99
Views
10
CrossRef citations to date
0
Altmetric
Original Research

Mapracorat, a selective glucocorticoid receptor agonist, causes apoptosis of eosinophils infiltrating the conjunctiva in late-phase experimental ocular allergy

, , , , &
Pages 745-757 | Published online: 10 Jun 2014

Abstract

Background

Mapracorat, a novel nonsteroidal selective glucocorticoid receptor agonist, has been proposed for the topical treatment of inflammatory disorders as it binds with high affinity and selectivity to the human glucocorticoid receptor and displays a potent anti-inflammatory activity, but seems to be less effective in transactivation of a number of genes, resulting in a lower potential for side effects. Contrary to classical glucocorticoids, mapracorat displays a reduced ability to increase intraocular pressure and in inducing myocilin, a protein linked to intraocular pressure elevation. Allergic conjunctivitis is the most common form of ocular allergy and can be divided into an early phase, developing immediately after allergen exposure and driven primarily by mast cell degranulation, and a late phase, developing from 6–10 hours after the antigen challenge, and characterized by conjunctival infiltration of eosinophils and other immune cells as well as by the production of cytokines and chemokines.

Methods

In this study, mapracorat was administered into the conjunctival sac of ovalbumin (OVA)-sensitized guinea pigs 2 hours after the induction of allergic conjunctivitis, with the aim of investigating its activity in reducing clinical signs of the late-phase ocular reaction and to determine its mechanism of anti-allergic effects with respect to apoptosis of conjunctival eosinophils and expression of the chemokines C-C motif ligand 5 (CCL5), C-C motif ligand 11 (CCL11), and interleukin-8 (IL-8) and the proinflammatory cytokines interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α).

Results

Mapracorat, administered into the conjunctival sac of OVA-sensitized guinea pigs 2 hours after allergen exposure, was effective in reducing clinical signs, eosinophil infiltration, and eosinophil peroxidase activity in the guinea pig conjunctiva; furthermore, it reduced conjunctival mRNA levels and protein expression of both CCL5 and CCL11. Mapracorat was more effective than dexamethasone in increasing, in conjunctival sections of OVA-treated guinea pigs, apoptotic eosinophils.

Conclusion

Mapracorat displays anti-allergic properties in controlling the late phase of ocular allergic conjunctivitis and is a promising candidate for the topical treatment of allergic eye disorders.

Introduction

Allergic conjunctivitis is the most common form of ocular allergy and is usually associated with type 1 hypersensitivity reactions, characterized by early-phase and late-phase responses. Common symptoms include ocular itching, hyperemia, tearing, and chemosis. The early-phase response is driven primarily by mast cell degranulation and develops immediately after exposure to the allergen. This is followed by the late-phase response after 6–10 hours, which is characterized by infiltration of eosinophils, neutrophils, and lymphocytes into the conjunctiva.Citation1,Citation2 Activation and recruitment of inflammatory cells and the release of cytokines, chemokines, adhesion molecules, and proteases contribute to more serious chronic forms.Citation3,Citation4

Topical glucocorticoids are among the most effective drugs for the treatment of allergic eye disease,Citation2 as they potently inhibit a wide range of cytokines and chemokines that contribute to eosinophil accumulation and activation.Citation5 However, glucocorticoid use is associated with increased intraocular pressure, risk of cataract formation, and decreased resistance to infections. There is, therefore, a pressing need for compounds with the anti-inflammatory potency of standard glucocorticoids but fewer or less troublesome side effects.Citation6

Most of the effects of glucocorticoids on target cells are mediated by the regulation of transcription of steroid-responsive genes. It is thought that anti-inflammatory effects of glucocorticoids are largely mediated by a mechanism known as transrepression.Citation7 The glucocorticoid receptor is recruited to DNA via direct or indirect interactions with other transcription factors, notably, members of the activating protein 1 and nuclear factor-kB families, and inhibits transcriptional activation by impairing recruitment of transcriptional coactivators or by promoting recruitment of corepressors.Citation8,Citation9 There is also evidence that glucocorticoid-mediated repression of inflammatory genes involves significant posttranscriptional and/or translational mechanisms,Citation9 and the requirement for de novo protein synthesis has been highlighted.Citation11,Citation12 In contrast, certain side effects seem to be mediated mainly through trans-activation, in which the glucocorticoid-bound glucocorticoid receptor directly binds to DNA through simple palindromic glucocorticoid response elements.Citation7,Citation8

A deeper understanding of the molecular mode of glucocorticoid action has led to the identification of novel selective glucocorticoid receptor agonists that should preserve the beneficial anti-inflammatory activity but offer a better side effect profile.Citation12 However, the utility of dissociated glucocorticoid ligands, as more effective anti-inflammatory compounds with fewer side effects, is still debated,Citation9Citation11 and studies aimed to investigate their pharmacological profile are needed. Schäcke et alCitation13 have reported the pharmacological characterization of mapracorat (also known as BOL-303242-X or ZK 245186), a nonsteroidal selective glucocorticoid receptor agonist, for the topical treatment of inflammatory disorders (). Mapracorat binds with high affinity and selectivity to the human glucocorticoid receptor and possesses potent anti-inflammatory activity, but seems to be less effective in transactivation of a number of genes,Citation13 resulting in a lower potential for metabolic and ocular side effects. Physicochemical and pharmacokinetic properties of mapracorat suggest that it is particularly suited for topical treatment, displaying a low systemic activity. In fact, in vitro mapracorat is rapidly metabolized by liver microsomes and, in vivo, it shows a high hepatic clearance and a high volume of distribution. Mapracorat has a short half-life (2 hours) after intravenous administration and a low bioavailability (9%) after topical administration.Citation13 These properties suggest that mapracorat meets the criteria to be considered a “soft” glucocorticoid agonist as it behaves like budesonide.Citation14

Figure 1 Chemical structure of mapracorat.

Figure 1 Chemical structure of mapracorat.

Ocular pharmacokinetics of mapracorat were investigated by Proksch et alCitation15 after topical administration over a wide range of doses in rabbits and monkeys. Mapracorat is rapidly absorbed and widely distributed into ocular tissues after conjunctival administration, with measurable levels up to 24 hours after dosing. Higher concentrations are detected in tears, followed by conjunctiva and cornea, with lower levels observed in iris/ciliary body and aqueous humor. Plasma levels are lower than those found for other glucocorticoids like dexamethasone. Taken together, these data suggest that mapracorat has a favorable profile of activity in comparison to classical glucocorticoids and may be suitable for topical ocular administration adopting a daily dosing regimen.

Mapracorat, topically administered as eye drops, displays a reduced ability to increase intraocular pressure in normotensive rabbits when compared to dexamethasoneCitation16 and behaves as a partial glucocorticoid receptor agonist in inducing myocilin, a protein linked to intraocular pressure elevation, in monkey trabecular meshwork cells.Citation17 Thus, it would be expected to possess an ocular safety profile superior to other glucocorticoids.

Previously, we reported that mapracorat promotes in vitro eosinophil spontaneous apoptosis and inhibits eosinophil migration and release of proinflammatory cytokines and chemokines from eosinophils,Citation18 mast cells,Citation17 or conjunctival epithelial cells, fibroblasts,Citation19 and corneal epithelial cells.Citation20

Exacerbations of allergic conjunctivitis cause eosinophils to infiltrate the conjunctiva, where they contribute to tissue derangement, inflammation, and remodeling.Citation3,Citation21 The chemokines C-C motif ligand 5 (CCL5) and C-C motif ligand 11 (CCL11) play a key role in recruiting eosinophils and other immune cells to the inflammation areaCitation22 and are expressed in the conjunctiva of patients with vernal keratoconjunctivitis.Citation23,Citation24 Eosinophil response to inflammatory stimuli leads to increased production of proinflammatory cytokines like interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) and chemokines including interleukin-8 (IL-8).Citation25

Together with other interventions, the removal of eosinophils from the conjunctival epithelium and stroma should contribute to the resolution of the conjunctivitis. To accomplish cell removal, investigations are focused on the pharmacology of apoptosis. This process is considered a noninflammatory model of cell death because apoptotic cells are eliminated through phagocytosis.Citation26

Glucocorticoid-mediated eosinophil apoptosis has been widely investigated with the adoption of in vitro cell models;Citation27Citation29 however, its occurrence in the airway tissue after glucocorticoid treatment of allergic diseases is still debated.Citation30Citation32 Studies demonstrating conjunctival eosinophil apoptosis, caused by glucocorticoid treatment at the conjunctival level, are scarce. To date, the potential anti-allergic activity of mapracorat in the eye, and whether eosinophil apoptosis and the chemokines CCL5 and CCL11 are the targets of its action, has been explored very little in vivo. This study specifically addressed these questions, adopting a guinea pig model of allergic conjunctivitis. Mapracorat was tested alongside dexamethasone, a traditional glucocorticoid used to treat ocular inflammation.Citation2 Both drugs were topically administered 2 hours after antigen challenge.

Materials and methods

Materials

Mapracorat was provided by Bausch & Lomb Inc. (Rochester, NY, USA) and dissolved in 10% polyethylene glycol 3350/1% polysorbate 80 in phosphate-buffered saline (PBS) pH 7.0 (vehicle). Dexamethasone 21 disodium phosphate was obtained from Visufarma (Rome, Italy) and diluted in phosphate-buffered saline (PBS). Ovalbumin (OVA) grade V, aluminum hydroxide, o-phenylenediamine, hydrogen peroxide 30%, Triton X-100™, peroxidase acidic isoenzyme from horseradish, RNAlater®, and TRI Reagent® were obtained from Sigma-Aldrich (St Louis, MO, USA). RNase-free DNase was from Thermo Fisher Scientific (Waltham, MA, USA). Rabbit specific horseradish peroxidase (HRP)/diaminobenzidine (DAB) detection kit, anti-CCL11, and anti-CCL5 antibodies were from Abcam (Cambridge, UK).

Induction of allergic conjunctivitis by active immunization and eosinophil detection

The experimental protocol was approved by the Ethical Committee for Animal Experiments of the University of Bologna, Bologna, Italy, and conformed to the EU Directive 2010/63/EU statement for animal experiments.

Allergic conjunctivitis was induced in male Dunkin-Hartley guinea pigs (250–300 g), purchased from Charles River Laboratory Inc. (Calco, Italy), as previously described.Citation33 Guinea pigs were intraperitoneally (ip) treated, except the negative controls, once a week for 2 weeks with 2 mL of a saline solution containing 100 μg/mL OVA and 20 mg/mL aluminum hydroxide as adjuvant. Three weeks after the first OVA treatment, all animals were examined to ensure that there was no sign of preexisting ocular inflammation and then challenged with 30 μL per eye of saline solution containing 100 mg/mL OVA or with 30 μL per eye of saline solution (controls). Over 97% of the sensitized animals treated with topical OVA developed an allergic ocular reaction; guinea pigs that did not respond to the challenge were excluded from the study. Mapracorat and dexamethasone (0.1%, 0.25%, 0.4%; w/v) or the vehicle alone were administered in the conjunctival sac of both eyes (30 μL/eye), 2 hours after OVA challenge (groups of five guinea pigs). Both compounds were administered in a range of doses, including those of dexamethasone used in clinical studies (0.1% or 0.2%) and that of mapracorat previously employed in another in vivo study (0.4%).Citation16

Conjunctival clinical symptoms were rated in a masked fashion on both eyes using the following scale: 0, no symptoms; 1, slight conjunctival redness with or without tears; 2, mild conjunctival redness with or without tears and mild chemosis; 3, mild conjunctival redness with or without tears and moderate chemosis; 4, severe conjunctival redness with tears and partial lid eversion; 5, severe conjunctival redness with tears and lids more than half closed. Pictures of both eyes were taken to evaluate the clinical score 30 minutes before and 1, 4, 6, 8, and 24 hours after OVA administration. The animals were euthanized 8 or 24 hours after OVA challenge by ip injection of Tanax® (3 mL/kg; Hoechst AG, Frankfurt am Main, Germany). Tarsal conjunctiva of both eyes was carefully excised and divided into separate samples for subsequent investigations. One sample was fixed in 10% buffered paraformaldehyde solution and paraffin embedded. Slides, 6 μm thick, were stained to determine the number of eosinophilsCitation33 or to carry out immunohistochemistry. To evaluate accumulation and distribution of eosinophils in the tarsal conjunctiva, slides were stained with Luna’s eosinophil stain. Slides were desiccated in xylene, stained with hematoxylin–Biebrich scarlet solution, differentiated in 1% acid alcohol, and subsequently stained with lithium carbonate. Eosinophil granules stain red-orange.Citation33 The number of eosinophils in each field was counted under light microscopy (500× magnification).

In separate conjunctival specimens, eosinophil peroxidase activity was measured as previously described.Citation33 Eosinophil peroxidase activity was also measured in ophthalmic lavage fluid (OLF) as previously described.Citation34 OLF was collected as follows: a 30 μL aliquot of buffer consisting of 0.3 M sucrose in 50 mM sodium acetate buffer pH 5.4 with 10 units/mL heparin was applied to the eye using a micropipette, without touching the eye. After two or three forced blinks, 30 μL of lavage fluid was collected. The lavage was repeated three times in each eye and the OLF was collected in the same tube.

Immunohistochemistry

On conjunctival slices, after paraffin removal and rehydration, heat-mediated antigen retrieval was performed with citrate buffer (pH 6.0) for 20 minutes and the sections were washed three times in Tris-buffered saline containing 0.025% of Tween 20® (TBS-T) (Cayman Chemical Company, MI, USA). Immunohistochemical staining was made using rabbit specific HRP/DAB detection kit (Abcam), following the manufacturer’s instructions. Briefly, specimens were incubated with Protein Block solution (Abcam) for 5 minutes at room temperature to block nonspecific background staining. After washing once in TBS-T, tissue sections were incubated with antibodies (100 μL) that recognized human CCL11 or CCL5 (diluted 1:250) overnight at 4°C. The specimens were washed in TBS-T and incubated with biotinylated goat anti-rabbit immunoglobulin G for 10 minutes at room temperature. Slides were then developed with streptavidin–biotin complex/HRP and DAB substrate, according to the manufacturer’s instructions. Hematoxylin was used as background staining. Sections were examined with a light microscope in a blinded manner and photomicrographs were taken with a 20× objective. Coded slides were further analyzed using a validated semiquantitative scoring method based on the percentage of conjunctival tissue showing positive immunoreactive staining.Citation35,Citation36 Immunohistochemical scores were assigned according to five categories (0: no staining; 1: <25% staining; 2: 25%–50% staining: 3, 50%–75% staining; and 4: >75% staining).

Detection of apoptotic cells with the terminal deoxynucleotidyl transferase dUTP nick end labeling technique

Sections of tarsal conjunctiva (6 μm) were deparaffinized, rehydrated, and permeabilized with proteinase K (20 μg/mL) for 20 minutes at room temperature. Apoptotic cells were visualized by a fluorescein isothiocyanate-linked terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assayCitation37 with the FragEL™ DNA Fragmentation Detection Kit (Merck Millipore, Billerica, MA, USA), following manufacturer instructions. No staining was evident in negative controls when omitting the terminal deoxynucleotidyl transferase enzyme. Slides were counterstained with the DNA-binding stain DAPI (4′,6-diamidino-2-phenylindole) to reveal pyknotic nuclei as well as the total number of cells. Apoptotic cells were defined as TUNEL-positive cells exhibiting apoptotic morphology (small cells with condensed nuclei). Specimens were analyzed with a microscope (Nikon Eclipse E800 [Nikon Instruments Europe BV, Amsterdam, the Netherlands]); TUNEL-positive cells per mm2 were counted and digital images were captured using a standard fluorescein filter (465–495 nm) and a filter for DAPI (330–380 nm).

Detection of apoptotic eosinophils

Combined staining with TUNEL and chromotrope-2R identifying apoptotic eosinophils was performed as previously described.Citation31,Citation38,Citation39 After deparaffinization, tissue sections were incubated with chromotrope-2R solution for 30 minutes. Slides were washed twice with tap water and then stained with the FragEL™ DNA kit, as described in the previous paragraph. Apoptotic eosinophils were defined as both chromotrope-2R-positive and TUNEL-positive cells exhibiting apoptotic morphology (small cells with condensed nuclei).

Quantitative reverse transcriptase polymerase chain reaction

Tarsal conjunctiva specimens were stored in RNAlater at −20°C. Total RNA was extracted, after homogenization, with TRI Reagent and quantified using a spectrophotometer. For each sample, 1 μg of total RNA was treated with RNase-free DNase for 15 minutes at 25°C, followed by incubation at 65°C for 10 minutes to inactivate DNase. The RNA samples were then converted into cDNA using oligo(dT), random primers, and the High-Capacity cDNA Reverse Transcription Kits (Life Technologies Italia), according to the manufacturer’s instructions. Real-time PCR was performed using GoTaq® qPCR Master Mix obtained from Promega Corporation (Madison, WI, USA). The protocol consisted of denaturation at 95°C for 10 minutes, followed by 40 cycles of 95°C denaturation (20 seconds) and 58°C annealing (1 minute). No-template controls and DNA melting curve analysis were used as controls to ensure the lack of contaminating DNA in the RNA preparations and to rule out primer-dimer formation, respectively. Induction of mRNA was determined from the threshold cycle (Ct) values normalized for glyceraldehyde 3-phosphate dehydrogenase (GAPDH) expression and then normalized to the value derived from conjunctivae of non-immunized guinea pigs. Primer sequences of GAPDH, IL-8, IL-1β, TNF-α, CCL11, and CCL5 have been previously publishedCitation40,Citation41 and are listed in . Primers were synthesized by Sigma-Aldrich.

Table 1 Primers employed in real-time polymerase chain reaction

Data analysis

Data are expressed as mean ± standard error of the mean or median ± standard error of the mean for the number of experiments indicated. Statistical comparisons were made by analysis of variance and post hoc Newman–Keuls test. Nonparametric analysis of the scores assigned to the conjunctival symptoms was done using the Friedman test followed by Dunn’s post hoc comparison. Significant differences among immunohistochemical scores were tested by the Kruskal–Wallis test followed by Dunn’s post hoc comparison. Data analysis was done using GraphPad Prism (v 4.0; GraphPad Software, Inc., La Jolla, CA, USA). P-values <0.05 were considered significant.

Results

Mapracorat and dexamethasone eye drops reduce late-phase allergic reaction in guinea pigs

Guinea pigs were actively immunized by ip injection of OVA and challenged with OVA instilled into the conjunctival sac. One hour after challenge, during the early-phase reaction, clinical observations revealed typical early-phase symptoms of allergic conjunctivitis: tearing and discharge, conjunctival redness, and chemosis. These clinical symptoms showed a second rise from 4–8 hours after allergen challenge in the late-phase reaction and then progressively decreased;Citation42 24 hours after challenge, conjunctiva did not present any relevant clinical symptom.

Mapracorat and dexamethasone (0.1%, 0.25%, and 0.4%) administered in the conjunctival sac of both eyes 2 hours after OVA challenge were equally effective in reducing inflammatory signs observed in the late phase. Both drugs caused a dose-dependent response and a 0.4% dose was the most effective ().

Figure 2 Map and Dex inhibit the clinical signs of late-phase conjunctival allergic inflammation to OVA in sensitized guinea pigs.

Notes: Map and Dex eye drops (0.1%, 0.25%, and 0.4%; w/v) or the vehicle (30 μL/eye) were administered in the conjunctival sac of both eyes 2 hours after OVA challenge (30 μL of 2.5% solution). Ctrls received the vehicle alone and were not treated with topical OVA. Each group comprised five guinea pigs, and the score, evaluated on both eyes (n=10) was based on changes observed 4, 6, and 8 hours after OVA challenge (2, 4, and 6 hours after drug treatment) for the symptoms of itching, swelling, redness, and lid eversion and was rated in a masked fashion using the following scale: 0, no symptoms; 1, slight conjunctival redness with or without tears; 2, mild conjunctival redness with or without tears and mild chemosis; 3, mild conjunctival redness with or without tears and moderate chemosis; 4, severe conjunctival redness with tears and partial lid eversion; 5, severe conjunctival redness with tears and lids more than half closed. Data are presented as boxes and whiskers. The boxes represent the 25th to 75th percentiles and horizontal lines within each box represent the median values of all the data for all time points together. The whiskers are the minimum and the maximum of all the data (n=10; both eyes were evaluated). § P<0.01 versus OVA + vehicle; # P<0.01 versus Ctrl (Friedman test followed by Dunn’s post hoc comparison).
Abbreviations: Ctrl, control; Dex, dexamethasone; Map, mapracorat; OVA, ovalbumin.
Figure 2 Map and Dex inhibit the clinical signs of late-phase conjunctival allergic inflammation to OVA in sensitized guinea pigs.

In addition to the clinical response, late-phase allergic reaction was evidenced by a significant increase in eosinophil peroxidase activity in the OLF collected 4, 6, and 8 hours after topical OVA challenge, taken as an index of the occurrence of eosinophils in tear fluids; conversely, 30 minutes before and 24 hours after OVA challenge, eosinophil peroxidase activity was not increased in the collected OLF. These data are indicative of conjunctival infiltration of circulating eosinophils that can also reach the tears. Mapracorat and dexamethasone were equally effective in reducing, in a dose-related manner, eosinophil peroxidase activity in tears ().

Figure 3 Map and Dex reduce eosinophil peroxidase activity in OLF.

Notes: Both drugs (0.1%, 0.25%, and 0.4%; w/v) or the vehicle (30 μL/eye) were administered in the conjunctival sac of both eyes 2 hours after OVA challenge (30 μL of 2.5% solution). Ctrls received the vehicle alone and were not treated with topical OVA. Each group comprised five guinea pigs and both eyes were evaluated (n=10). Values are the mean ± standard error of the mean of ten values (samples of both eyes of five guinea pigs were collected) per group. *P<0.05; § P<0.01 versus OVA + vehicle; # P<0.01 versus Ctrl (two-way ANOVA with Bonferroni post test).
Abbreviations: Br–, bromide; Ctrl, control; Dex, dexamethasone; EPO, eosinophil peroxidase; Map, mapracorat; OD, optical density; OLF, ophthalmic lavage fluid; OVA, ovalbumin.
Figure 3 Map and Dex reduce eosinophil peroxidase activity in OLF.

The occurrence of a late-phase allergic reaction was confirmed by histological analysis of tarsal conjunctiva specimens, obtained from guinea pigs sacrificed 24 hours after OVA challenge, containing numerous infiltrating eosinophils (). Similarly, a significant increase of conjunctival eosinophil peroxidase activity was measured (). Mapracorat and dexamethasone were equally effective in reducing, in a dose-related manner, eosinophil infiltration and eosinophil peroxidase activity in the guinea pig conjunctiva ().

Figure 4 Map and Dex eye drops reduce eosinophil infiltration by OVA in guinea pig conjunctiva.

Notes: Map or Dex eye drops (0.1%, 0.25%, and 0.4%; w/v) or the vehicle (30 μL/eye) were administered in the conjunctival sac of both eyes 2 hours after OVA challenge (30 μL of 2.5% solution). Ctrls received the vehicle alone and were not treated with topical OVA. (A) Effect of Map or Dex eye drops on conjunctival eosinophil infiltration 24 hours after topical challenge with OVA. (B) Effect of Map or Dex eye drops on conjunctival eosinophil peroxidase levels 24 hours after topical challenge with OVA. Ctrls received the vehicle alone and were not challenged with topical OVA. Each group comprised five guinea pigs, and both eyes were evaluated (n=10). *P<0.05; § P<0.01 versus OVA + vehicle; # P<0.01 versus Ctrl one-way ANOVA with Newman-Keuls post test.
Abbreviations: Ctrl, control; Dex, dexamethasone; EPO, eosinophil peroxidase; Map, mapracorat; OVA, ovalbumin.
Figure 4 Map and Dex eye drops reduce eosinophil infiltration by OVA in guinea pig conjunctiva.

Mapracorat and dexamethasone eye drops increase conjunctival eosinophil apoptotic cells in late-phase allergic reaction in guinea pigs

OVA challenge did not cause any significant elevation of apoptotic, TUNEL-positive cells lying scattered in the conjunctival tissue in comparison to controls at a 24-hour time point (). Interestingly, we observed that mapracorat and dexamethasone, administered 2 hours after OVA, were effective in increasing, in a dose-related manner, the number of apoptotic TUNEL-positive cells in guinea pig conjunctiva in comparison to OVA-treated animals. Mapracorat eye drops caused a more marked conjunctival cell apoptosis compared to the same dose of dexamethasone ().

Figure 5 Map and Dex eye drops increase, in a dose-related manner, the number of positive apoptotic cells evaluated by the TUNEL technique in tarsal conjunctival sections prepared from guinea pigs sacrificed 24 hours after OVA challenge.

Notes: Ctrls received the vehicle alone and were not treated with topical OVA. (A) Representative micrographs. Apoptotic cells appear rounded and bright green. Nuclei were counterstained with DAPI. (B) Values are the mean ± standard error of the mean (n=10; both eyes of five guinea pigs were evaluated). *P<0.05, § P<0.01 versus OVA + vehicle; # P<0.01 versus Dex at the same dose (one-way ANOVA with Newman-Keuls post test). Scale bar equals 50 μm.
Abbreviations: Ctrl, control; DAPI, 4′,6-diamidino-2-phenylindole; Dex, dexamethasone; Map, mapracorat; OVA, ovalbumin; TUNEL, terminal deoxynu cleotidyl transferase dUTP nick end labeling.
Figure 5 Map and Dex eye drops increase, in a dose-related manner, the number of positive apoptotic cells evaluated by the TUNEL technique in tarsal conjunctival sections prepared from guinea pigs sacrificed 24 hours after OVA challenge.

To characterize the nature of apoptotic cells detected in conjunctival specimens, we adopted a combined staining with TUNEL and chromotrope-2R that allows identification of tissue apoptotic eosinophils. As shown in , histological analysis performed in guinea pigs sacrificed 24 hours after OVA challenge showed numerous eosinophils, evidenced as red cells by chromotrope-2R staining, infiltrating the tarsal conjunctiva. Under these conditions, only very rare apoptotic, non-eosinophilic cells (stained as green cells) were evidenced by the TUNEL procedure. In fact, adopting this procedure, TUNEL- and chromotrope-2R-positive apoptotic eosinophils appear as yellow cells. The number of apoptotic eosinophils was increased, in a dose-related manner, in mapracorat- or dexamethasone-treated guinea pig conjunctiva as compared to OVA-treated animals. TUNEL-positive green cells when counterstained with chromotrope-2R appear as yellow cells. Mapracorat was more effective than dexamethasone in increasing the number of apoptotic cells (). No TUNEL staining was detected in control conjunctival sections or in absence of the deoxynucleotidyl transferase enzyme (data not shown). Thus, both glucocorticoid receptor agonists induce, 24 hours after OVA challenge, a significant apoptosis of conjunctival eosinophil.

Figure 6 Map and Dex eye drops increase, in a dose-related manner, the number of positive apoptotic eosinophils detected by TUNEL staining and chromotrope-2R counterstaining, in tarsal conjunctival sections prepared from guinea pigs sacrificed 24 hours after OVA challenge.

Notes: Ctrls received the vehicle alone and were not treated with topical OVA. Each group comprised five guinea pigs and both eyes were evaluated (n=10). Representative micrographs, in which TUNEL-positive cells appear as distinct, rounded, bright green nuclei and chromotrope-2R-positive cells appear as red spots, are shown. Colocalization of TUNEL and chromotrope-2R staining originates yellow cells (insets show enlargements). Scale bar equals 50 μm.
Abbreviations: Ctrl, control; Dex, dexamethasone; Map, mapracorat; OVA, ovalbumin; TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling.
Figure 6 Map and Dex eye drops increase, in a dose-related manner, the number of positive apoptotic eosinophils detected by TUNEL staining and chromotrope-2R counterstaining, in tarsal conjunctival sections prepared from guinea pigs sacrificed 24 hours after OVA challenge.

As previously stated in this paper, late-phase allergic reaction is characterized by conjunctival eosinophil infiltration that starts 4 hours after allergen challenge. To investigate if apoptotic eosinophils are detected in the conjunctiva as early as 8 hours after OVA challenge, we performed a separate experiment in which we observed that mapracorat or dexamethasone (0.4%) did not elevate the number of TUNEL-positive cells in conjunctival specimens of OVA-challenged guinea pigs sacrificed 6 hours after drug treatment (8 hours after OVA), although a significant number of infiltrating chromotrope-2R-positive eosinophils was evidenced (). Thus, eosinophil apoptosis induced by glucocorticoid receptor agonists is a late event that has been detected 22 hours after drug treatment and has also been observed in human eosinophils cultured in vitro for at least 24 hours.Citation18

Figure 7 Map and Dex (0.4%; w/v) eye drops do not cause any apparent increase in the number of positive apoptotic eosinophils detected by TUNEL procedure in tarsal conjunctival sections prepared from guinea pigs sacrificed 6 hours after Map and Dex treatment (8 hours after OVA challenge).

Notes: Contrary to apoptotic eosinophils, non-apoptotic eosinophils infiltrating the conjunctiva, evidenced by chromotrope-2R, were increased by OVA challenge and reduced by Map and Dex. Ctrls were not treated with OVA. Representative micrographs, in which TUNEL-positive cells appear as distinct, rounded, bright green nuclei and chromotrope-2R-positive cells appear as red spots, are shown. Each group comprised five guinea pigs, and both eyes were evaluated (n=10).
Abbreviations: Ctrl, control; Dex, dexamethasone; Map, mapracorat; OVA, ovalbumin; TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling.
Figure 7 Map and Dex (0.4%; w/v) eye drops do not cause any apparent increase in the number of positive apoptotic eosinophils detected by TUNEL procedure in tarsal conjunctival sections prepared from guinea pigs sacrificed 6 hours after Map and Dex treatment (8 hours after OVA challenge).

Mapracorat and dexamethasone eye drops reduce conjunctival chemokines and cytokines mRNA levels and CCL5 and CCL11 protein expression

OVA challenge induced a significant elevation of mRNA levels and protein expression of CCL5 and CCL11 in tarsal conjunctival specimens after 24 hours. Mapracorat and dexamethasone, administered 2 hours after OVA, were equally effective in reducing, in a dose-related manner, conjunctival mRNA levels () and protein expression () of both chemokines.

Figure 8 Map and Dex eye drops, administered 2 hours after antigen challenge, reduce conjunctival CCL5 (A), CCL11 (B), IL-1β (C), IL-8 (D), and TNF-α (E) mRNA levels in guinea pigs sacrificed 24 hours after OVA challenge.

Notes: Ctrls received the vehicle alone and were not treated with topical OVA. mRNA levels were measured by real time polymerase chain reaction using GAPDH as internal control. Values are the mean ± standard error of the mean (n=10; samples of both eyes of five guinea pigs were assayed). *P<0.05; § P<0.01 versus OVA + vehicle; # P<0.01 versus Ctrl (one-way ANOVA with Newman-Keuls post test).
Abbreviations: CCL5, chemokine C-C motif ligand 5; CCL11, chemokine C-C motif ligand 11; Ctrl, control; Dex, dexamethasone; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; IL-1β, interleukin-1β; IL-8, interleukin-8; Map, mapracorat; OVA, ovalbumin; TNF-α, tumor necrosis factor-α.
Figure 8 Map and Dex eye drops, administered 2 hours after antigen challenge, reduce conjunctival CCL5 (A), CCL11 (B), IL-1β (C), IL-8 (D), and TNF-α (E) mRNA levels in guinea pigs sacrificed 24 hours after OVA challenge.

Figure 9 Map and Dex eye drops, administered 2 hours after antigen challenge, reduce conjunctival CCL5 and CCL11 protein levels in guinea pigs sacrificed 24 hours after OVA challenge.

Notes: Ctrls received the vehicle alone and were not treated with topical OVA. (A and B) Representative conjunctival samples showing, respectively, the detection of CCL5 and CCL11 by immunohistochemistry. (C and D) Immunohistochemical scores were assigned according to five categories (0: no staining; 1: 25% staining; 2: 25%–50% staining: 3, 50%–75% staining; and 4: >75% staining). Median values ± standard error of the mean are reported (n=10; samples of both eyes of five guinea pigs/group were assayed). *P<0.05; § P<0.01 versus OVA + vehicle; # P<0.01 versus Ctrl (Dunn’s post hoc test after Kruskal–Wallis).
Abbreviations: CCL5, chemokine C-C motif ligand 5; CCL11, chemokine C-C motif ligand 11; Ctrl, control; Dex, dexamethasone; IHC, immunohistochemistry; Map, mapracorat; OVA, ovalbumin.
Figure 9 Map and Dex eye drops, administered 2 hours after antigen challenge, reduce conjunctival CCL5 and CCL11 protein levels in guinea pigs sacrificed 24 hours after OVA challenge.

Furthermore, both glucocorticoid agonists induced a dose-dependent decrease of conjunctival mRNA levels of IL-1β, IL-8, and TNF-α ().

Discussion

Mapracorat is a selective glucocorticoid receptor agonist with demonstrated anti-inflammatory properties for the topical treatment of inflammatory conditions.Citation13,Citation16 Previously, we reported that this agent inhibits the release of cytokines and chemokines from human mast cells.Citation18 In agreement with our findings, Zhang et alCitation43 and Cavet et alCitation19,Citation20 reported that mapracorat may act, with similar activity and potency as dexamethasone, to prevent the release of cytokines and chemokines in various primary human ocular cells, including conjunctival epithelial cells and fibroblasts. These effects help to explain the action of mapracorat in reducing the conjunctival symptoms that we preliminarily observed in OVA-sensitized guinea pigs by administering mapracorat before allergen exposure and thus showing its activity in preventing the early phase of ocular allergy.Citation18 In this former modelCitation18, a prophylactic treatment of mapracorat was effective in alleviating clinical signs of conjunctivitis as it may act upstream, preventing mast cell degranulation observed in the initial allergy cascade. Conversely, in the present study, topical mapracorat was administered after the induction of allergic conjunctivitis, when mast cells were already degranulated, with the aim of investigating its activity in the late-phase ocular reaction. We found that mapracorat, as well as dexamethasone, reduce conjunctival eosinophil accumulation and promote their apoptosis. We focused on eosinophils since these cells mediate unique cytotoxic and inflammatory effects and are crucial for the development of allergic disorders, including conjunctivitis.Citation4 Besides selective migration, longer cell survival and decreased apoptosis are relevant to tissue-specific accumulation of these inflammatory cells.Citation29

Glucocorticoids are the most effective anti-inflammatory drugs used to treat eosinophil disorders as they can prevent eosinophil accumulation and activation. In vitro models have suggested that glucocorticoids mediate their effects on eosinophil activity by an induction of eosinophil apoptosis;Citation27,Citation44,Citation45 however, the in vivo therapeutic relevance of such explanations is still questionable. Apoptotic eosinophils have previously been observed in parasite-infected and glucocorticoid-treated rat gutCitation45 whereas, as regards airway tissue, several studies have reported that glucocorticoids resolve established eosinophilic inflammation in vivo without inducing any detectable apoptosis of tissue eosinophils.Citation31,Citation38,Citation39,Citation46 These cited authors have proposed that eosinophils could efficiently be eliminated by egression into the airway lumen, where they may undergo apoptosis much more frequently than cells resident in tissue compartments. Interestingly, adopting the same histological procedures developed by Uller et al and Uller et al to ascertain any significant apoptosis in airway tissue,Citation31,Citation38 we show here, for the first time, that mapracorat and dexamethasone may cause a dose-dependent increase of eosinophil apoptosis in the conjunctiva. In agreement with our previous study carried out in vitro on human eosinophils,Citation18 we confirm that mapracorat is more effective than dexamethasone in increasing spontaneous eosinophil apoptosis. This unique profile might be due to differences in mapracorat versus glucocorticoids such as dexamethasone in the binding to the glucocorticoid receptor, which leads to a change in receptor conformation and could induce differential binding with other cofactors and/or with glucocorticoid recognition elements residing in the promoter of target genes.Citation47

Despite only one single glucocorticoid receptor gene having been discovered to date, several receptor isoforms may be generated via alternative splicing or via alternative translation initiation mechanisms.Citation48 These isoforms have distinct tissue distribution patterns and are capable of regulating unique sets of genes and mediating glucocorticoid-induced apoptosis in distinct manners.Citation49 For instance, the glucocorticoid receptor-C3 isoform induces the expression of proapoptotic molecules more efficiently than the D3 isoform.Citation50 Therefore, it should be considered that mapracorat may induce apoptosis more efficiently than dexamethasone by binding to certain glucocorticoid receptor isoforms expressed in eosinophils. In addition, it has been reported that components of cytoskeleton may facilitate glucocorticoid-induced apoptosis.Citation50 Thus, mapracorat could also influence these events more effectively that dexamethasone.

The reduced eosinophilia in mapracorat- and dexamethasone-treated groups may, in part, reflect a reduced recruitment of these cells to the conjunctiva. In fact, we noted that both glucocorticoid receptor agonists significantly suppress eosinophil accumulation in tear fluid collected 4–8 hours after allergen challenge when eosinophil apoptosis was not yet detected. Our findings, showing a reduced, dose-dependent reduction in mRNA and protein expression of CCL5 and CCL11 and in mRNA levels of IL-1β, IL-8, and TNF-α, suggest that these are related to the mapracorat- or dexamethasone-mediated reduction of conjunctival eosinophilia and confirm that these chemokines and cytokines may represent useful markers with which to monitor the anti-allergic efficacy of mapracorat and other glucocorticoids. Interestingly, CCL5 and CCL11 have been pointed out as two major chemokines elevated in the conjunctiva of patients with vernal keratoconjunctivitis.Citation24,Citation26

Development of therapeutic glucocorticoid receptor agonists with full anti-inflammatory properties and reduced potential for side effects is an active area of research.Citation9,Citation11,Citation51 Transrepressive mechanisms are thought to account for the majority of anti-inflammatory effects mediated through the glucocorticoid receptor; however, recent studies highlight, in some cell types and inflammatory conditions, the necessity for induction of anti-inflammatory properties for maximal therapeutic effects.Citation52 Induction of these anti-inflammatory proteins likely involves transactivation, whether this is via glucocorticoid responsive element binding, more complex tethering, or composite actions incorporating other transcription factors, remains to be elucidated. Therefore, it is essential to verify the anti-inflammatory activity of novel glucocorticoid receptor ligands in vivo in models, such as the one adopted in this study. As reported,Citation9Citation11 it would be better to search for “differential” compounds that show the most favorable functional profiles than for glucocorticoid ligands that distinguish transrepression and transactivation.

Clinical interest for mapracorat is also supported by several clinical trials completed in the last few years testing for allergic conjunctivitis,Citation53 dry eye syndrome,Citation54 and the treatment of ocular inflammation and pain following cataract surgery.Citation55 Trial results, which are not yet released by Bausch & Lomb, will be fundamental to confirm mapracorat’s therapeutic effects in clinical conditions involving ocular inflammation.

Conclusion

We confirm that mapracorat is a promising candidate for the topical treatment of allergic eye disorders. It easily reaches conjunctival tissue when administered topically,Citation15 and some of its cellular targets may contribute to eosinophil apoptosis and/or to prevention of eosinophil recruitment and activation, in addition to inhibiting the release of cytokines and chemokines.

Acknowledgments

This study was supported by a grant from the University of Bologna, Bologna, Italy (Ricerca Fondamentale Orientata)

Disclosure

The authors report no conflicts of interest in this work. Megan E Cavet is an employee of Bausch & Lomb. Bausch & Lomb did not participate in the study design; the collection, analysis, and interpretation of data; the writing of the report; or the decision to submit this paper for publication.

References

  • Leonardi A Motterle L Bortolotti M Allergy and the eye Clin Exp Immunol 2008 153 Suppl 1 17 21 18721324
  • Bielory BP O’Brien TP Bielory L Management of seasonal allergic conjunctivitis: guide to therapy Acta Ophthalmol 2012 90 399 407 22067457
  • Guglielmetti S Dart JK Calder V Atopic keratoconjunctivitis and atopic dermatitis Curr Opin Allergy Clin Immunol 2010 10 478 485 20720488
  • Irkec MT Bozkurt B Molecular immunology of allergic conjunctivitis Curr Opin Allergy Clin Immunol 2012 12 534 539 22885886
  • Chauhan S Leach CH Kunz S Bloom JW Miesfeld RL Glucocorticoid regulation of human eosinophil gene expression J Steroid Biochem Mol Biol 2003 84 441 452 12732289
  • Bielory BP Perez VL Bielory L Treatment of seasonal allergic conjunctivitis with ophthalmic corticosteroids: in search of the perfect ocular corticosteroids in the treatment of allergic conjunctivitis Curr Opin Allergy Clin Immunol 2010 10 469 477 20720489
  • De Bosscher K Haegeman G Elewaut D Targeting inflammation using selective glucocorticoid receptor modulators Curr Opin Pharmacol 2010 10 497 504 20493772
  • Ratman D Vanden Berghe W Dejager L How glucocorticoid receptors modulate the activity of other transcription factors: a scope beyond tethering Mol Cell Endocrinol 2013 380 41 54 23267834
  • Newton R Holden NS Separating transrepression and transactivation: a distressing divorce for the glucocorticoid receptor? Mol Pharmacol 2007 72 799 809 17622575
  • Clark AR Belvisi MG Maps and legends: the quest for dissociated ligands of the glucocorticoid receptor Pharmacol Ther 2012 134 54 67 22212616
  • Newton R Anti-inflammatory glucocorticoids: changing concepts Eur J Pharmacol 2014 724 231 236 23747654
  • De Bosscher K Selective glucocorticoid receptor modulators J Steroid Biochem Mol Biol 2010 120 96 104 20206690
  • Schäcke H Zollner TM Döcke WD Characterization of ZK 245186, a novel, selective glucocorticoid receptor agonist for the topical treatment of inflammatory skin diseases Br J Pharmacol 2009 158 1088 1103 19422381
  • Bodor N Buchwald P Corticosteroid design for the treatment of asthma: structural insights and the therapeutic potential of soft corticosteroids Curr Pharm Des 2006 12 25 3241 3260 17020532
  • Proksch JW Lowe ER Ward KW Ocular pharmacokinetics of mapracorat, a novel selective glucocorticoid receptor agonist, in rabbits and monkeys Drug Metab Dispos 2011 39 1181 1187 21441467
  • Shafiee A Bucolo C Budzynski E Ward KW López FJ In vivo ocular efficacy profile of mapracorat, a novel selective glucocorticoid receptor agonist, in rabbit models of ocular disease Invest Ophthalmol Vis Sci 2011 52 1422 1430 20881303
  • Pfeffer BA DeWitt CA Salvador-Silva M Cavet ME López FJ Ward KW Reduced myocilin expression in cultured monkey trabecular meshwork cells induced by a selective glucocorticoid receptor agonist: comparison with steroids Invest Ophthalmol Vis Sci 2010 51 437 446 19696178
  • Baiula M Spartà A Bedini A Eosinophil as a cellular target of the ocular anti-allergic action of mapracorat, a novel selective glucocorticoid receptor agonist Mol Vis 2011 17 3208 3223 22194647
  • Cavet ME Volhejn S Harrington KL Zhang JZ Anti-allergic effects of mapracorat, a novel selective glucocorticoid receptor agonist, in human conjunctival fibroblasts and epithelial cells Mol Vis 2013 19 1515 1525 23878502
  • Cavet ME Harrington KL Ward KW Zhang JZ Mapracorat, a novel selective glucocorticoid receptor agonist, inhibits hyperosmolar-induced cytokine release and MAPK pathways in human corneal epithelial cells Mol Vis 2010 16 1791 1800 20824100
  • Chambless SL Trocme S Developments in ocular allergy Curr Opin Allergy Clin Immunol 2004 4 431 434 15349044
  • Bisset LR Schmid-Grendelmeier P Chemokines and their receptors in the pathogenesis of allergic asthma: progress and perspective Curr Opin Pulm Med 2005 11 35 42 15591886
  • Abu El-Asrar AM Struyf S Al-Kharashi SA Missotten L Van Damme J Geboes K Chemokines in the limbal form of vernal keratoconjunctivitis Br J Ophthalmol 2000 84 1360 1366 11090473
  • Leonardi A Jose PJ Zhan H Calder VL Tear and mucus eotaxin-1 and eotaxin-2 in allergic keratoconjunctivitis Ophthalmology 2003 110 487 492 12623809
  • Kvarnhammar AM Cardell LO Pattern-recognition receptors in human eosinophils Immunology 2012 136 1 11 20 22242941
  • Duffin R Leitch AE Fox S Haslett C Rossi AG Targeting granulocyte apoptosis: mechanisms, models, and therapies Immunol Rev 2010 236 28 40 20636806
  • McColl A Michlewska S Dransfield I Rossi AG Effects of glucocorticoids on apoptosis and clearance of apoptotic cells ScientificWorldJournal 2007 7 1165 1181 17704849
  • Druilhe A Létuvé S Pretolani M Glucocorticoid-induced apoptosis in human eosinophils: mechanisms of action Apoptosis 2003 8 481 495 12975579
  • Baiula M Bedini A Carbonari G Dattoli SD Spampinato S Therapeutic targeting of eosinophil adhesion and accumulation in allergic conjunctivitis Front Pharmacol 2012 3 203 23271999
  • Woolley KL Gibson PG Carty K Wilson AJ Twaddell SH Woolley MJ Eosinophil apoptosis and the resolution of airway inflammation in asthma Am J Respir Crit Care Med 1996 154 237 243 8680686
  • Uller L Emanuelsson CA Andersson M Erjefält JS Greiff L Persson CG Early phase resolution of mucosal eosinophilic inflammation in allergic rhinitis Respir Res 2010 11 54 20459697
  • Uller L Persson CG Erjefält JS Resolution of airway disease: removal of inflammatory cells through apoptosis, egression or both? Trends Pharmacol Sci 2006 27 461 466 16876880
  • Qasem AR Bucolo C Baiula M Contribution of alpha4beta1 integrin to the antiallergic effect of levocabastine Biochem Pharmacol 2008 76 751 762 18680729
  • Kato M Imoto K Miyake H Oda T Miyaji S Nakamura M Apafant, a potent platelet-activating factor antagonist, blocks eosinophil activation and is effective in the chronic phase of experimental allergic conjunctivitis in guinea pigs J Pharmacol Sci 2004 95 435 442 15286429
  • Walker RA Quantification of immunohistochemistry – issues concerning methods, utility and semiquantitative assessment I Histopathology 2006 49 406 410 16978204
  • Kelly MM King EM Rider CF Corticosteroid-induced gene expression in allergen-challenged asthmatic subjects taking inhaled budesonide Br J Pharmacol 2012 165 1737 1747 21827450
  • Gavrieli Y Sherman Y Ben-Sasson SA Identification of programmed cell death in situ via specific labeling of nuclear DNA fragmentation J Cell Biol 1992 119 493 501 1400587
  • Uller L Andersson M Greiff L Persson CG Erjefält JS Occurrence of apoptosis, secondary necrosis, and cytolysis in eosinophilic nasal polyps Am J Respir Crit Care Med 2004 170 742 747 15229095
  • Uller L Lloyd CM Rydell-Törmänen K Persson CG Erjefält JS Effects of steroid treatment on lung CC chemokines, apoptosis and transepithelial cell clearance during development and resolution of allergic airway inflammation Clin Exp Allergy 2006 36 111 121 16393273
  • Kubo S Kobayashi M Masunaga Y Cytokine and chemokine expression in cigarette smoke-induced lung injury in guinea pigs Eur Respir J 2005 26 993 1001 16319327
  • Lacy HM Bowlin AK Hennings L Scurlock AM Nagarajan UM Rank RG Essential role for neutrophils in pathogenesis and adaptive immunity in Chlamydia caviae ocular infections Infect Immun 2011 79 1889 1897 21402767
  • Choi SH Bielory L Late-phase reaction in ocular allergy Curr Opin Allergy Clin Immunol 2008 8 438 444 18769198
  • Zhang JZ Cavet ME VanderMeid KR Salvador-Silva M López FJ Ward KW BOL-303242-X, a novel selective glucocorticoid receptor agonist, with full anti-inflammatory properties in human ocular cells Mol Vis 2009 15 2606 2616 20011631
  • Zhang X Moilanen E Kankaanranta H Enhancement of human eosinophil apoptosis by fluticasone propionate, budesonide, and beclomethasone Eur J Pharmacol 2000 406 325 332 11040338
  • Kawabori S Soda K Perdue MH Bienenstock J The dynamics of intestinal eosinophil depletion in rats treated with dexamethasone Lab Invest 1991 64 224 233 1997734
  • Uller L Persson CG Källström L Erjefält JS Lung tissue eosinophils may be cleared through luminal entry rather than apoptosis: effects of steroid treatment Am J Respir Crit Care Med 2001 164 1948 1956 11734451
  • Barnes PJ Glucocorticosteroids: current and future directions Br J Pharmacol 2011 163 29 43 21198556
  • Lu NZ Cidlowski JA Translational regulatory mechanisms generate N-terminal glucocorticoid receptor isoforms with unique transcriptional target genes Mol Cell 2005 18 331 342 15866175
  • Lu NZ Collins JB Grissom SF Cidlowski JA Selective regulation of bone cell apoptosis by translational isoforms of the glucocorticoid receptor Mol Cell Biol 2007 27 7143 7160 17682054
  • Wu I Shin SC Cao Y Selective glucocorticoid receptor translational isoforms reveal glucocorticoid-induced apoptotic transcriptomes Cell Death Dis 2013 4 e453 23303127
  • Catley M Dissociated steroids ScientificWorldJournal 2007 7 421 430 17450306
  • Clark AR Anti-inflammatory functions of glucocorticoid-induced genes Mol Cell Endocrinol 2007 275 79 97 17561338
  • Bausch & Lomb Incorporated Mapracorat Ophthalmic Formulation in Subjects With Allergic Conjunctivitis Available from: http://www.clinicaltrials.gov/ct2/show/NCT01289431?term=mapracorat&rank=6. NLM identifier: NCT01289431 Accessed March 20, 2014
  • Bausch & Lomb Incorporated Study to Assess the Safety and Efficacy of BOL-303242-X Ophthalmic Suspension in Dry Eye Syndrome Available from: http://www.clinicaltrials.gov/ct2/show/NCT01163643?term=nct01163643&rank=1. NLM identifier: NCT01163643 Accessed March 20, 2014
  • Bausch & Lomb Incorporated Mapracorat Ophthalmic Suspension, 3% for the Treatment of Ocular Inflammation and Pain Following Cataract Surgery Available from: http://www.clinicaltrials.gov/ct2/show/NCT01591161?term=mapracorat&rank=3. NLM identifier: NCT01591161 Accessed March 20, 2014