine hydrochloride (DIC) (>500 μM), QNB (>300 μM, 6% methanol), oxyphenonium (OXY) (>3 mM), mepenzolate bromide (MEP) (>10 mM), and himbacine (HIM) (>500 μM). These data were removed along with their corresponding CRE-Luc values in the final analysis. Remaining CRE-Luc data were normalized to RLuc (CRE-Luc/RLuc = LucN), and then either the fold-change LucN (agonist treatment: [treated well − well without agonist] ÷ well without agonist) or the percent maximum response (antagonist treatment: [treated well ÷ well without antagonist] × 100) was calculated. For graphing and IC50 calculations, molar concentrations were graphed on the x-axis, transformed using the algorithm X = Log(X), and then nonlinear regression analysis was performed. Graphical data are represented as curve-fitted linear regressions of the means of the transformed molar concentration values ± SEM (GraphPad Prism, Version 6.07); n = 3 or 4 experiments, performed in duplicate. Inhibitory constants (Ki(CP)) were estimated from these data by the method of Cheng and Prusoff (pKi(CP) = IC50 ÷ (1 + ([A]/EC50)),49,50 where [A] = agonist concentration used, and EC50 was the agonist concentration required to elicit a 50% response, as determined by previous experiments using our assay system. Data are reported as either IC50 (assay/system-dependent inhibitory potency), Ki (absolute inhibitory potency), or EC50 (assay/system-dependent excitatory potency) values. These values are all concentration-dependent; therefore, the smaller the value, the more effective the ligand. 
Results
Human mAChR M4
Carbachol treatment of transiently transfected cells resulted in an increased induction of CRE-Luc–mediated luminescence (maximum 87-fold increase) with an EC50 = 5.6 μM; 10 μM was selected as the fixed agonist concentration, as it resulted in a submaximal induction of CRE-Luc (approximately 80% of maximum response). Relative IC50 values of mAChR antagonists, ranked in order from highest to lowest potency at human mAChR M4, were QNB (115 pM) > atropine (390 pM) ≥ oxyphenonium (400 pM) > mepenzolate (1 nM) > MT3 (8 nM) > himbacine (10 nM) > dicyclomine (15 nM) > pirenzepine (23 nM) > tropicamide (155 nM) (Fig. 1). Inhibitory constants (Ki), estimated from these data by the method of Cheng and Prusoff,49 were in good agreement with previously published affinity constant data for these ligands, either at human M4 receptor or in mammalian tissues (Table 1). 
Figure 1
 
Inhibitory potencies of various mAChR antagonists at the human M4 receptor. Antagonists are categorized into full myopia inhibition (open/colored symbols), partial myopia inhibition (partially filled symbols), and no myopia inhibition (filled symbols) according to previously published research,14,16,19 and ranked in descending order from most to least potent in our receptor assay (IC50 ± 95% CI). MT3 (yellow) and atropine (blue) are indicated separately as they are the most researched anti-myopia mAChR antagonist in humans (atropine), and the most potent anti-myopia drug found to date in chicks (MT3). ATR, atropine sulfate; OXY, oxyphenonium bromide; PRZ, pirenzepine dihydrochloride; CI, confidence interval.
Figure 1
 
Inhibitory potencies of various mAChR antagonists at the human M4 receptor. Antagonists are categorized into full myopia inhibition (open/colored symbols), partial myopia inhibition (partially filled symbols), and no myopia inhibition (filled symbols) according to previously published research,14,16,19 and ranked in descending order from most to least potent in our receptor assay (IC50 ± 95% CI). MT3 (yellow) and atropine (blue) are indicated separately as they are the most researched anti-myopia mAChR antagonist in humans (atropine), and the most potent anti-myopia drug found to date in chicks (MT3). ATR, atropine sulfate; OXY, oxyphenonium bromide; PRZ, pirenzepine dihydrochloride; CI, confidence interval.
Chicken mAChR M4
Carbachol treatment resulted in an increased induction of CRE-Luc–mediated luminescence (maximum 287-fold increase), similar to results reported previously,44 with an EC50 = 2 μM; 10 μM was chosen as the fixed concentration, as it gave a large but submaximal activation of CRE-Luc induction (approximately 90% of maximum response). The behavior of antagonists at the cM4 receptor did not mimic perfectly their behavior at human M4. Relative IC50 values for cM4, in order from highest to lowest potency, were QNB (310 pM) > oxyphenonium (560 pM) > atropine (710 pM) > mepenzolate (4 nM) > himbacine (6 nM) > dicyclomine (41 nM) > pirenzepine (49 nM) > tropicamide (245 nM) > MT3 (450 nM) (Fig. 2). When converted by the Cheng-Prusoff correction, the IC50 data for atropine, QNB, and pirenzepine are in good agreement with previously published Ki values obtained by utilizing the same receptor clone38,51 (Table 1). Although most antagonists acted with comparable relative inhibitory potencies at the cM4 receptor and the human M4 receptor, the Ki for MT3 in blocking cM4 was 56 times less potent (IC50 = 450 vs. 8 nM, respectively; P < 0.0001, unpaired t-test, 2-tailed). Thus, relative to all other mAChR antagonists tested, MT3 was the weakest antagonist at chick cM4
Figure 2
 
Inhibitory potencies of various mAChR antagonists at the chicken cM4 receptor. Antagonists are categorized into full myopia inhibition (open/colored symbols), partial myopia inhibition (partially filled symbols), and no myopia inhibition (filled symbols) according to previously published research,14,16,19 and ranked in descending order from most to least potent in our receptor assay (IC50 ± 95% CI). MT3 (yellow) and atropine (blue) are indicated separately as they are the most researched anti-myopia mAChR antagonist in humans (atropine), and the most potent anti-myopia drug found to date in chicks (MT3). ATR, atropine sulfate; OXY, oxyphenonium bromide; PRZ, pirenzepine dihydrochloride; CI, confidence interval.
Figure 2
 
Inhibitory potencies of various mAChR antagonists at the chicken cM4 receptor. Antagonists are categorized into full myopia inhibition (open/colored symbols), partial myopia inhibition (partially filled symbols), and no myopia inhibition (filled symbols) according to previously published research,14,16,19 and ranked in descending order from most to least potent in our receptor assay (IC50 ± 95% CI). MT3 (yellow) and atropine (blue) are indicated separately as they are the most researched anti-myopia mAChR antagonist in humans (atropine), and the most potent anti-myopia drug found to date in chicks (MT3). ATR, atropine sulfate; OXY, oxyphenonium bromide; PRZ, pirenzepine dihydrochloride; CI, confidence interval.
Human α2A-Adrenoceptor
Clonidine treatment of cells transiently transfected with hADRA2A resulted in increased CRE-Luc–mediated luminescence (maximum 26-fold increase), with an EC50 = 430 nM; 1 μM was chosen as the fixed concentration, as it gave a submaximal activation of CRE-Luc (approximately 78% of maximum response). Although increased cAMP synthesis is not the usual response to agonist treatment of Gi-coupled receptors, this Gs-coupling effect has been reported previously in CHO cells transfected with the hADRA2A receptor (also known as α2C10).43 Relative IC50 values for the mAChR antagonists at the hADRA2A receptor, ranked in order from highest to lowest potency, were MT3 (15 nM) > himbacine (17 μM) > atropine (45 μM) > oxyphenonium (463 μM) > pirenzepine (867 μM) > tropicamide (1.5 mM) > QNB (260 μM; maximum 82% inhibition) > mepenzolate (798 μM; maximum 68% inhibition) > > dicyclomine (no detectable activity) (Fig. 3). The IC50 for yohimbine, an α2-adrenergic antagonist, was 5 nM (calculated Ki(CP) = 2 nM). These data are in good agreement with previously published values for the affinity of MT3 (Ki(CP) = 5 nM)29,52 and yohimbine (Ki = 400 pM–6 nM)5355 at the hADRA2A receptor (Table 2). Thus, our data demonstrated unequivocally that myopia-inhibiting mAChR antagonists can bind to hADRA2A, although with affinities much lower than those with which they interact with mAChRs. The noteworthy exception was MT3, which bound to hADRA2A with an inhibitory potency (IC50 = 15 nM) comparable to that for its interaction with the human M4 mAChR (IC50 = 8 nM). 
Figure 3
 
Inhibitory potencies of mAChR antagonists at hADRA2A. Antagonists are categorized into full myopia inhibition (open/colored symbols), partial myopia inhibition (partially filled symbols), and no myopia inhibition (filled symbols) according to previously published research,14,16,19 and ranked in descending order from most to least potent in our receptor assay (IC50 ± 95% CI). MT3 (yellow) and atropine (blue) are indicated separately as they are the most researched anti-myopia mAChR antagonist in humans (atropine), and the most potent anti-myopia drug found to date in chicks (MT3). In this assay, dicyclomine is indicated by an open circle with a cross, because it had no activity at the hADRA2A receptor at the concentrations tested. YOH, yohimbine hydrochloride; ATR, atropine sulfate; OXY, oxyphenonium bromide; PRZ, pirenzepine dihydrochloride; CI, confidence interval.
Figure 3
 
Inhibitory potencies of mAChR antagonists at hADRA2A. Antagonists are categorized into full myopia inhibition (open/colored symbols), partial myopia inhibition (partially filled symbols), and no myopia inhibition (filled symbols) according to previously published research,14,16,19 and ranked in descending order from most to least potent in our receptor assay (IC50 ± 95% CI). MT3 (yellow) and atropine (blue) are indicated separately as they are the most researched anti-myopia mAChR antagonist in humans (atropine), and the most potent anti-myopia drug found to date in chicks (MT3). In this assay, dicyclomine is indicated by an open circle with a cross, because it had no activity at the hADRA2A receptor at the concentrations tested. YOH, yohimbine hydrochloride; ATR, atropine sulfate; OXY, oxyphenonium bromide; PRZ, pirenzepine dihydrochloride; CI, confidence interval.
Table 2
 
Binding Data for α2-Adrenergic Drugs and mAChR Antagonists at hADRA2A
Table 2
 
Binding Data for α2-Adrenergic Drugs and mAChR Antagonists at hADRA2A
Controls
Three different control experiments were conducted to verify that the changes in CRE-Luc, reflecting increases in intracellular cAMP, were due to activation of the transfected receptor and not stimulation of endogenous receptor signaling pathways. First, cells were transfected with the pcDNA 3.1(+) plasmid only. There was no significant change in luminescence in response to carbachol or clonidine treatment (Supplementary Fig. S1). Second, the impacts of elevating intracellular calcium using a calcium ionophore (CI A23187), or activating protein kinase C with phorbol 12-myristate 13-acetate (PMA) were tested. CI A23187 treatment, resulting in an increase in intracellular Ca2+, could in principle cause activation of Ca2+-dependent cyclase and an increase in cellular cAMP, thereby mimicking the action of Gq-coupled GPCRs (Supplementary Fig. S2). Alternatively, the protein kinase C (PKC) activator, PMA, could mimic the potential action of Gq-protein–coupled receptors via phospholipase action, and raise both inositol triphosphate and diacylglycerol levels by activating cAMP response element binding protein (CREB)—thus causing increased luminescence without an increase in cellular cAMP (Supplementary Fig. S2). Importantly, we observed no significant change in LucN with treatment of either CI A23187 (1 μM) or PMA (10 μM), especially when compared to the response resulting from agonist treatment of transfected cells (Supplementary Fig. S2). 
Discussion
The primary finding of our study is that the relative potencies of myopia-inhibiting mAChR antagonists to block the adrenergic hADRA2A receptor are correlated better with their ability to inhibit chick FDM than are the relative inhibitory potencies of the same compounds to block the muscarinic M4/cM4 receptors (Tables 1, 2; Fig. 4). The data correlating the relative abilities of the mAChR antagonists for myopia inhibition versus their relative Ki values for blocking the chick and human mAChRs show MT3 as an obvious outlier (Figs. 4a, 4b, yellow circles). Yet, the same correlation plot paradigm shows that the relative Ki of MT3 to block hADRA2A is tightly correlated, as are the Ki values of the other mAChR antagonists, with the ability to attenuate chick FDM (Fig. 4c, yellow circle). These findings support the hypothesis that myopia inhibition by high-concentration mAChR antagonists may be mediated by receptors other than mAChR M4/cM4, and our data point to members of the α-adrenoceptor family as possible alternatives. A secondary finding is that, although the relative inhibitory potencies of most myopia-inhibiting mAChR antagonists for blocking the chick cM4 compared with the human M4 receptors are in good agreement, MT3 is an anomaly; it has markedly reduced Ki for the chick cM4 receptor compared with the human M4 receptor. This result is likely due to differences in the ligand-binding sites on the two receptors. 
Figure 4
 
Correlations between rank order inhibitory potency (x-axis) and rank order inhibition of axial elongation in chicks with experimentally induced FDM (y-axis) for mAChR antagonists at M4 (a), cM4 (b), and hADRA2A (c). Rank orders for both parameters are 1 = least potent (pIC50) or effective (% axial length inhibition) and 9 = most potent (pIC50) or effective (% axial length inhibition). A line of unity (slope = 1) has been superimposed upon the graphs for visual reference. Inhibitory potencies of mAChR antagonists, and their abilities to inhibit FDM in the chick, correlate significantly for hADRA2A only (r = 0.87, P < 0.01; Spearman's ranked correlation). Atropine and MT3 are shown in color (atropine = blue, MT3 = yellow). Full inhibition of axial elongation (open/colored symbols), partial inhibition of axial elongation (partially filled symbols), and inhibition of axial elongation (filled symbols) according to previously published research in the chick.14,16,19
Figure 4
 
Correlations between rank order inhibitory potency (x-axis) and rank order inhibition of axial elongation in chicks with experimentally induced FDM (y-axis) for mAChR antagonists at M4 (a), cM4 (b), and hADRA2A (c). Rank orders for both parameters are 1 = least potent (pIC50) or effective (% axial length inhibition) and 9 = most potent (pIC50) or effective (% axial length inhibition). A line of unity (slope = 1) has been superimposed upon the graphs for visual reference. Inhibitory potencies of mAChR antagonists, and their abilities to inhibit FDM in the chick, correlate significantly for hADRA2A only (r = 0.87, P < 0.01; Spearman's ranked correlation). Atropine and MT3 are shown in color (atropine = blue, MT3 = yellow). Full inhibition of axial elongation (open/colored symbols), partial inhibition of axial elongation (partially filled symbols), and inhibition of axial elongation (filled symbols) according to previously published research in the chick.14,16,19
Distribution of Drugs Within Ocular Tissues After Different Delivery Methods
A question to ask about the use of atropine for myopia inhibition (in humans and animals) is, Does the high concentration of antagonist, as administered, reflect its concentration at the site of action in the eye? Currently, the site of action for atropine-mediated myopia inhibition is unknown, but best guesses identify the retina, choroid, and/or sclera.56 In chicks, intravitreal injection is the usual method of delivery, and concentrations of 1 to 100 mM have been used for mAChR antagonist-mediated inhibition of FDM16,57,58 (Table 1). Unfortunately, no studies exist on the ocular distribution of atropine after intravitreal injection into chick eyes; but one study has investigated the ocular distribution, in chicks, of a single dose of intravitreally injected radioactively labeled pirenzepine.59 At 1 hour after injection, reported concentrations (percentage of the original injected amount) were 6% in the retina, 1.5% in the choroid, and 2.5% in the sclera.59 Based on these data, and considering that 100 mM was used to achieve full myopia inhibition in the chick,16 estimated concentrations for ocular tissues would be retina = 6 mM, choroid = 1.5 mM, and sclera = 2.5 mM. These estimated concentrations greatly exceed those that would be expected to have mAChR-only effects, and fall within the range of concentrations that would enable pirenzepine to block the hADRA2A receptor under in vitro conditions, as calculated by our assay (Ki(CP) = 250 μM; Table 2). 
In humans, atropine for myopia inhibition is delivered in the form of daily eye drops (0.01%–1%). Under experimental conditions, delivery of a single dose of 2% [3H]-atropine to the conjunctival sac of albino rabbits resulted in a distribution of the original concentration of 0.09% in the sclera, 0.05% in the choroid, and 0.008% in the retina, 1 hour after instillation60; similar results (retina: 0.01%; choroid and sclera 0.38%) were seen 30 minutes after topical application of [C14]-atropine.61 Therefore, the concentrations of atropine in the eye after a single topical application of 0.01% to 1% (0.13–13 μM in the sclera, 0.07–7 μM in the choroid, and 0.01–1.1 μM in the retina, extrapolated from single-dose studies in albino rabbit60,61) are within the range of what we have found to be the IC50 for atropine's ability to block the hADRA2A in in vitro cell culture preparations (our value = 45 μM; value reported by others = 1–10 μM).30,31 The ocular distribution of atropine after multiple topical doses has not been determined, but studies monitoring the accumulation of atropine in humans have shown that atropine buildup can occur. Serum levels of atropine were undetectable after a single inhalation dose of 12 to 25 ng/μL atropine sulfate, but were raised to 1.13 to 5.23 ng/μL when treatment was administered every 4 to 6 hours over a period of 48 hours.62 Pigmented tissues (RPE, iris, ciliary body, choroid, and retina) are especially apt at retaining atropine, which may lead to an initially weaker but significantly prolonged effect.63,64 It is possible, then, that topical application of 0.01% atropine daily for long periods (up to 2 years) could result in ocular concentrations much higher than those seen after a single dose, with intracellular organelles such as melanin granules acting as reservoirs for slow release of the drug and possibly providing local concentrations that would be sufficient to interact with non-mAChRs. 
Pirenzepine has also been tested for the treatment of childhood myopia, with twice-daily applications of concentrations up to 2%; but the results did not support superiority of clinical efficacy of pirenzepine treatment over atropine treatment,6568 and further studies have seemingly been abandoned.69 Ocular distribution of a single dose of topically applied [3H]-pirenzepine (2%) has also been tested in the rabbit eye; concentrations in the ocular tissues 1 hour after administration were roughly half those calculated for atropine (retina: 0.004%, choroid: 0.021%, sclera: 0.052%)—possibly because atropine, being more lipophilic, is better at penetrating the ocular tissues and is lost less rapidly than pirenzepine.60 The estimated concentrations of pirenzepine delivered to ocular tissues should be sufficient to activate all mammalian mAChR subtypes fully,70 but are expected to fall below the concentration required to achieve a significant blockade of hADRA2A signaling (retina: 2 μM, choroid: 12 μM, sclera: 30 μM) as calculated by our assay (Ki(cp) = 250 μM, Table 2). 
mAChR Antagonists at M4, cM4, and hADRA2A Receptors: Correlations With Myopia Inhibition
The relative inhibitory potencies of myopia-inhibiting mAChR antagonists at the hADRA2A receptor, but not at the chick cM4 and human M4 mAChRs, are well correlated with their reported ability to inhibit chick FDM14,16,19 (Fig. 4). We have confirmed previous reports of high-affinity binding of MT329,52 (Ki(CP) = 5 nM) to hADRA2A, and demonstrated that atropine and himbacine can also bind to this receptor, albeit with potencies much lower than their relative Ki values for blocking the mAChRs. All other mAChR antagonists tested had very low potency for blocking hADRA2A, but 100% inhibition of clonidine-mediated effects was achieved with oxyphenonium, tropicamide, and pirenzepine—drugs that were reported to inhibit myopia fully in the chick when used at high concentrations. The maximum inhibitory effects achieved by QNB (partial myopia inhibition in chick) and mepenzolate (no myopia inhibition in chick) were 82% and 68%, respectively, and dicyclomine (no myopia inhibition in chick) had no discernable effect at the concentrations tested. At the human and chick M4/cM4 receptors, atropine, QNB, mepenzolate, and dicyclomine are all comparably potent inhibitors, with Ki values in the nanomolar range; yet atropine is the only one of these that can inhibit chick FDM.16 Most interesting is the behavior of MT3 at M4/cM4. It is the most effective drug found to date for mitigating chick FDM,14,15 and we confirmed its high potency for the human M4 receptor. However, we found it to be the least potent of all drugs tested at the chick cM4 receptor. This disconnect, between the potency of MT3 at cM4 and its effectiveness against chick FDM, supports our hypothesis that myopia inhibition is not mediated by the M4/cM4 receptor. 
Difference in Binding of MT3 at M4 Versus cM4
When attempting to assign functional consequences to treatment with a particular ligand, it is important to consider the mechanism of binding of that ligand to its target receptor. Generally, orthosteric binding sites, where the transmitters or primary signaling molecules bind, are highly conserved (100%) between receptors of the same class and between species.71 “Selective” modulatory ligands such as MT3, however, will bind to receptor amino acid residues that are less conserved, usually outside the orthosteric pocket, in the extracellular loops (EL2 and EL3) of the target GPCR.39 In such instances, substitution of even a single amino acid can have a significant impact on ligand–receptor binding characteristics.39,40 Thus, while decreased conservation of receptor sequences in the extracellular loops aids in selectivity of a ligand for a certain receptor subtype, the increasingly stringent requirements for binding can also result in loss of activity at the same receptor subtype in other species. This finding is what we observed in our assay: MT3 bound poorly to cM4 compared to M4. Chicken and human M4 share only a 71.4% to 72.7% sequence identity in EL3 and EL2, respectively (Supplementary Fig. S3). Therefore, it is likely that the decreased activity of MT3 at cM4 is caused by amino acid differences in its EL2 and EL3 sequences, compared to those of M4. All other drugs tested in our assay did not differ significantly in their binding behavior at M4 versus cM4. This result was expected; these ligands are known to be orthosteric inhibitors of ACh at mAChRs, and the reported orthosteric binding site of the mAChR M4/cM4 receptors is 100% conserved between human and chick71 (Supplementary Fig. S3). 
Ocular α-Adrenoceptors and Possible Mechanism of Action
Sympathetic innervation in the vertebrate eye services structures such as the dilator pupillae, the ciliary muscle and epithelium, and ocular blood vessels within and outside the choroid.72 Alpha2-adrenoceptors are best known as autoreceptors, but they can also act as heteroreceptors.41,73 Alpha2-adrenoceptors that act as autoreceptors are located on presynaptic adrenergic neurons; they are activated by epinephrine/norepinephrine and inhibit the release of their own neurotransmitters. Alpha2-adrenoceptors that act as heteroreceptors are located presynaptically on nonadrenergic neurons, and there is evidence that α2-adrenoceptors can modulate the release of neurotransmitters such as GABA, dopamine, and serotonin in the rat brain and retina.7479 Interestingly, both GABA and dopamine have been implicated in the regulation of eye growth.8088 In the rat retina, the epinephrine-synthesizing enzyme phenylethanolamine N-methyltransferase (PNMT) has been localized to amacrine cell populations89 and retinal epinephrine levels have been reported to rise upon light exposure90,91; but there is no further evidence for a role of epinephrine or norepinephrine in retinal visual processing of any vertebrate species examined. In spite of this, α-adrenoceptors are present on retinal neurons. In rat retina, human, and primate tissue, ADRA2A immunoreactivity has been localized to the nonpigmented ciliary epithelium, cornea and conjunctiva, and retinal ganglion cells and amacrine cells.92 In a transgenic mouse expressing an N-terminal hemagglutinin epitope (HA)-tagged α2A-receptor, driven by the endogenous ADRA2A gene locus, ADRA2A gene expression was localized to horizontal cells, inner nuclear layer, inner plexiform layer, and the retinal ganglion cell layer (Rao SS, et al. IOVS 2011;52:ARVO E-Abstract 2059). In chick, immunoreactive ADRA2A has been localized to retinal Müller cells, amacrine cells, and ganglion cells (Costa GV, et al. IOVS 2012;53:ARVO E-Abstract 6545).93 
Ocular α2-adrenoceptors are implicated in the pathogenesis of glaucoma, and α2-adrenoceptor agonists, such as brimonidine and apraclonidine, are clinically approved for management of intraocular pressure (IOP). The prevalence of glaucoma is said to be correlated with the prevalence of myopia,94,95 and new evidence suggests that these α2-adrenoceptor agonists can inhibit myopia in chicks (Carr BJ, Stell WK. IOVS 2016;57:ARVO E-Abstract 4738) and guinea pigs.43 It is still unknown which neural circuits or receptors might mediate these effects, but Liu et al.43 suggested that attenuation of IOP is responsible for inhibition of myopia in guinea pigs. A direct mechanical influence of IOP (within the physiological range) on axial elongation, as hypothesized by Pruett96 and others,9799 does not seem likely, however. Human clinical studies have found no relationship,100103 an increase in IOP after but not preceding the onset of myopia,99,104 or a small decrease in IOP before onset of childhood myopia103; animal studies have produced equally equivocal results.97,105 A possible reason for this could be measurement bias induced by utilizing indentation or applanation tonometry, which calculates IOP from the amount of pressure required to flatten an area of the cornea. The accuracy of tonometry is dependent on factors such as corneal thickness106 and scleral compliance,107,108 and increased scleral compliance (scleral creep) has been observed in humans with pathologic myopia109 and mammals with experimentally induced myopia.110,111 Thus, if the sclera is more compliant than normal, it may lead to a mistaken measurement bias toward lower IOP. In contrast, α2-adrenoceptor agonists also were reported to effectively inhibit FDM in chicks (Carr BJ, Stell WK. IOVS 2016;57:ARVO E-Abstract 4738), in which there is no increase in scleral compliance in response to myopiagenic stimuli111; IOP measurements were not recorded in that study, however, so we cannot exclude a role completely. The relationship between the actions and receptor targets of the compounds we studied and their impact on IOP and myopia remains an interesting topic for future work. Notwithstanding the possible role of IOP in myopia, our work establishes clearly that action of atropine via the mAChR M4 receptor is highly unlikely to be a factor. 
Alpha2-adrenoceptor agonism in the retina can also result in the upregulation of neuroprotective agents and mechanisms, such as an increase in basic fibroblast growth factor (bFGF) in rat photoreceptors112,113 or activation of the extracellular signal-regulated kinase signaling pathway in chick Müller cells.93 Interestingly, both bFGF and extracellular signal-regulated kinase signaling have been implicated in protection against chick FDM114,115 and guinea pig lens-induced myopia.42,116,117 Finally, brimonidine might be involved in the regulation of nitric oxide synthase in retinal arterioles, where it may activate endothelial nitric oxide synthase (eNOS), resulting in vasodilation, as reported in pigs.118 It is important to note that although there are no intraretinal blood vessels in the chick retina, it is served by blood vessels in the choroid and an intravitreal structure called the pecten. Whether α2-adrenoceptor ligands can also modulate NOS (neuronal NOS, eNOS, or inducible NOS) activity or expression in retinal neurons is worth investigating further, since synthesis and release of nitric oxide are necessary for myopia inhibition by atropine, and have been associated strongly with reduction of eye growth.57 
Caveats and Considerations
There are some discrepancies in the ability of mamba toxins to inhibit myopia in different animal models. Although MT3 inhibits FDM effectively in chicks14 and tree shrews,17 it seems to be ineffective at inhibiting the axial elongation induced by lens-induced myopia (LIM) in the same animals.15,17 Conversely, MT7, a mamba toxin subtype that is highly selective for mAChR M1 and is not known to act at any other receptor,28,29 inhibits LIM, but not the axial elongation induced by FDM, in the tree shrew.17 MT7 has no effect in the chick,119 which is expected because chicks do not possess the M1 subtype. Pirenzepine, however, is effective at inhibiting chick FDM, possibly due to binding to the cM2 receptor, which MT7 does not recognize, or to off-target binding at α2-adrenoceptors as evidenced by our work. These differences in treatment effectiveness of mamba toxins are in line with evidence suggesting that the circuits involved in LIM and FDM are in some ways different,120122 and that—whatever the mechanism affected by atropine (and other myopia-inhibiting mAChR antagonists) is—the results of our study may be unique to the circuitry of FDM, and not LIM. 
Although we have shown that some mAChR antagonists will bind hADRA2A at high concentrations—comparable to those found to be effective against chick FDM—we have not ruled out other α-adrenoceptor subtypes, non-M4 muscarinic subtypes (i.e., mAChR M1/cM2), or non-mAChR/nonadrenergic receptor targets. The high concentrations of these drugs required to inhibit FDM in the chick model impede our ability to identify any single receptor as the key target for the impact of any compound on myopia, because of the distinct possibility of many unidentified off-target receptor-mediated effects.21 Furthermore, because of technical issues we could not overcome, we were unable to express a functional chick ADRA2A receptor in our HEK cell expression system, which would have enabled us to compare the “nonspecific” affinities of mAChR antagonists directly for the chick versus human α2A-adrenoceptors. What can be concluded with confidence from our data, however, is that M4/cM4 cannot be seen as the unique or even the leading receptor candidate for mediating the inhibition of FDM by atropine (and other mAChR antagonists) in the chick bioassay. 
Summary
Our data provide new information about the relative potencies of muscarinic antagonists at the human and chick mAChR M4/cM4 receptors and the human α2A-adrenoceptor. This is the first study to characterize the inhibitory action of oxyphenonium, mepenzolate, MT3, dicyclomine, and tropicamide explicitly at chicken cM4, and the first to test specifically and report in detail, the potential “off-target” actions of multiple mAChR antagonists at hADRA2A. Consequently, these data provide a concentration range over which these drugs should be expected to act in a mAChR-specific manner, and a concentration range at which they can be predicted to have “off-target” effects at hADRA2A and possibly other receptors. The focus of thinking about the anti-myopia action of atropine as muscarinic, combined with the lack of knowledge of the mechanism of atropine's therapeutic actions (in the setting of either human or chick myopia), is a deterrent to the discovery of more effective anti-myopia agents with fewer side effects. The data presented here make a compelling case for continued investigation into non-mAChR targets for atropine-mediated myopia inhibition, with α-adrenoceptors being provocative candidates for novel therapeutic interventions. 
Acknowledgments
Supported by a Natural Sciences and Engineering Research Council of Canada (NSERC) Discovery Grant (RGPIN/131-2013) and the Foundation Fighting Blindness Canada EYEGEYE Research Training Fund (WKS), a Canadian Institutes of Health Research (CIHR) grant (MOP13759) (MDH), and an NSERC PGS-D award (PGSD2-476058-2015), Dr. D. Grant Gall Graduate Traineeship, Queen Elizabeth II Scholarships, and Odd Fellow Rebekah Visual Research Fund Awards (BJC). Core support was provided by the University of Calgary Core DNA Services and the Live Cell Imaging Facility, funded by the Snyder Institute for Chronic Disease, at the Cumming School of Medicine, University of Calgary. 
Disclosure: B.J. Carr, None; K. Mihara, None; R. Ramachandran, None; M. Saifeddine, None; N.M. Nathanson, None; W.K. Stell, None; M.D. Hollenberg, None 
References
Foster PJ, Jiang Y. Epidemiology of myopia. Eye (Lond). 2014; 28: 202–208.
Ikuno Y. Overview of the complications of high myopia. Retina. 2017; 37: 2347–2351.
Dolgin E. The myopia boom. Nature. 2015; 519: 276–278.
Chia A, Chua WH, Cheung YB, et al. Atropine for the treatment of childhood myopia: safety and efficacy of 0.5%, 0.1%, and 0.01% doses (atropine for the treatment of myopia 2). Ophthalmology. 2012; 119: 347–354.
Chua WH, Balakrishnan V, Chan YH, et al. Atropine for the treatment of childhood myopia. Ophthalmology. 2006; 113: 2285–2291.
Tong L, Huang XL, Koh AL, Zhang X, Tan DT, Chua WH. Atropine for the treatment of childhood myopia: effect on myopia progression after cessation of atropine. Ophthalmology. 2009; 116: 572–579.
Chia A, Lu QS, Tan D. Five-year clinical trial on atropine for the treatment of myopia 2: myopia control with atropine 0.01% eyedrops. Ophthalmology. 2016; 123: 391–399.
Clark TY, Clark RA. Atropine 0.01% eyedrops significantly reduce the progression of childhood myopia. J Ocul Pharmacol Ther. 2015; 31: 541–545.
Huang J, Wen D, Wang Q, et al. Efficacy comparison of 16 interventions for myopia control in children: a network meta-analysis. Ophthalmology. 2016; 123: 697–708.
Morgan IG, He M. An important step forward in myopia prevention: low-dose atropine. Ophthalmology. 2016; 123: 232–233.
Galvis V, Tello A, Parra MM, et al. Topical atropine in the control of myopia. Med Hypothesis Discov Innov Ophthalmol. 2016; 5: 78–88.
Pineles SL, Kraker RT, VanderVeen DK, et al. Atropine for the prevention of myopia progression in children: a report by the American Academy of Ophthalmology. Ophthalmology. 2017; 124: 1857–1866.
Liang JS, Carsi-Gabrenas J, Krajewski JL, et al. Anti-muscarinic toxins from Dendroaspis angusticeps. Toxicon. 1996; 34: 1257–1267.
McBrien NA, Arumugam B, Gentle A, Chow A, Sahebjada S. The M4 muscarinic antagonist MT-3 inhibits myopia in chick: evidence for site of action. Ophthalmic Physiol Opt. 2011; 31: 529–539.
Nickla DL, Yusupova Y, Totonelly K. The muscarinic antagonist MT3 distinguishes between form deprivation- and negative lens-induced myopia in chicks. Curr Eye Res. 2015; 40: 962–967.
Luft WA, Ming Y, Stell WK. Variable effects of previously untested muscarinic receptor antagonists on experimental myopia. Invest Ophthalmol Vis Sci. 2003; 44: 1330–1338.
Arumugam B, McBrien NA. Muscarinic antagonist control of myopia: evidence for M4 and M1 receptor-based pathways in the inhibition of experimentally-induced axial myopia in the tree shrew. Invest Ophthalmol Vis Sci. 2012; 53: 5827–5837.
McBrien NA, Stell WK, Carr B. How does atropine exert its anti-myopia effects? Ophthalmic Physiol Opt. 2013; 33: 373–378.
Cottriall CL, Truong HT, McBrien NA. Inhibition of myopia development in chicks using himbacine: a role for M4 receptors? Neuroreport. 2001; 12: 2453–2456.
Fischer AJ, Miethke P, Morgan IG, Stell WK. Cholinergic amacrine cells are not required for the progression and atropine-mediated suppression of form-deprivation myopia. Brain Res. 1998; 794: 48–60.
Schwahn HN, Kaymak H, Schaeffel F. Effects of atropine on refractive development, dopamine release, and slow retinal potentials in the chick. Vis Neurosci. 2000; 17: 165–176.
McBrien NA, Jobling AI, Truong HT, Cottriall CL, Gentle A. Expression of muscarinic receptor subtypes in tree shrew ocular tissues and their regulation during the development of myopia. Mol Vis. 2009; 15: 464–475.
Vessey KA, Cottriall CL, McBrien NA. Muscarinic receptor protein expression in the ocular tissues of the chick during normal and myopic eye development. Brain Res Dev Brain Res. 2002; 135: 79–86.
McBrien NA, Cottriall CL, Annies R. Retinal acetylcholine content in normal and myopic eyes: a role in ocular growth control? Vis Neurosci. 2001; 18: 571–580.
Lind GJ, Chew SJ, Marzani D, Wallman J. Muscarinic acetylcholine receptor antagonists inhibit chick scleral chondrocytes. Invest Ophthalmol Vis Sci. 1998; 39: 2217–2231.
Barathi VA, Weon SR, Beuerman RW. Expression of muscarinic receptors in human and mouse sclera and their role in the regulation of scleral fibroblasts proliferation. Mol Vis. 2009; 15: 1277–1293.
Nickla DL, Zhu X, Wallman J. Effects of muscarinic agents on chick choroids in intact eyes and eyecups: evidence for a muscarinic mechanism in choroidal thinning. Ophthalmic Physiol Opt. 2013; 33: 245–256.
Potter LT. Snake toxins that bind specifically to individual subtypes of muscarinic receptors. Life Sci. 2001; 68: 2541–2547.
Näreoja K, Kukkonen JP, Rondinelli S, Toivola DM, Meriluoto J, Nasman J. Adrenoceptor activity of muscarinic toxins identified from mamba venoms. Br J Pharmacol. 2011; 164: 538–550.
Chang KC, Hahn KH. Is α-adrenoceptor blockade responsible for atropine flush? Eur J Pharmacol. 1995; 284: 331–334.
Nedergaard OA, Schrold J. Presynaptic muscarinic and alpha-adrenergic receptor blocking effect of atropine on the noradrenergic neurones of the rabbit pulmonary artery. Blood Vessels. 1979; 16: 163–168.
Troilo D, Gottlieb MD, Wallman J. Visual deprivation causes myopia in chicks with optic nerve section. Curr Eye Res. 1987; 6: 993–999.
Wallman J, Turkel J, Trachtman J. Extreme myopia produced by modest change in early visual experience. Science. 1978; 201: 1249–1251.
Wallman J, Gottlieb MD, Rajaram V, Fugate-Wentzek LA. Local retinal regions control local eye growth and myopia. Science. 1987; 237: 73–77.
Schaeffel F, Glasser A, Howland HC. Accommodation, refractive error and eye growth in chickens. Vision Res. 1988; 28: 639–657.
Wilson KT, Sivak JG, Callender MG. Induced refractive anomalies affect chick orbital bone structure. Exp Eye Res. 1997; 64: 675–682.
Schaeffel F, Feldkaemper M. Animal models in myopia research. Clin Exp Optom. 2015; 98: 507–517.
Tietje KM, Nathanson NM. Embryonic chick heart expresses multiple muscarinic acetylcholine receptor subtypes. Isolation and characterization of a gene encoding a novel m2 muscarinic acetylcholine receptor with high affinity for pirenzepine. J Biol Chem. 1991; 266: 17382–17387.
Kukkonen A, Peräkylä M, Akerman KE, Näsman J. Muscarinic toxin 7 selectivity is dictated by extracellular receptor loops. J Biol Chem. 2004; 279: 50923–50929.
Radić Z, Duran R, Vellom DC, Li Y, Cervenansky C, Taylor P. Site of fasciculin interaction with acetylcholinesterase. J Biol Chem. 1994; 269: 11233–11239.
Starke K. Presynaptic autoreceptors in the third decade: focus on α2-adrenoceptors. J Neurochem. 2001; 78: 685–693.
Liu Y, Wang Y, Lv H, Jiang X, Zhang M, Li X. α-adrenergic agonist brimonidine control of experimentally induced myopia in guinea pigs: a pilot study. Mol Vis. 2017; 23: 785–798.
Eason MG, Kurose H, Holt BD, Raymond JR, Liggett SB. Simultaneous coupling of alpha 2-adrenergic receptors to two G-proteins with opposing effects. Subtype-selective coupling of α2C10, α2C4, and α2C2 adrenergic receptors to Gi and Gs. J Biol Chem. 1992; 267: 15795–15801.
Migeon JC, Nathanson NM. Differential regulation of cAMP-mediated gene transcription by m1 and m4 muscarinic acetylcholine receptors. Preferential coupling of m4 receptors to Gi α2. J Biol Chem. 1994; 269: 9767–9773.
Hakak Y, Shrestha D, Goegel MC, Behan DP, Chalmers DT. Global analysis of G-protein-coupled receptor signaling in human tissues. FEBS Lett. 2003; 550: 11–17.
Luo J, Busillo JM, Benovic JL. M3 muscarinic acetylcholine receptor-mediated signaling is regulated by distinct mechanisms. Mol Pharmacol. 2008; 74: 338–347.
Sanjana NE, Shalem O, Zhang F. Improved vectors and genome-wide libraries for CRISPR screening. Nat Methods. 2014; 11: 783–784.
Shalem O, Sanjana NE, Hartenian E, et al. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science. 2014; 343: 84–87.
Cheng Y, Prusoff WH. Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction. Biochem Pharmacol. 1973; 22: 3099–3108.
Munson PJ, Rodbard D. An exact correction to the “Cheng-Prusoff” correction. J Recept Res. 1988; 8: 533–546.
Tietje KM, Goldman PS, Nathanson NM. Cloning and functional analysis of a gene encoding a novel muscarinic acetylcholine receptor expressed in chick heart and brain. J Biol Chem. 1990; 265: 2828–2834.
Blanchet G, Collet G, Mourier G, et al. Polypharmacology profiles and phylogenetic analysis of three-finger toxins from mamba venom: case of aminergic toxins. Biochimie. 2014; 103: 109–117.
Laurila JM, Xhaard H, Ruuskanen JO, et al. The second extracellular loop of α2A-adrenoceptors contributes to the binding of yohimbine analogues. Br J Pharmacol. 2007; 151: 1293–1304.
Ruuskanen JO, Laurila J, Xhaard H, et al. Conserved structural, pharmacological and functional properties among the three human and five zebrafish α2A-adrenoceptors. Br J Pharmacol. 2005; 144: 165–177.
Zheng W, Lei L, Lalchandani S, Sun G, Feller DR, Miller DD. Yohimbine dimers exhibiting binding selectivities for human α2a- versus α2b-adrenergic receptors. Bioorg Med Chem Lett. 2000; 10: 627–630.
Wallman J, Winawer J. Homeostasis of eye growth and the question of myopia. Neuron. 2004; 43: 447–468.
Carr BJ, Stell WK. Nitric oxide (NO) mediates the inhibition of form-deprivation myopia by atropine in chicks. Sci Rep. 2016; 6: 9.
Schmid KL, Wildsoet CF. Inhibitory effects of apomorphine and atropine and their combination on myopia in chicks. Optom Vis Sci. 2004; 81: 137–147.
Cottriall CL, McBrien NA, Annies R, Leech EM. Prevention of form-deprivation myopia with pirenzepine: a study of drug delivery and distribution. Ophthalmic Physiol Opt. 1999; 19: 327–335.
Tigges M, Iuvone PM, Fernandes A, et al. Effects of muscarinic cholinergic receptor antagonists on postnatal eye growth of rhesus monkeys. Optom Vis Sci. 1999; 76: 397–407.
Janes RG, Stiles JF. The penetration of C14-labeled atropine into the eye; a comparison of methods of application. AMA Arch Ophthalmol. 1959; 62: 69–74.
Kradjan WA, Smallridge RC, Davis R, Verma P. Atropine serum concentrations after multiple inhaled doses of atropine sulfate. Clin Pharmacol Ther. 1985; 38: 12–15.
Salazar M, Patil PN. An explanation for the long duration of mydriatic effect of atropine in eye. Invest Ophthalmol. 1976; 15: 671–673.
Salazar M, Shimada K, Patil PN. Iris pigmentation and atropine mydriasis. J Pharmacol Exp Ther. 1976; 197: 79–88.
Bartlett JD, Niemann K, Houde B, Allred T, Edmondson MJ, Crockett RS. A tolerability study of pirenzepine ophthalmic gel in myopic children. J Ocul Pharmacol Ther. 2003; 19: 271–279.
Siatkowski RM, Cotter S, Miller JM, Scher CA, Crockett RS, Novack GD. Safety and efficacy of 2% pirenzepine ophthalmic gel in children with myopia: a 1-year, multicenter, double-masked, placebo-controlled parallel study. Arch Ophthalmol. 2004; 122: 1667–1674.
Siatkowski RM, Cotter SA, Crockett RS, Miller JM, Novack GD, Zadnik K. Two-year multicenter, randomized, double-masked, placebo-controlled, parallel safety and efficacy study of 2% pirenzepine ophthalmic gel in children with myopia. J AAPOS. 2008; 12: 332–339.
Tan DT, Lam DS, Chua WH, Shu-Ping DF, Crockett RS. One-year multicenter, double-masked, placebo-controlled, parallel safety and efficacy study of 2% pirenzepine ophthalmic gel in children with myopia. Ophthalmology. 2005; 112: 84–91.
Kang P. Optical and pharmacological strategies of myopia control. Clin Exp Optom. 2018; 101: 321–332.
Moriya H, Takagi Y, Nakanishi T, Hayashi M, Tani T, Hirotsu I. Affinity profiles of various muscarinic antagonists for cloned human muscarinic acetylcholine receptor (mAChR) subtypes and mAChRs in rat heart and submandibular gland. Life Sci. 1999; 64: 2351–2358.
Thal DM, Sun B, Feng D, et al. Crystal structures of the M1 and M4 muscarinic acetylcholine receptors. Nature. 2016; 531: 335–340.
McDougal DH, Gamlin PD. Autonomic control of the eye. Compr Physiol. 2015; 5: 439–473.
Gilsbach R, Hein L. Are the pharmacology and physiology of α2 adrenoceptors determined by α2-heteroreceptors and autoreceptors respectively? Br J Pharmacol. 2012; 165: 90–102.
Mongeau R, Weiss M, de Montigny C, Blier P. Effect of acute, short- and long-term milnacipran administration on rat locus coeruleus noradrenergic and dorsal raphe serotonergic neurons. Neuropharmacology. 1998; 37: 905–918.
Jackisch R, Haaf A, Jeltsch H, Lazarus C, Kelche C, Cassel JC. Modulation of 5-hydroxytryptamine release in hippocampal slices of rats: effects of fimbria-fornix lesions on 5HT1B-autoreceptor and α2-heteroreceptor function. Brain Res Bull. 1999; 48: 49–59.
Forray MI, Bustos G, Gysling K. Noradrenaline inhibits glutamate release in the rat bed nucleus of the stria terminalis: in vivo microdialysis studies. J Neurosci Res. 1999; 55: 311–320.
Zha DJ, Wang ZM, Lin Y, et al. Effects of noradrenaline on the GABA response in rat isolated spiral ganglion neurons in culture. J Neurochem. 2007; 103: 57–66.
Philbin KE, Bateman RJ, Mendelowitz D. Clonidine, an α2-receptor agonist, diminishes GABAergic neurotransmission to cardiac vagal neurons in the nucleus ambiguus. Brain Res. 2010; 1347: 65–70.
Iuvone PM, Rauch AL. Alpha2-adrenergic receptors influence tyrosine hydroxylase activity in retinal dopamine neurons. Life Sci. 1983; 33: 2455–2463.
Schmid KL, Strasberg G, Rayner CL, Hartfield PJ. The effects and interactions of GABAergic and dopaminergic agents in the prevention of form deprivation myopia by brief periods of normal vision. Exp Eye Res. 2013; 110: 88–95.
Stone RA, Liu J, Sugimoto R, Capehart C, Zhu X, Pendrak K. GABA, experimental myopia, and ocular growth in chick. Invest Ophthalmol Vis Sci. 2003; 44: 3933–3946.
Cheng ZY, Wang XP, Schmid KL, Han XG. Inhibition of form-deprivation myopia by a GABAAOr receptor antagonist, (1,2,5,6-tetrahydropyridin-4-yl) methylphosphinic acid (TPMPA), in guinea pigs. Graefes Arch Clin Exp Ophthalmol. 2014; 252: 1939–1946.
Feldkaemper M, Schaeffel F. An updated view on the role of dopamine in myopia. Exp Eye Res. 2013; 114: 106–119.
Rohrer B, Spira AW, Stell WK. Apomorphine blocks form-deprivation myopia in chickens by a dopamine D2-receptor mechanism acting in retina or pigmented epithelium. Vis Neurosci. 1993; 10: 447–453.
Stone RA, Lin T, Laties AM, Iuvone PM. Retinal dopamine and form-deprivation myopia. Proc Natl Acad Sci U S A. 1989; 86: 704–706.
Ganesan P, Wildsoet CF. Pharmaceutical intervention for myopia control. Expert Rev Ophthalmol. 2010; 5: 759–787.
George A, Schmid KL, Pow DV. Retinal serotonin, eye growth and myopia development in chick. Exp Eye Res. 2005; 81: 616–625.
Ohngemach S, Hagel G, Schaeffel F. Concentrations of biogenic amines in fundal layers in chickens with normal visual experience, deprivation, and after reserpine application. Vis Neurosci. 1997; 14: 493–505.
Cohen J, Hadjiconstantinou M. Identification of epinephrine and phenylethanolamine N-methyltransferase activity in rat retina. Fed Proc. 1984; 43: 2725–2728.
Hadjiconstantinou M, Cohen J, Neff NH. Epinephrine: a potential neurotransmitter in retina. J Neurochem. 1983; 41: 1440–1444.
Hadjiconstantinou M, Mariani AP, Panula P, Joh TH, Neff NH. Immunohistochemical evidence for epinephrine-containing retinal amacrine cells. Neuroscience. 1984; 13: 547–551.
Woldemussie E, Wijono M, Pow D. Localization of alpha 2 receptors in ocular tissues. Vis Neurosci. 2007; 24: 745–756.
Harun-Or-Rashid M, Lindqvist N, Hallböök F. Transactivation of EGF receptors in chicken Müller cells by alpha2A-adrenergic receptors stimulated by brimonidine. Invest Ophthalmol Vis Sci. 2014; 55: 3385–3394.
Fong DS, Epstein DL, Allingham RR. Glaucoma and myopia: are they related? Int Ophthalmol Clin. 1990; 30: 215–218.
Mitchell P, Hourihan F, Sandbach J, Wang JJ. The relationship between glaucoma and myopia: the Blue Mountains Eye Study. Ophthalmology. 1999; 106: 2010–2015.
Pruett RC. Progressive myopia and intraocular pressure: what is the linkage? A literature review. Acta Ophthalmol Suppl. 1988; 185: 117–127.
Nickla DL, Wildsoet C, Wallman J. The circadian rhythm in intraocular pressure and its relation to diurnal ocular growth changes in chicks. Exp Eye Res. 1998; 66: 183–193.
Jiang WJ, Wu JF, Hu YY, et al. Intraocular pressure and associated factors in children: the Shandong children eye study. Invest Ophthalmol Vis Sci. 2014; 55: 4128–4134.
Quinn GE, Berlin JA, Young TL, Ziylan S, Stone RA. Association of intraocular pressure and myopia in children. Ophthalmology. 1995; 102: 180–185.
Manny RE, Deng L, Crossnoe C, Gwiazda J. IOP, myopic progression and axial length in a COMET subgroup. Optom Vis Sci. 2008; 85: 97–105.
Lee AJ, Saw SM, Gazzard G, Cheng A, Tan DT. Intraocular pressure associations with refractive error and axial length in children. Br J Ophthalmol. 2004; 88: 5–7.
Li S, Li SM, Wang XL, et al. Distribution and associations of intraocular pressure in 7- and 12-year-old Chinese children: the Anyang Childhood Eye Study. PLoS One. 2017; 12: e0181922.
Goss DA, Caffey TW. Clinical findings before the onset of myopia in youth: 5. Intraocular pressure. Optom Vis Sci. 1999; 76: 286–291.
Edwards MH, Brown B. IOP in myopic children: the relationship between increases in IOP and the development of myopia. Ophthalmic Physiol Opt. 1996; 16: 243–246.
Schmid KL, Hills T, Abbott M, Humphries M, Pyne K, Wildsoet CF. Relationship between intraocular pressure and eye growth in chick. Ophthalmic Physiol Opt. 2003; 23: 25–33.
Doughty MJ, Zaman ML. Human corneal thickness and its impact on intraocular pressure measures: a review and meta-analysis approach. Surv Ophthalmol. 2000; 44: 367–408.
François J. The importance of scleral rigidity in ocular tonometry. Indian J Ophthalmol. 1960; 8: 16–24.
Green K. Techniques of intraocular pressure determination. Lens Eye Toxic Res. 1990; 7: 485–489.
Curtin BJ, Teng CC. Scleral changes in pathological myopia. Trans Am Acad Ophthalmol Otolaryngol. 1958; 62: 777–788; discussion 788–790.
Siegwart JTJr, Norton TT. Regulation of the mechanical properties of tree shrew sclera by the visual environment. Vision Res. 1999; 39: 387–407.
Phillips JR, Khalaj M, McBrien NA. Induced myopia associated with increased scleral creep in chick and tree shrew eyes. Invest Ophthalmol Vis Sci. 2000; 41: 2028–2034.
Wen R, Cheng T, Li Y, Cao W, Steinberg RH. α2-adrenergic agonists induce basic fibroblast growth factor expression in photoreceptors in vivo and ameliorate light damage. J Neurosci. 1996; 16: 5986–5992.
Chao HM, Chidlow G, Melena J, Wood JP, Osborne NN. An investigation into the potential mechanisms underlying the neuroprotective effect of clonidine in the retina. Brain Res. 2000; 877: 47–57.
Rohrer B, Stell WK. Basic fibroblast growth factor (bFGF) and transforming growth factor beta (TGF-β) act as stop and go signals to modulate postnatal ocular growth in the chick. Exp Eye Res. 1994; 58: 553–561.
Penha AM, Burkhardt E, Schaeffel F, Feldkaemper MP. Effects of intravitreal insulin and insulin signaling cascade inhibitors on emmetropization in the chick. Mol Vis. 2012; 18: 2608–2622.
Chen BY, Wang CY, Chen WY, Ma JX. Altered TGF-β2 and bFGF expression in scleral desmocytes from an experimentally-induced myopia guinea pig model. Graefes Arch Clin Exp Ophthalmol. 2013; 251: 1133–1144.
Tian XD, Cheng YX, Liu GB, et al. Expressions of type I collagen, α2 integrin and β1 integrin in sclera of guinea pig with defocus myopia and inhibitory effects of bFGF on the formation of myopia. Int J Ophthalmol. 2013; 6: 54–58.
Rosa RHJr, Hein TW, Yuan Z, et al. Brimonidine evokes heterogeneous vasomotor response of retinal arterioles: diminished nitric oxide-mediated vasodilation when size goes small. Am J Physiol Heart Circ Physiol. 2006; 291: H231–H238.
Arumugam B. Strategies to Inhibit Myopia: Pharmacological and Optical Approaches [doctoral thesis]. Melbourne: Optometry and Vision Sciences, University of Melbourne; 2011.
Morgan IG, Ashby RS, Nickla DL. Form deprivation and lens-induced myopia: are they different? Ophthalmic Physiol Opt. 2013; 33: 355–361.
Schaeffel F, Hagel G, Bartmann M, Kohler K, Zrenner E. 6-Hydroxy dopamine does not affect lens-induced refractive errors but suppresses deprivation myopia. Vision Res. 1994; 34: 143–149.
Choh V, Lew MY, Nadel MW, Wildsoet CF. Effects of interchanging hyperopic defocus and form deprivation stimuli in normal and optic nerve-sectioned chicks. Vision Res. 2006; 46: 1070–1079.
Kashihara K, Varga EV, Waite SL, Roeske WR, Yamamura HI. Cloning of the rat M3, M4 and M5 muscarinic acetylcholine receptor genes by the polymerase chain reaction (PCR) and the pharmacological characterization of the expressed genes. Life Sci. 1992; 51: 955–971.
Jolkkonen M, van Giersbergen PL, Hellman U, Wernstedt C, Karlsson E. A toxin from the green mamba Dendroaspis angusticeps: amino acid sequence and selectivity for muscarinic m4 receptors. FEBS Lett. 1994; 352: 91–94.
Lazareno S, Birdsall NJ. Pharmacological characterization of acetylcholine-stimulated [35S]-GTP gamma S binding mediated by human muscarinic m1-m4 receptors: antagonist studies. Br J Pharmacol. 1993; 109: 1120–1127.
Eglen RM, Whiting RL. Competitive and non-competitive antagonism exhibited by “selective” antagonists at atrial and ileal muscarinic receptor subtypes. Br J Pharmacol. 1987; 90: 701–707.
Lazareno S, Buckley NJ, Roberts FF. Characterization of muscarinic M4 binding sites in rabbit lung, chicken heart, and NG108-15 cells. Mol Pharmacol. 1990; 38: 805–815.
Bylund DB, Rudeen PK, Petterborg LJ, Ray-Prenger C. Identification of α2-adrenergic receptors in chicken pineal gland using [3H]rauwolscine. J Neurochem. 1988; 51: 81–86.
Newman-Tancredi A, Nicolas JP, Audinot V, et al. Actions of α2 adrenoceptor ligands at α2A and 5-HT1A receptors: the antagonist, atipamezole, and the agonist, dexmedetomidine, are highly selective for α2A adrenoceptors. Naunyn Schmiedebergs Arch Pharmacol. 1998; 358: 197–206.
Randall WC, Baldwin JJ, Cresson EL, Tolman RL, Weppelman RM, Lyon TF. Multiple central α2 adrenoceptors of avian and mammalian species. Biochem Pharmacol. 1983; 32: 1933–1940.
Figure 1
 
Inhibitory potencies of various mAChR antagonists at the human M4 receptor. Antagonists are categorized into full myopia inhibition (open/colored symbols), partial myopia inhibition (partially filled symbols), and no myopia inhibition (filled symbols) according to previously published research,14,16,19 and ranked in descending order from most to least potent in our receptor assay (IC50 ± 95% CI). MT3 (yellow) and atropine (blue) are indicated separately as they are the most researched anti-myopia mAChR antagonist in humans (atropine), and the most potent anti-myopia drug found to date in chicks (MT3). ATR, atropine sulfate; OXY, oxyphenonium bromide; PRZ, pirenzepine dihydrochloride; CI, confidence interval.
Figure 1
 
Inhibitory potencies of various mAChR antagonists at the human M4 receptor. Antagonists are categorized into full myopia inhibition (open/colored symbols), partial myopia inhibition (partially filled symbols), and no myopia inhibition (filled symbols) according to previously published research,14,16,19 and ranked in descending order from most to least potent in our receptor assay (IC50 ± 95% CI). MT3 (yellow) and atropine (blue) are indicated separately as they are the most researched anti-myopia mAChR antagonist in humans (atropine), and the most potent anti-myopia drug found to date in chicks (MT3). ATR, atropine sulfate; OXY, oxyphenonium bromide; PRZ, pirenzepine dihydrochloride; CI, confidence interval.
Figure 2
 
Inhibitory potencies of various mAChR antagonists at the chicken cM4 receptor. Antagonists are categorized into full myopia inhibition (open/colored symbols), partial myopia inhibition (partially filled symbols), and no myopia inhibition (filled symbols) according to previously published research,14,16,19 and ranked in descending order from most to least potent in our receptor assay (IC50 ± 95% CI). MT3 (yellow) and atropine (blue) are indicated separately as they are the most researched anti-myopia mAChR antagonist in humans (atropine), and the most potent anti-myopia drug found to date in chicks (MT3). ATR, atropine sulfate; OXY, oxyphenonium bromide; PRZ, pirenzepine dihydrochloride; CI, confidence interval.
Figure 2
 
Inhibitory potencies of various mAChR antagonists at the chicken cM4 receptor. Antagonists are categorized into full myopia inhibition (open/colored symbols), partial myopia inhibition (partially filled symbols), and no myopia inhibition (filled symbols) according to previously published research,14,16,19 and ranked in descending order from most to least potent in our receptor assay (IC50 ± 95% CI). MT3 (yellow) and atropine (blue) are indicated separately as they are the most researched anti-myopia mAChR antagonist in humans (atropine), and the most potent anti-myopia drug found to date in chicks (MT3). ATR, atropine sulfate; OXY, oxyphenonium bromide; PRZ, pirenzepine dihydrochloride; CI, confidence interval.
Figure 3
 
Inhibitory potencies of mAChR antagonists at hADRA2A. Antagonists are categorized into full myopia inhibition (open/colored symbols), partial myopia inhibition (partially filled symbols), and no myopia inhibition (filled symbols) according to previously published research,14,16,19 and ranked in descending order from most to least potent in our receptor assay (IC50 ± 95% CI). MT3 (yellow) and atropine (blue) are indicated separately as they are the most researched anti-myopia mAChR antagonist in humans (atropine), and the most potent anti-myopia drug found to date in chicks (MT3). In this assay, dicyclomine is indicated by an open circle with a cross, because it had no activity at the hADRA2A receptor at the concentrations tested. YOH, yohimbine hydrochloride; ATR, atropine sulfate; OXY, oxyphenonium bromide; PRZ, pirenzepine dihydrochloride; CI, confidence interval.
Figure 3
 
Inhibitory potencies of mAChR antagonists at hADRA2A. Antagonists are categorized into full myopia inhibition (open/colored symbols), partial myopia inhibition (partially filled symbols), and no myopia inhibition (filled symbols) according to previously published research,14,16,19 and ranked in descending order from most to least potent in our receptor assay (IC50 ± 95% CI). MT3 (yellow) and atropine (blue) are indicated separately as they are the most researched anti-myopia mAChR antagonist in humans (atropine), and the most potent anti-myopia drug found to date in chicks (MT3). In this assay, dicyclomine is indicated by an open circle with a cross, because it had no activity at the hADRA2A receptor at the concentrations tested. YOH, yohimbine hydrochloride; ATR, atropine sulfate; OXY, oxyphenonium bromide; PRZ, pirenzepine dihydrochloride; CI, confidence interval.
Figure 4
 
Correlations between rank order inhibitory potency (x-axis) and rank order inhibition of axial elongation in chicks with experimentally induced FDM (y-axis) for mAChR antagonists at M4 (a), cM4 (b), and hADRA2A (c). Rank orders for both parameters are 1 = least potent (pIC50) or effective (% axial length inhibition) and 9 = most potent (pIC50) or effective (% axial length inhibition). A line of unity (slope = 1) has been superimposed upon the graphs for visual reference. Inhibitory potencies of mAChR antagonists, and their abilities to inhibit FDM in the chick, correlate significantly for hADRA2A only (r = 0.87, P < 0.01; Spearman's ranked correlation). Atropine and MT3 are shown in color (atropine = blue, MT3 = yellow). Full inhibition of axial elongation (open/colored symbols), partial inhibition of axial elongation (partially filled symbols), and inhibition of axial elongation (filled symbols) according to previously published research in the chick.14,16,19
Figure 4
 
Correlations between rank order inhibitory potency (x-axis) and rank order inhibition of axial elongation in chicks with experimentally induced FDM (y-axis) for mAChR antagonists at M4 (a), cM4 (b), and hADRA2A (c). Rank orders for both parameters are 1 = least potent (pIC50) or effective (% axial length inhibition) and 9 = most potent (pIC50) or effective (% axial length inhibition). A line of unity (slope = 1) has been superimposed upon the graphs for visual reference. Inhibitory potencies of mAChR antagonists, and their abilities to inhibit FDM in the chick, correlate significantly for hADRA2A only (r = 0.87, P < 0.01; Spearman's ranked correlation). Atropine and MT3 are shown in color (atropine = blue, MT3 = yellow). Full inhibition of axial elongation (open/colored symbols), partial inhibition of axial elongation (partially filled symbols), and inhibition of axial elongation (filled symbols) according to previously published research in the chick.14,16,19
Table 1
 
Binding Data for Muscarinic Antagonists Tested at mAChR M4 and cM4 in Our Assay
Table 1
 
Binding Data for Muscarinic Antagonists Tested at mAChR M4 and cM4 in Our Assay
Table 2
 
Binding Data for α2-Adrenergic Drugs and mAChR Antagonists at hADRA2A
Table 2
 
Binding Data for α2-Adrenergic Drugs and mAChR Antagonists at hADRA2A
Supplement 1
×
×

This PDF is available to Subscribers Only

Sign in or purchase a subscription to access this content. ×

You must be signed into an individual account to use this feature.

×