Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Nov 1.
Published in final edited form as: Exp Eye Res. 2016 Sep 4;152:10–33. doi: 10.1016/j.exer.2016.08.020

Glucocorticoid action in human corneal epithelial cells establishes roles for corticosteroids in wound healing and barrier function of the eye

Mahita Kadmiel a, Agnes Janoshazi a, Xiaojiang Xu b, John A Cidlowski a,*
PMCID: PMC5097880  NIHMSID: NIHMS818507  PMID: 27600171

Abstract

Glucocorticoids play diverse roles in almost all physiological systems of the body, including both anti-inflammatory and immunosuppressive roles. Synthetic glucocorticoids are one of the most widely prescribed drugs and are used in the treatment of conditions such as autoimmune diseases, allergies, ocular disorders and certain types of cancers. In the interest of investigating glucocorticoid actions in the cornea of the eye, we established that multiple cell types in mouse corneas express functional glucocorticoid receptor (GR) with corneal epithelial cells having robust expression. To define glucocorticoid actions in a cell type-specific manner, we employed immortalized human corneal epithelial (HCE) cell line to define the glucocorticoid transcriptome and elucidated its functions in corneal epithelial cells. Over 4000 genes were significantly regulated within 6 hours of dexamethasone treatment, and genes associated with cell movement, cytoskeletal remodeling and permeability were highly regulated. Real-time in vitro wound healing assays revealed that glucocorticoids delay wound healing by attenuating cell migration. These functional alterations were associated with cytoskeletal remodeling at the wounded edge of a scratch-wounded monolayer. However, glucocorticoid treatment improved the organization of tight-junction proteins and enhanced the epithelial barrier function. Our results demonstrate that glucocorticoids profoundly alter corneal epithelial gene expression and many of these changes likely impact both wound healing and epithelial cell barrier function.

Keywords: Cornea, glucocorticoids, gene expression, wound healing, migration, cytoskeleton, epithelial integrity

1. Introduction

Glucocorticoids are steroid hormones that have a critical role in regulating stress response in the body. Endogenous glucocorticoids in humans are necessary for life and they are synthesized by the adrenal cortex in a tight regulation by the hypothalamic-pituitary-adrenal axis. Both endogenous glucocorticoids and their synthetic derivatives used in patient treatment signal through their canonical receptor, the glucocorticoid receptor (gene name NR3C1) that belongs to the super family of nuclear receptors. Glucocorticoid actions span a wide range of cellular and systemic effects including cell cycle, cell movement, glucose homeostasis and fluid regulation. They are most known for their anti-inflammatory and immunosuppressive roles. Due to their potent immunosuppressive property, glucocorticoids have been exploited pharmacologically and they have become one of the largest selling class of drugs in the world today. The nearly ubiquitous expression of the glucocorticoid receptor suggests a role for glucocorticoid signaling in every cell type, which is further supported by studies establishing that glucocorticoid signaling is indeed cell type-specific. For example, glucocorticoids exert an anti-apoptotic role in cardiomyocytes (1) while exerting a pro-apoptotic role in lymphocytes (2). Cell type specificity of glucocorticoid signaling diversifies the actions of glucocorticoids and therefore, there is a need to understand the role of glucocorticoids in a cell/tissue-specific manner.

The cornea is the clear part of the eye that covers the iris, pupil and the anterior chamber. By providing a physical barrier, the cornea protects the interior of the eye from external agents such as bacteria, viruses and debris. By refracting light through the lens and onto the retina where the light signal converts into vision, the cornea also plays an important role in maintaining vision. Synthetic glucocorticoids have been widely used to successfully treat several ocular disorders, however, the functions of glucocorticoid receptor signaling in the eye, particularly in the cornea are largely under studied. Corticosteroids are also used in transplant surgeries such as in lens transplantation and keratoplasty, to minimize graft rejection. Corticosteroids are used in treating sight-threatening conditions of the cornea such as corneal inflammation and corneal neovascularization (3). Despite the fact that glucocorticoids have numerous benefits in treating ocular conditions, some patients receiving chronic glucocorticoid treatment are susceptible to increase in intraocular pressure that could develop into steroid-induced glaucoma and eventually loss of vision (4). Opacification of the lens or cataract formation are also adverse events seen in sustained corticosteroid use. Glucocorticoids have also been reported to be synthesized in the human ocular surface and they have the ability to regulate corneal immune response (5-7). In the cornea, glucocorticoids have been shown to regulate their circadian rhythm (8,9), inhibit blood and lymphatic vessel growth (10,11), curb inflammation (12-15), and increase epithelial integrity under a hypoxic challenge (16), as well as retard wound healing in rabbits (17). Although it has been established that corticosteroids are effective in treating diseases of the cornea, the molecular functions in specific cell types where they occur have not been fully characterized.

In the current investigation, we establish that mouse corneas express functional GR with strong expression of GR by the corneal epithelial cells. Subsequently, we employed immortalized corneal epithelial cell line derived from human cornea to understand glucocorticoid signaling in a single cell type. Here we demonstrate that glucocorticoids can profoundly alter the gene expression profile of human corneal epithelial cells. Ingenuity pathway analysis (IPA) of the glucocorticoid transcriptome revealed that glucocorticoid signaling in corneal epithelial cells was enriched for genes involved in pathways associated with inflammatory diseases and organismal growth and development. Additionally, Ingenuity Pathway Analysis indicated that glucocorticoid signaling in corneal epithelial cells may regulate cellular functions, such as cell movement and cell growth, by altering the expression of a large cohort of genes. Since cornea is at the interface with the environment and prone to injuries, we focused our further analysis of glucocorticoid signaling in corneal epithelial cells on wound healing which included processes such as cell migration, cytoskeletal remodeling and epithelial permeability. Real time in vitro wound healing assays demonstrated that glucocorticoid treatment delayed wound healing of HCE cell monolayer by altering their cytoskeleton. Interestingly, the distribution of tight junction proteins and paracellular permeability in response to glucocorticoid treatment indicated that glucocorticoids enhance barrier function in corneal epithelial cells. The study presented here provides a new understanding of the diversity of glucocorticoid actions on corneal epithelial cell wound healing and barrier function.

2. Materials and methods

2.1 Animals

Wild type C57BL/6 female mice aged 2-months old purchased from Charles River Laboratories were used for all animal experiments. For dexamethasone treatment studies, mice were adrenalectomized at Charles River Laboratories to remove endogenous glucocorticoids and were rested for a week after the surgery before being shipped to the National Institute of Environmental Health Sciences (NIEHS). Upon arrival at NIEHS, the animals were rested for 7-10 days before being treated. For dexamethasone treatment experiment, each mouse was treated with vehicle in the left eye and dexamethasone in the right eye. Dexamethasone was purchased from Steraloids and was prepared in Refresh artificial tears manufactured by Allergan, Irvine, CA. For each animal, one eye received 3 microliters of vehicle (Refresh artificial tears) or dexamethasone prepared at a concentration of 1mg/ml. Six hours after the treatment, mice were euthanized by cervical dislocation and eyes were enucleated and corneas were dissected immediately and stored in RNA later (Qiagen) at 4°C overnight. Six corneas were pooled to generate one sample of RNA, therefore, requiring 24 corneas/treatment to generate an n of 4. RNA was extracted using Trizol and chloroform and purified using RNeasy Micro kit and Dnase digested (Qiagen). For immunofluorescence studies, mice were euthanized by cervical dislocation and eyes were enucleated from euthanized animals. Eyes were fresh frozen in Optimal Cutting Temperature (O.C.T.) Compound (VWR, Pennsylvania) and six-micron sections were prepared. Sections were stained at 4°C overnight wi th antibodies to glucocorticoid receptor (Cell Signaling, cat#3660, 1:300). Hoechst 33342 and Alexa Fluor 488 Phalloidin (both from Life Technologies, New York) were used to visualize nuclei and actin filaments, respectively. Z-stack images were taken using the Zeiss LSM710 and Zen 2012 software and Image J software were used to process the images.

2.2 Cell culture and treatment

A widely studied immortalized human corneal epithelial cell line (HCE) obtained from RIKEN was used (18). HCE cells were cultured in DMEM/F12 medium supplemented with 5% fetal bovine serum, 5ug/ml insulin, 10ng/ml human epidermal growth factor, 0.5% dimethyl sulfoxide and antibiotics. Anti-glucocorticoid-RU486 (mifepristone) were purchased from Steraloids. Cells were incubated in DMEM/F12 medium containing 5% charcoal stripped fetal bovine serum for 18-24hours before being treated with vehicle or dexamethasone or RU486.

2.3 RNA Isolation and Quantitative RT-PCR Analysis

Total RNA was isolated using the RNeasy Kit (micro kit for Trizol/Chloroform extracted mouse corneal RNA and mini kit for human cells) and DNase digested using the RNase-Free DNase Kit (Qiagen) according to the manufacturer’s protocol. The abundance of individual mRNAs was determined using a Taqman one-step RT-PCR method on a 7900HT sequence detection system (Applied Biosystems). Pre-developed Taqman primer probe sets for GILZ (Hs00608272_m1, Mm00726417_s1), FKBP5 (Mm00487406_m1), TNFRSF11b (Hs00900358_m1), BDNF (Hs00380947_m1), EREG (Hs00154995_m1), NGF (Hs00171458_m1) and PPIB (Hs00168719_m1, Mm00478295_m1) were purchased from Life Technologies, Grand Island, NY. Target gene expression was normalized to the housekeeping gene PPIB, which is not regulated by glucocorticoids.

2.4 SDS-PAGE and Immunoblot Analyses

Cells were washed with ice-cold phosphate buffered saline and lysed in SDS sample buffer (Life Technologies) supplemented with B-mercaptoethanol (final concentration 2.5%). Samples were sonicated on ice for 5 seconds and boiled for 5 mins and 104° centigrade. Equal amounts of protein was loaded and run on precast 10% Tris Mini Protean TGX gels (Bio-Rad) and transferred to nitrocellulose. The membranes were blocked for an hour in LI-COR Blocking buffer at room temperature and then incubated overnight at 4C with primary antibody to GR(19) (1:1000 dilution) or B-actin (Millipore, 1:20,000 dilution). Blots were washed and incubated with goat anti-rabbit IRDye 680-conjugated secondary antibody and/or goat anti-mouse IRDye800-conjugated secondary antibody (Rockland Immunochemicals) for one hour at room temperature. LICOR Odyssey scanner was used to visualize the western blot.

2.5 Microarray and data analysis

Global gene expression analysis was performed on RNA isolated from cells treated with vehicle or Dexamethasone (100nM) for 6 hours (n = 4 biological replicates/treatment). Specifically, gene expression analysis was conducted using Agilent Whole Human Genome 4×44 multiplex format oligo arrays (014850) (Agilent Technologies) following the Agilent 1-color microarray-based gene expression analysis protocol. Starting with 400ng of total RNA, Cy3 labeled cRNA was produced according to manufacturer’s protocol. For each sample, 1.65ug of Cy3 labeled cRNAs were fragmented and hybridized for 17 hours in a rotating hybridization oven. Slides were washed and then scanned with an Agilent Scanner. Data was obtained using the Agilent Feature Extraction software (v9.5), using the 1-color defaults for all parameters. The Agilent Feature Extraction Software performed error modeling, adjusting for additive and multiplicative noise.

Differential gene expression was examined using the Partek Genomics Suite V 6.6 (Partek, Inc., St. Louis, MO, USA). To identify differentially expressed probe sets, analysis of variance (ANOVA) was performed and significant changes in gene expression were defined on the basis of p-value (p < 0.01). Partek Genomics Suite was further used to generate heat maps for visual analyses and to support generation of hierarchical clustering dendrograms. Lists of significant probe sets were further analyzed using Ingenuity Pathway Analysis (IPA, Content version 27216297) (Ingenuity Systems, Redwood City, CA). Enrichment or overlap was determined by IPA using Fisher’s exact test (p < 0.05). Pathdesigner feature of IPA was used to build pathways of glucocorticoid-regulated genes. Gene network of genes involved in Cell Movement was extracted from STRING (Version10, http://string-db.org/) and Visualized using Gephi (Version 0.9.1).

2.6 In vitro wound healing assay

HCE cells were grown to 90% confluence in 12-well plates in DMEM/F12 medium containing 5% charcoal stripped bovine serum and antibiotics. The cells were then treated with vehicle or dexamethasone or RU486 or both in the same medium containing charcoal stripped fetal bovine serum. After treatment for 24hours, a scratch was made using a sterile 200ul yellow pipette tip in the middle of the confluent monolayer. The wells were washed with the respective treatment media to remove detached and dead cells. The wells were replaced with fresh medium containing the respective treatments. Three to five bright-field images were taken along the scratch area every thirty minutes for up to 30 hours (until the scratch wound is healed) using an incubator setup with a Zeiss LSM 710 confocal microscope using a low-magnification objective of 5X to cover a large area of the healing monolayer. Images were taken only in X and Y planes. Area of wound closure was measured using the following formula:

Percent wound closed=((Area of wound at 0hr- area of wound at 18hr)area of wound at 0hr100).

2.7 Lamellipodia and Filopodia Visualization and Quantification

HCE cells were grown to 90% confluence in glass-bottom dishes and in DMEM/F12 medium containing 5% charcoal stripped bovine serum and antibiotics. The cells were then treated with vehicle or dexamethasone (1000nM) in the same medium containing charcoal stripped medium. After treatment for 24hours, a scratch was made using a sterile 10ul pipette tip in the middle of the confluent monolayer. The wells were replaced with fresh medium containing the respective treatments. The media was discarded and the plates were washed with the respective treatment media to remove detached and dead cells. To visualize the cell membrane with lamellipodia and filopodia, CellMask™ Deep Red Plasma membrane Stain (Catalog # C10046) was added to the plates at 0.1% concentration in HCE culture medium immediately after creating a scratch wound in the monolayer and imaging at high-magnification (40X oil-immersion objective) was initiated within 10 minutes of making the scratch. Zeiss LSM 780 confocal microscope equipped with an incubator set at 37 degrees centigrade and 5% carbon dioxide was used. Z-stack images scanning a total depth of 6-10 microns (0.75 micron per Z-section) were taken continuously for an hour to visualize the dynamic lamellipodia and filopodia at the healing edge. Maximum intensity projection of a Z-stack of images was made to count the number of filopodia in each image. The number of filopodia on each of the three to five fields (each field had a scratched edge of 250 microns) were manually counted at different time-points starting from 10 minutes to 1 hour after creating a scratch wound and the average number of filopodia were represented.

2.8 Permeability Assays

HCE cells were plated grown to confluence in 12-well transwell plates (Corning Costar Transwell, 0.4uM pore size). Twenty-four hours prior to treatment, the medium was replaced with DMEM/F12 containing 5% charcoal stripped fetal bovine serum. Cells were treated with either vehicle or 100nM dexamethasone for 24 hours. At the end of incubation, FITC Dextran -10kD at a final concentration of 1mg/ml final concentration (purchased from Sigma) was carefully added to the medium in the insert and the plates were returned back to the incubator. After an hour of incubation, media was collected from the bottom wells and the relative units of fluorescence of FITC dextran diffused through the monolayer in the inserts were measured using a fluorescent plate reader.

2.9 Proliferation Assays

For proliferation assays, HCE cells were plated at a density of 8×105 cells per well in 6-well cell culture plates. Twenty-four hours after plating, cells were treated with vehicle or dexamethasone (100nM or 1000nM) in DMEM/F12 medium containing charcoal stripped serum. Trypsinized cells and dead floating cells in the supernatant from each well at all time-points (24, 48, and 72 hours post treatment) were counted with Countess Automated Cell Counter (Invitrogen) using chamber slides with a 1:1 dilution of cells to Trypan blue stain 0.4% (Invitrogen). Each sample was counted in duplicate. Average number of viable and dead cells were calculated from 4 independent experiments.

2.10 Flow Cytometric Analysis

Cell proliferation was assessed by flow cytometry. HCE cells were grown in 6-well cell culture plates and treated with vehicle or dexamethasone (100nM or 1000nM) for 24, 48 and 72 hours. After treatment, cells (including floating cells) were collected by trypsinization and propidium iodide was added to identify dead cells. Cells were excited using a 488-nm argon laser and emission was detected at 585 nm. Analysis was carried out using a Becton Dickinson FACSort flow cytometer (Franklin Lakes, NJ, USA) and CELLQuest software (Becton Dickinson Immunocytometry Systems, San Jose, CA, USA).

2.11 Statistical Analysis

The data are represented as mean+/− standard error of the mean. Unless indicated otherwise, a student’s t-test was performed to determine statistical significance of results. A p value of < 0.01 (**) or <0.05 (*) was considered statistically significant.

3. RESULTS

3.1 Glucocorticoid receptors in the mouse cornea

To determine if the glucocorticoid receptor is present in the adult mouse cornea, glucocorticoid receptor expression was examined in 2 month-old wild type female mice by immunofluorescence. Nuclei in all layers of the cornea- the corneal epithelium, the stroma and the endothelium are stained positive for the glucocorticoid receptor (Figure 1). This data suggests that the glucocorticoid receptor may play a role in regulating the function of the adult cornea. Particularly interesting was the robust expression of the glucocorticoid receptor in all the cells of the corneal epithelium. To establish the functionality of GR in the mouse cornea, adult wild type female mice were treated with vehicle or dexamethasone eye drops and 6 hours later Glucocorticoid-Induced Leucine Zipper (Gilz) and FK506 Binding protein 5 (Fkbp5), the two classical target genes of glucocorticoid receptor signaling were quantified by RT-PCR. Glucocorticoid treatment induced the expression of Gilz (Fig1B) and Fkbp5 (Fig1C) demonstrating the presence of an active glucocorticoid signaling system in the mouse cornea. In order to evaluate the function of the glucocorticoid receptor in the corneal epithelium, an immortalized human corneal epithelial cell line was utilized for subsequent studies.

Figure 1.

Figure 1

Glucocorticoid Receptor signaling in the adult mouse cornea. A) Immunofluorescence of wild type adult female mouse cornea showing glucocorticoid receptor expression (red) in all the layers of the corneal epithelium, in the corneal stromal cells and in the corneal endothelial cells. Phalloidin and Hoechst were used to visualize actin (green) and nuclei (blue), respectively. A merge of all three channels is presented in the fourth panel. Scale bar: 20µm. B) & C) Adrenalectomized wild type mice were treated for 6 hours with vehicle or dexamethasone eye drops and GILZ mRNA (B) and FKBP5 mRNA (C) were evaluated by quantitative RT-PCR. Results were normalized to PPIB gene expression (housekeeping gene). Data represent mean ± standard error of mean from four biological replicates. Student’s t-test was used to determine statistical significance compared to the vehicle-treated cells; *** p<0.001.

3.2 Human corneal epithelial cells express a functional glucocorticoid receptor signaling system

Glucocorticoid effects on human corneal epithelial cells have been previously reported for a few target genes (16,20,21), however, the genome wide actions of glucocorticoids have not been established. To address this issue, we performed western blotting in immortalized human corneal epithelial cell line to first determine if glucocorticoid receptor is expressed (18). Our studies demonstrate that glucocorticoid receptors are expressed by human corneal epithelial cells (Figure 2A). Subsequently, we elucidated the ability of the glucocorticoid receptor to undergo translocation to the nucleus following ligand binding. HCE cells treated either with vehicle or 100nM dexamethasone for an hour were fixed and immunofluorescence was performed. Glucocorticoid treatment resulted in translocation of glucocorticoid receptor to the nucleus, which was otherwise mostly cytoplasmic in location (Figure 2B). In order to establish the functionality of GR in these cells, we treated HCE cells with different doses of dexamethasone (0, 1, 10, 100 and 1000nM) for 6 hours, followed by RT-PCR for the expression of GILZ mRNA. Glucocorticoid treatment resulted in a dose-dependent induction of GILZ expression (Figure 2C). Glucocorticoid-induced upregulation of GILZ is mediated via the glucocorticoid receptor, because GILZ induction was inhibited in the presence of RU486- a glucocorticoid receptor antagonist (Figure 2D). These data indicate that transcriptionally active glucocorticoid receptor is expressed by HCE cells.

Figure 2.

Figure 2

Corneal epithelial cells express functional glucocorticoid receptors. A) Glucocorticoid receptor protein level expressed by immortalized human corneal epithelial (HCE) cells evaluated by immunoblot. Actin was used as the loading control. B) Nuclear translocation of glucocorticoid receptor by immunofluorescence in HCE cells treated with 100nM dexamethasone for 1 hour at 37 degrees centigrade. Glucocorticoid receptor expression is in red and Hoechst staining of the nuclei is in blue. C) HCE cells were treated with vehicle or four different concentrations of dexamethasone for 6hrs and GILZ mRNA was measure by quantitative RT-PCR. D) HCE cells were treated for 6 hours with vehicle, dexamethasone (100nM), RU486- an antagonist of glucocorticoid receptor (1000nM), or both and GILZ mRNA was evaluated by quantitative RT-PCR. Results were normalized to PPIB gene expression (housekeeping gene). Data represent mean ± standard error of mean from three or four independent experiments. Student’s t-test was used to determine statistical significance compared to the vehicle-treated cells; * p<0.05 and ** p<0.01.

3.3 Global gene expression changes induced by glucocorticoid treatment in HCE cells

Glucocorticoids are known to regulate numerous genes in a variety of tissues and cell types from rodents and humans (22-24), but very little is known about their genome wide actions in specific corneal cell types. For example, Liu et al have prepared primary corneal fibroblasts from male human donors and they found that very long-term dexamethasone treatment (16 hours) greatly altered both the gene and microRNA profiles in human corneal fibroblasts (25). To our knowledge, no such global gene expression studies have been performed on human corneal epithelial cells. Since cornea is a mixture of different cell types and because glucocorticoid regulation is known to work in a cell-type specific manner, we focused our studies on the human corneal epithelial cells. We performed whole-genome microarray on human corneal epithelial cells. HCE cells were treated with 100nM dexamethasone for only 6 hours. Glucocorticoids significantly altered the expression of 4439 genes expressed in HCE cells (Figure 3A). Of the significantly regulated genes, 2046 genes (about 46.1%) were upregulated, while 2393 genes (about 53.9%) were downregulated by glucocorticoid treatment (Figure 3B). Ingenuity Pathway Analysis software was used to elucidate the biological significance of the genetic signature elicited by dexamethasone-treatment of HCE cells. The predicted top-ranked biological functions regulated by glucocorticoids are cell movement (679 genes), cell growth and proliferation (1135 genes), cell development (1002 genes), cell death and survival (918 genes) and cell morphology (685 genes) (Figure 3C). These data suggest that glucocorticoid treatment alters expression of genes involved in migration, growth and trauma. Since injuries affecting the corneal epithelium are the most common types of eye injuries reported (26), and because glucocorticoids are commonly prescribed to suppress inflammation in an injured cornea, we focused our functional analysis on the repair process involved in wound healing. Analysis of the 679 cell movement associated genes indicates that 294 genes (43.3%) were up-regulated and 385 genes (56.7%) were downregulated by dexamethasone (Figure 3D and Table1). Based on the literature, a network of all the 679 genes encompasses different nodes of genes regulating diverse cellular functions related to cell movement.

Figure 3.

Figure 3

Figure 3

Figure 3

Figure 3

Figure 3

Genome-wide regulation by glucocorticoids in HCE cells. HCE cells were treated with vehicle or 100nM dexamethasone (Dex). RNA was isolated and gene expression was analyzed using whole mouse genome 4×44 multiplex format Aglient oligo array. A) Heat map of genes regulated significantly by 100nM dexamethasone in 6 hours (ANOVA p<0.01); Red represents upregulated genes and blue represents downregulated genes in each of the 4 replicates/group. B) Bar-graph is representing glucocorticoid-regulated genes in red (upregulated) and in green (downregulated). C) Glucocorticoid-regulated gene list from HCE cells obtained by microarray were analyzed using Ingenuity Pathway Analysis (IPA) software. IPA predicted glucocorticoid treatment to regulate several molecular and cellular functions, of which the top 5 are listed in the table; Cell movement was ranked as the top most molecular and cellular function regulated by glucocorticoids in HCE cells. D) Gene network of Cell Movement genes identifying different nodes of genes regulating various cellular functions associated with cellular movement; Bar-graph is representing glucocorticoid-regulated cell movement genes in red (upregulated, 294 genes) and in green (downregulated, 385 genes). E-G) Glucocorticoid-regulated genes associated in diseases and functions involving lamellipodia (E), filopodia (F) and permeability (G). Green indicates repression and red indicates upregulation of gene expression; A family of genes with some members upregulated and some members downregulated are indicated in both green and red. The black lines/arrows indicate direct interaction either at the gene/protein level as indicated by Ingenuity Pathway Analysis.

Table 1.

Dexamethasone regulation of genes associated in cell movement

Up regulated genes
Number Symbol Entrez Gene Name Fold
Change
1 PDE2A phosphodiesterase 2A 9.80859
2 TSC22D3 TSC22 domain family member 3 8.37242
3 ALOX5AP arachidonate 5-lipoxygenase-activating protein 5.66178
4 FAM65B family with sequence similarity 65 member B 4.66935
5 PER1 period circadian clock 1 4.65227
6 KLF6 Kruppel-like factor 6 4.38594
7 MMP7 matrix metallopeptidase 7 4.25261
8 ERBB4 erb-b2 receptor tyrosine kinase 4 3.90165
9 EDN2 endothelin 2 3.82819
10 PRKG1 protein kinase, cGMP-dependent, type I 3.7749
11 CGA glycoprotein hormones, alpha polypeptide 3.76295
12 DMBT1 deleted in malignant brain tumors 1 3.72217
13 PTGS2 prostaglandin-endoperoxide synthase 2 (prostaglandin G/H synthase
and cyclooxygenase)
3.6601
14 FGF8 fibroblast growth factor 8 3.36341
15 PLAT plasminogen activator, tissue type 3.26515
16 TREM1 triggering receptor expressed on myeloid cells 1 3.25299
17 FAM43A family with sequence similarity 43 member A 3.07726
18 TLR2 toll-like receptor 2 3.05242
19 COL4A3 collagen, type IV, alpha 3 (Goodpasture antigen) 2.91731
20 ACKR3 atypical chemokine receptor 3 2.91065
21 CCBE1 collagen and calcium binding EGF domains 1 2.89472
22 IGK immunoglobulin kappa locus 2.77607
23 GCNT1 glucosaminyl (N-acetyl) transferase 1, core 2 2.72011
24 FFAR4 free fatty acid receptor 4 2.71943
25 BMP7 bone morphogenetic protein 7 2.63977
26 SCNN1A sodium channel, non voltage gated 1 alpha subunit 2.601
27 MAFB v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog B 2.59413
28 C2 complement component 2 2.56885
29 RGCC regulator of cell cycle 2.5588
30 ZAP70 zeta chain of T cell receptor associated protein kinase 70kDa 2.52303
31 PDIA2 protein disulfide isomerase family A member 2 2.52092
32 PTGES prostaglandin E synthase 2.49236
33 G6PC glucose-6-phosphatase, catalytic subunit 2.47483
34 WNT11 wingless-type MMTV integration site family member 11 2.45361
35 ERRFI1 ERBB receptor feedback inhibitor 1 2.42978
36 LMO7 LIM domain 7 2.42814
37 TWIST1 twist family bHLH transcription factor 1 2.39978
38 SERPINA5 serpin peptidase inhibitor, clade A (alpha-1 antiproteinase,
antitrypsin), member 5
2.39942
39 EDNRB endothelin receptor type B 2.39193
40 GP1BA glycoprotein Ib platelet alpha subunit 2.38223
41 SFRP5 secreted frizzled-related protein 5 2.3733
42 KCNH2 potassium channel, voltage gated eag related subfamily H, member
2
2.33941
43 ST8SIA2 ST8 alpha-N-acetyl-neuraminide alpha-2,8-sialyltransferase 2 2.31557
44 PTGER2 prostaglandin E receptor 2 2.3143
45 TDGF1 teratocarcinoma-derived growth factor 1 2.31039
46 TREM2 triggering receptor expressed on myeloid cells 2 2.30895
47 BARX2 BARX homeobox 2 2.29272
48 TNS4 tensin 4 2.2516
49 SDR9C7 short chain dehydrogenase/reductase family 9C, member 7 2.24862
50 SOX10 SRY-box 10 2.23462
51 NME8 NME/NM23 family member 8 2.22648
52 HYAL1 hyaluronoglucosaminidase 1 2.22431
53 SFRP2 secreted frizzled-related protein 2 2.22094
54 ANXA2 annexin A2 2.21004
55 CSF1R colony stimulating factor 1 receptor 2.20717
56 MYCN v-myc avian myelocytomatosis viral oncogene neuroblastoma
derived homolog
2.18662
57 PNOC prepronociceptin 2.18369
58 POU3F2 POU class 3 homeobox 2 2.14487
59 LHX6 LIM homeobox 6 2.13937
60 NCAM1 neural cell adhesion molecule 1 2.12964
61 UNC5C unc-5 netrin receptor C 2.12805
62 SAA1 serum amyloid A1 2.12535
63 ARHGEF6 Rac/Cdc42 guanine nucleotide exchange factor 6 2.12256
64 SEPT4 septin 4 2.09427
65 SEMA6D semaphorin 6D 2.08409
66 AHSG alpha-2-HS-glycoprotein 2.04678
67 FOXO4 forkhead box O4 2.03975
68 DUSP1 dual specificity phosphatase 1 2.03074
69 PIGR polymeric immunoglobulin receptor 2.02612
70 CSPG4 chondroitin sulfate proteoglycan 4 2.02415
71 SERPINA3 serpin peptidase inhibitor, clade A (alpha-1 antiproteinase,
antitrypsin), member 3
2.02111
72 FGD4 FYVE, RhoGEF and PH domain containing 4 2.01625
73 DACT2 dishevelled-binding antagonist of beta-catenin 2 1.99964
74 OVOL2 ovo-like zinc finger 2 1.99502
75 HTRA3 HtrA serine peptidase 3 1.98175
76 LOX lysyl oxidase 1.96085
77 SNAI2 snail family zinc finger 2 1.95867
78 ST3GAL3 ST3 beta-galactoside alpha-2,3-sialyltransferase 3 1.95677
79 DKK1 dickkopf WNT signaling pathway inhibitor 1 1.95068
80 CAV3 caveolin 3 1.93759
81 PDPK1 3-phosphoinositide dependent protein kinase 1 1.93146
82 SGK1 serum/glucocorticoid regulated kinase 1 1.92796
83 CX3CL1 chemokine (C-X3-C motif) ligand 1 1.90246
84 RECK reversion-inducing-cysteine-rich protein with kazal motifs 1.90193
85 TAF7L TATA-box binding protein associated factor 7 like 1.89943
86 CXCL9 chemokine (C-X-C motif) ligand 9 1.89439
87 SERPINE1 serpin peptidase inhibitor, clade E (nexin, plasminogen activator
inhibitor type 1), member 1
1.89231
88 ESAM endothelial cell adhesion molecule 1.8895
89 MUC2 mucin 2, oligomeric mucus/gel-forming 1.88395
90 CCKBR cholecystokinin B receptor 1.88322
91 MTSS1 metastasis suppressor 1 1.87278
92 SRCIN1 SRC kinase signaling inhibitor 1 1.87237
93 SCNN1G sodium channel, non voltage gated 1 gamma subunit 1.87131
94 GUCY1A3 guanylate cyclase 1, soluble, alpha 3 1.86393
95 BCAN brevican 1.84483
96 MMP28 matrix metallopeptidase 28 1.84228
97 A4GALT alpha 1,4-galactosyltransferase 1.83856
98 ARRDC3 arrestin domain containing 3 1.82678
99 IL17C interleukin 17C 1.80878
100 TNFRSF18 tumor necrosis factor receptor superfamily member 18 1.80542
101 LMCD1 LIM and cysteine-rich domains 1 1.79297
102 APBB2 amyloid beta (A4) precursor protein-binding, family B, member 2 1.78296
103 GPR173 G protein-coupled receptor 173 1.76787
104 CD22 CD22 molecule 1.7662
105 ARID5B AT-rich interaction domain 5B 1.76014
106 COL2A1 collagen, type II, alpha 1 1.73823
107 TREML2 triggering receptor expressed on myeloid cells like 2 1.72252
108 CBLB Cbl proto-oncogene B, E3 ubiquitin protein ligase 1.71418
109 DMD dystrophin 1.70442
110 PTGDR2 prostaglandin D2 receptor 2 1.6963
111 CSF1 colony stimulating factor 1 1.69629
112 DEFB1 defensin beta 1 1.69534
113 AHNAK AHNAK nucleoprotein 1.69458
114 ALPPL2 alkaline phosphatase, placental like 2 1.69249
115 ITGB6 integrin subunit beta 6 1.69233
116 KDR kinase insert domain receptor 1.69116
117 NFKBIA nuclear factor of kappa light polypeptide gene enhancer in B-cells
inhibitor, alpha
1.68325
118 THBD thrombomodulin 1.68252
119 DSG3 desmoglein 3 1.68104
120 IL6R interleukin 6 receptor 1.67241
121 AZGP1 alpha-2-glycoprotein 1, zinc-binding 1.67019
122 CKLF chemokine-like factor 1.6601
123 FABP5 fatty acid binding protein 5 (psoriasis-associated) 1.65888
124 DNAJB4 DnaJ heat shock protein family (Hsp40) member B4 1.65114
125 STAB2 stabilin 2 1.64157
126 GUCY1B3 guanylate cyclase 1, soluble, beta 3 1.64096
127 STAB1 stabilin 1 1.6352
128 HOXC9 homeobox C9 1.63051
129 KRT6B keratin 6B, type II 1.61929
130 IL6ST interleukin 6 signal transducer 1.61917
131 GGT5 gamma-glutamyltransferase 5 1.61518
132 PTH1R parathyroid hormone 1 receptor 1.61329
133 FOXO1 forkhead box O1 1.6106
134 OPRD1 opioid receptor, delta 1 1.61012
135 TNFSF8 tumor necrosis factor superfamily member 8 1.60148
136 TG thyroglobulin 1.59772
137 GNAQ guanine nucleotide binding protein (G protein), q polypeptide 1.59632
138 MYO9A myosin IXA 1.58634
139 TXNDC2 thioredoxin domain containing 2 (spermatozoa) 1.57836
140 WNT5A wingless-type MMTV integration site family member 5A 1.56678
141 ROR1 receptor tyrosine kinase-like orphan receptor 1 1.56674
142 CRMP1 collapsin response mediator protein 1 1.55331
143 ATF3 activating transcription factor 3 1.55296
144 NTF4 neurotrophin 4 1.54373
145 C5AR2 complement component 5a receptor 2 1.54316
146 CD209 CD209 molecule 1.54084
147 TGFBR2 transforming growth factor beta receptor II 1.53985
148 HLA-G major histocompatibility complex, class I, G 1.53451
149 CDH4 cadherin 4 1.5343
150 SPARCL1 SPARC like 1 1.53001
151 SFTPA1 surfactant protein A1 1.52362
152 FAP fibroblast activation protein alpha 1.51341
153 MUC1 mucin 1, cell surface associated 1.50915
154 HIPK2 homeodomain interacting protein kinase 2 1.50556
155 LAMA3 laminin subunit alpha 3 1.50543
156 ADGRL3 adhesion G protein-coupled receptor L3 1.50267
157 NDRG1 N-myc downstream regulated 1 1.5009
158 DNAH11 dynein, axonemal, heavy chain 11 1.49615
159 HTRA1 HtrA serine peptidase 1 1.49229
160 DKK3 dickkopf WNT signaling pathway inhibitor 3 1.48976
161 IRX2 iroquois homeobox 2 1.48728
162 AKAP12 A-kinase anchoring protein 12 1.48594
163 CD8A CD8a molecule 1.48588
164 CTGF connective tissue growth factor 1.47837
165 PROK1 prokineticin 1 1.47053
166 MAVS mitochondrial antiviral signaling protein 1.46925
167 IRS2 insulin receptor substrate 2 1.46551
168 ATP2B4 ATPase, Ca++ transporting, plasma membrane 4 1.46535
169 SEMA5A semaphorin 5A 1.46424
170 AFAP1L1 actin filament associated protein 1 like 1 1.46064
171 MAPT microtubule associated protein tau 1.45851
172 FOSL2 FOS like antigen 2 1.45689
173 NFIA nuclear factor I/A 1.45523
174 EGFR epidermal growth factor receptor 1.44947
175 ST3GAL4 ST3 beta-galactoside alpha-2,3-sialyltransferase 4 1.4478
176 ALPP alkaline phosphatase, placental 1.44571
177 TUBB2B tubulin beta 2B class IIb 1.4385
178 NTN1 netrin 1 1.43575
179 CXCL5 chemokine (C-X-C motif) ligand 5 1.43458
180 CRYAB crystallin alpha B 1.43261
181 PLD1 phospholipase D1 1.42163
182 CD59 CD59 molecule 1.42106
183 KLF4 Kruppel-like factor 4 (gut) 1.41722
184 VNN2 vanin 2 1.41559
185 PLXNA2 plexin A2 1.40612
186 CMA1 chymase 1, mast cell 1.40552
187 SLC30A4 solute carrier family 30 (zinc transporter), member 4 1.40175
188 UNC5B unc-5 netrin receptor B 1.39967
189 MARVELD MARVEL domain containing 3 1.3954
190 ABLIM1 actin binding LIM protein 1 1.39124
191 FBLIM1 filamin binding LIM protein 1 1.39083
192 JUNB jun B proto-oncogene 1.38982
193 KLF8 Kruppel-like factor 8 1.38587
194 CHRNA7 cholinergic receptor, nicotinic alpha 7 1.38155
195 ITGB7 integrin subunit beta 7 1.37875
196 ROBO3 roundabout guidance receptor 3 1.37754
197 PXN paxillin 1.37578
198 CTSC cathepsin C 1.37552
199 FLNB filamin B 1.37067
200 CAMK2G calcium/calmodulin-dependent protein kinase II gamma 1.36898
201 GPC1 glypican 1 1.36368
202 TRIP10 thyroid hormone receptor interactor 10 1.36183
203 GLI3 GLI family zinc finger 3 1.35695
204 PDCD4 programmed cell death 4 (neoplastic transformation inhibitor) 1.35597
205 TSC1 tuberous sclerosis 1 1.35562
206 IL15 interleukin 15 1.35092
207 TNFRSF1A tumor necrosis factor receptor superfamily member 1A 1.34708
208 GLI2 GLI family zinc finger 2 1.34616
209 DAB2 Dab, mitogen-responsive phosphoprotein, homolog 2 (Drosophila) 1.3451
210 CEBPD CCAAT/enhancer binding protein delta 1.34448
211 NOD1 nucleotide binding oligomerization domain containing 1 1.34023
212 RHOB ras homolog family member B 1.33951
213 PDLIM1 PDZ and LIM domain 1 1.33705
214 ALOX15 arachidonate 15-lipoxygenase 1.33616
215 GPR161 G protein-coupled receptor 161 1.33444
216 CRIP2 cysteine-rich protein 2 1.32923
217 ARHGEF18 Rho/Rac guanine nucleotide exchange factor 18 1.32742
218 CEBPE CCAAT/enhancer binding protein epsilon 1.32704
219 ETS2 v-ets avian erythroblastosis virus E26 oncogene homolog 2 1.31809
220 SIRPA signal-regulatory protein alpha 1.3081
221 IFNGR1 interferon gamma receptor 1 1.30326
222 APBA3 amyloid beta (A4) precursor protein-binding, family A, member 3 1.29918
223 CREB3L1 cAMP responsive element binding protein 3-like 1 1.29846
224 WASF2 WAS protein family member 2 1.29785
225 KRT16 keratin 16, type I 1.29531
226 ANO6 anoctamin 6 1.29504
227 FPR1 formyl peptide receptor 1 1.2919
228 GBA glucosidase, beta, acid 1.28636
229 NEURL1 neuralized E3 ubiquitin protein ligase 1 1.28481
230 EFNB1 ephrin-B1 1.28338
231 REPS2 RALBP1 associated Eps domain containing 2 1.28076
232 CAV1 caveolin 1 1.27821
233 FHOD1 formin homology 2 domain containing 1 1.2777
234 TBXAS1 thromboxane A synthase 1 1.27669
235 ID3 inhibitor of DNA binding 3, dominant negative helix-loop-helix protein 1.27432
236 KLK7 kallikrein related peptidase 7 1.27282
237 CTSB cathepsin B 1.2722
238 E2F2 E2F transcription factor 2 1.26991
239 ATF2 activating transcription factor 2 1.26929
240 ANG angiogenin, ribonuclease, RNase A family, 5 1.26295
241 FOXQ1 forkhead box Q1 1.26212
242 IGFBP3 insulin like growth factor binding protein 3 1.24814
243 KLF5 Kruppel-like factor 5 (intestinal) 1.24315
244 PEX11B peroxisomal biogenesis factor 11 beta 1.24302
245 CTBP2 C-terminal binding protein 2 1.23906
246 GADD45A growth arrest and DNA damage inducible alpha 1.23381
247 UNK unkempt family zinc finger 1.22682
248 EPHB2 EPH receptor B2 1.22406
249 SEPT11 septin 11 1.21872
250 FLCN folliculin 1.21512
251 LIMK2 LIM domain kinase 2 1.2114
252 ALOX5 arachidonate 5-lipoxygenase 1.21048
253 STARD13 StAR related lipid transfer domain containing 13 1.20956
254 KIF1C kinesin family member 1C 1.20914
255 CEBPB CCAAT/enhancer binding protein beta 1.20899
256 CFB complement factor B 1.20881
257 PRKCZ protein kinase C, zeta 1.20575
258 PML promyelocytic leukemia 1.20095
259 WASF1 WAS protein family member 1 1.19908
260 MYO1E myosin IE 1.19684
261 PPARA peroxisome proliferator-activated receptor alpha 1.1953
262 MFI2 antigen p97 (melanoma associated) identified by monoclonal
antibodies 133.2 and 96.5
1.19307
263 THRA thyroid hormone receptor, alpha 1.19212
264 PLCG2 phospholipase C gamma 2 1.19
265 NOL3 nucleolar protein 3 1.18725
266 DCTN2 dynactin subunit 2 1.18711
267 SWAP70 SWAP switching B-cell complex 70kDa subunit 1.18608
268 TYRO3 TYRO3 protein tyrosine kinase 1.18563
269 OGG1 8-oxoguanine DNA glycosylase 1.18292
270 DOK1 docking protein 1 1.17558
271 LIMS2 LIM-type zinc finger domains 2 1.16949
272 GRHL2 grainyhead like transcription factor 2 1.16062
273 CDC42BPB CDC42 binding protein kinase beta 1.16052
274 CDK5RAP3 CDK5 regulatory subunit associated protein 3 1.15733
275 ROBO4 roundabout guidance receptor 4 1.15231
276 MAPK7 mitogen-activated protein kinase 7 1.14122
277 NISCH nischarin 1.1397
278 TXN thioredoxin 1.13966
279 BATF3 basic leucine zipper transcription factor, ATF-like 3 1.1359
280 SPSB1 splA/ryanodine receptor domain and SOCS box containing 1 1.13579
281 RRAS related RAS viral (r-ras) oncogene homolog 1.10437
282 LPAR4 lysophosphatidic acid receptor 4 1.0704
283 LILRB1 leukocyte immunoglobulin like receptor B1 1.06714
284 GPR18 G protein-coupled receptor 18 1.06277
285 SELP selectin P 1.06242
286 SH2D3C SH2 domain containing 3C 1.06103
287 IL16 interleukin 16 1.05912
288 ESR1 estrogen receptor 1 1.05874
289 PLXNC1 plexin C1 1.05869
290 BCL11B B-cell CLL/lymphoma 11B 1.05823
291 CYP19A1 cytochrome P450 family 19 subfamily A member 1 1.05594
292 CCR8 chemokine (C-C motif) receptor 8 1.05587
293 EPB41L4B erythrocyte membrane protein band 4.1 like 4B 1.05141
294 RARRES2 retinoic acid receptor responder (tazarotene induced) 2 1.04311
Down regulated genes
Number Symbol Entrez Gene Name Fold
Change
1 PHLDA1 pleckstrin homology-like domain, family A, member 1 −8.46535
2 TNFRSF11B tumor necrosis factor receptor superfamily member 11b −7.88483
3 PDE4B phosphodiesterase 4B −5.22171
4 IL6 interleukin 6 −4.46666
5 FGF5 fibroblast growth factor 5 −4.31595
6 CD36 CD36 molecule −3.92846
7 IL1B interleukin 1 beta −3.74371
8 EREG epiregulin −3.51023
9 CCL2 chemokine (C-C motif) ligand 2 −3.41548
10 RUNX2 runt-related transcription factor 2 −2.95184
11 IGFBP5 insulin like growth factor binding protein 5 −2.92668
12 CXCL8 chemokine (C-X-C motif) ligand 8 −2.91859
13 CYP26A1 cytochrome P450 family 26 subfamily A member 1 −2.91556
14 PLAU plasminogen activator, urokinase −2.90533
15 IL24 interleukin 24 −2.88468
16 ST3GAL5 ST3 beta-galactoside alpha-2,3-sialyltransferase 5 −2.88077
17 SEMA3A semaphorin 3A −2.87725
18 TNFSF15 tumor necrosis factor superfamily member 15 −2.81580
19 FGFR1 fibroblast growth factor receptor 1 −2.76805
20 DIO2 deiodinase, iodothyronine, type II −2.73236
21 TXK TXK tyrosine kinase −2.62840
22 SEMA3D semaphorin 3D −2.58360
23 FGFBP1 fibroblast growth factor binding protein 1 −2.51183
24 KCNA3 potassium channel, voltage gated shaker related subfamily A,
member 3
−2.50982
25 FAT3 FAT atypical cadherin 3 −2.46791
26 CCRL2 chemokine (C-C motif) receptor-like 2 −2.45201
27 NPPB natriuretic peptide B −2.41923
28 ETV1 ets variant 1 −2.41900
29 LIF leukemia inhibitory factor −2.41840
30 CD28 CD28 molecule −2.40401
31 EHF ets homologous factor −2.39617
32 SERPINB3 serpin peptidase inhibitor, clade B (ovalbumin), member 3 −2.37556
33 SEMA3E semaphorin 3E −2.35253
34 CSF2 colony stimulating factor 2 −2.34563
35 ST8SIA4 ST8 alpha-N-acetyl-neuraminide alpha-2,8-sialyltransferase 4 −2.32584
36 CEMIP cell migration inducing protein, hyaluronan binding −2.29345
37 FOSL1 FOS like antigen 1 −2.28415
38 TFPI2 tissue factor pathway inhibitor 2 −2.26858
39 BDNF brain-derived neurotrophic factor −2.24540
40 NR0B1 nuclear receptor subfamily 0 group B member 1 −2.23654
41 OPRM1 opioid receptor, mu 1 −2.23159
42 CCL3 chemokine (C-C motif) ligand 3 −2.22620
43 CYP1B1 cytochrome P450 family 1 subfamily B member 1 −2.20031
44 SCRN3 secernin 3 −2.13833
45 CSH1/CSH2 chorionic somatomammotropin hormone 1 −2.12536
46 SPRY4 sprouty RTK signaling antagonist 4 −2.12339
47 RGS4 regulator of G-protein signaling 4 −2.08806
48 NR2F1 nuclear receptor subfamily 2 group F member 1 −2.08334
49 IL23A interleukin 23 subunit alpha −2.08326
50 IFIT2 interferon induced protein with tetratricopeptide repeats 2 −2.07608
51 FOXA1 forkhead box A1 −2.06692
52 F3 coagulation factor III, tissue factor −2.05004
53 ITGA2 integrin subunit alpha 2 −2.04652
54 CCDC40 coiled-coil domain containing 40 −2.04553
55 ZBTB18 zinc finger and BTB domain containing 18 −2.03725
56 GDNF glial cell derived neurotrophic factor −2.02472
57 CLDN11 claudin 11 −2.02083
58 IER3 immediate early response 3 −1.97907
59 NOG noggin −1.97215
60 BDKRB2 bradykinin receptor B2 −1.96909
61 CLDN1 claudin 1 −1.95484
62 GREM1 gremlin 1, DAN family BMP antagonist −1.95480
63 CCL3L3 chemokine (C-C motif) ligand 3-like 3 −1.94494
64 ADAM19 ADAM metallopeptidase domain 19 −1.93804
65 ETV5 ets variant 5 −1.93529
66 STC1 stanniocalcin 1 −1.92768
67 CDH11 cadherin 11 −1.92589
68 CXCL1 chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating
activity, alpha)
−1.92384
69 CADM1 cell adhesion molecule 1 −1.92359
70 TNFRSF21 tumor necrosis factor receptor superfamily member 21 −1.91959
71 E2F5 E2F transcription factor 5, p130-binding −1.91884
72 CD44 CD44 molecule (Indian blood group) −1.91830
73 ADAMTS1 ADAM metallopeptidase with thrombospondin type 1 motif 1 −1.89520
74 SMAD3 SMAD family member 3 −1.86856
75 ADAM8 ADAM metallopeptidase domain 8 −1.86707
76 HBEGF heparin-binding EGF-like growth factor −1.85743
77 SOX9 SRY-box 9 −1.84049
78 NOV nephroblastoma overexpressed −1.83286
79 IRF8 interferon regulatory factor 8 −1.83215
80 GDF15 growth differentiation factor 15 −1.82966
81 HAVCR2 hepatitis A virus cellular receptor 2 −1.82840
82 LIPE lipase, hormone-sensitive −1.82349
83 SHC4 SHC (Src homology 2 domain containing) family member 4 −1.82181
84 ITGB8 integrin subunit beta 8 −1.81877
85 PRKCE protein kinase C, epsilon −1.81237
86 PTHLH parathyroid hormone-like hormone −1.79945
87 SPRY2 sprouty RTK signaling antagonist 2 −1.77757
88 PHLDA2 pleckstrin homology-like domain, family A, member 2 −1.76688
89 F2RL1 coagulation factor II (thrombin) receptor-like 1 −1.75500
90 HDAC9 histone deacetylase 9 −1.75136
91 CAMK2D calcium/calmodulin-dependent protein kinase II delta −1.74928
92 VEGFA vascular endothelial growth factor A −1.74782
93 IL21R interleukin 21 receptor −1.74709
94 MAP2K6 mitogen-activated protein kinase kinase 6 −1.74136
95 GPER1 G protein-coupled estrogen receptor 1 −1.73288
96 ADRA2A adrenoceptor alpha 2A −1.72940
97 CCND1 cyclin D1 −1.72596
98 CDCP1 CUB domain containing protein 1 −1.72023
99 CD274 CD274 molecule −1.70099
100 WNT7B wingless-type MMTV integration site family member 7B −1.69513
101 MMP13 matrix metallopeptidase 13 −1.69399
102 WNT7A wingless-type MMTV integration site family member 7A −1.69365
103 SOX7 SRY-box 7 −1.68947
104 AQP3 aquaporin 3 (Gill blood group) −1.68904
105 NGF nerve growth factor (beta polypeptide) −1.68514
106 VEGFC vascular endothelial growth factor C −1.67461
107 CFTR cystic fibrosis transmembrane conductance regulator −1.67311
108 CEACAM1 carcinoembryonic antigen related cell adhesion molecule 1 −1.67013
109 BCL3 B-cell CLL/lymphoma 3 −1.66934
110 EGF epidermal growth factor −1.66451
111 NUAK1 NUAK family, SNF1-like kinase, 1 −1.66163
112 MAP2K3 mitogen-activated protein kinase kinase 3 −1.65913
113 NUAK2 NUAK family, SNF1-like kinase, 2 −1.65090
114 PTX3 pentraxin 3 −1.65079
115 IL11 interleukin 11 −1.64764
116 TFAP2A transcription factor AP-2 alpha (activating enhancer binding
protein 2 alpha)
−1.64024
117 DUSP6 dual specificity phosphatase 6 −1.63858
118 EPAS1 endothelial PAS domain protein 1 −1.63589
119 DNMBP dynamin binding protein −1.62689
120 PLAUR plasminogen activator, urokinase receptor −1.62192
121 SLIT2 slit guidance ligand 2 −1.61888
122 AFAP1 actin filament associated protein 1 −1.61378
123 TM4SF1 transmembrane 4 L six family member 1 −1.60547
124 DLC1 DLC1 Rho GTPase activating protein −1.60302
125 DLX2 distal-less homeobox 2 −1.59963
126 IL15RA interleukin 15 receptor subunit alpha −1.59691
127 DUSP4 dual specificity phosphatase 4 −1.59289
128 RASGRP1 RAS guanyl releasing protein 1 (calcium and DAG-regulated) −1.58550
129 LTB lymphotoxin beta −1.58178
130 CLDN4 claudin 4 −1.57162
131 NR2F2 nuclear receptor subfamily 2 group F member 2 −1.56903
132 OSMR oncostatin M receptor −1.56874
133 LRP8 LDL receptor related protein 8 −1.56623
134 STX6 syntaxin 6 −1.56481
135 ST6GALNAC
5
ST6 (alpha-N-acetyl-neuraminyl-2,3-beta-galactosyl-1,3)-N-
acetylgalactosaminide alpha-2,6-sialyltransferase 5
−1.56360
136 ABL2 ABL proto-oncogene 2, non-receptor tyrosine kinase −1.56352
137 TCF4 transcription factor 4 −1.55623
138 MYO10 myosin X −1.55431
139 PTAFR platelet-activating factor receptor −1.55164
140 SASH1 SAM and SH3 domain containing 1 −1.54902
141 NRG1 neuregulin 1 −1.54870
142 SFRP4 secreted frizzled-related protein 4 −1.54760
143 OLR1 oxidized low density lipoprotein (lectin-like) receptor 1 −1.54270
144 MMP1 matrix metallopeptidase 1 −1.54030
145 LYN LYN proto-oncogene, Src family tyrosine kinase −1.53885
146 CLDN3 claudin 3 −1.53743
147 SH3KBP1 SH3-domain kinase binding protein 1 −1.53570
148 EGLN3 egl-9 family hypoxia-inducible factor 3 −1.52322
149 PIK3CD phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit
delta
−1.51461
150 BACH2 BTB and CNC homology 1, basic leucine zipper transcription
factor 2
−1.51327
151 FST follistatin −1.50979
152 SOCS3 suppressor of cytokine signaling 3 −1.50825
153 ADORA2B adenosine A2b receptor −1.50713
154 ETV6 ets variant 6 −1.50633
155 S100A7A S100 calcium binding protein A7A −1.50499
156 AMOTL1 angiomotin like 1 −1.49759
157 TMSB10/TM
SB4X
thymosin beta 10 −1.49466
158 P2RY6 pyrimidinergic receptor P2Y, G-protein coupled, 6 −1.49425
159 RARRES1 retinoic acid receptor responder (tazarotene induced) 1 −1.49203
160 CXADR coxsackie virus and adenovirus receptor −1.49154
161 MMP2 matrix metallopeptidase 2 −1.48914
162 CCK cholecystokinin −1.48872
163 ODC1 ornithine decarboxylase 1 −1.48278
164 LDLR low density lipoprotein receptor −1.48242
165 SLC12A2 solute carrier family 12 (sodium/potassium/chloride transporter),
member 2
−1.48129
166 SPNS2 spinster homolog 2 (Drosophila) −1.47624
167 EPHB3 EPH receptor B3 −1.47347
168 HSP90AA1 heat shock protein 90kDa alpha family class A member 1 −1.47258
169 BDKRB1 bradykinin receptor B1 −1.46892
170 ITGA6 integrin subunit alpha 6 −1.46736
171 CMTM8 CKLF-like MARVEL transmembrane domain containing 8 −1.46426
172 C5AR1 complement component 5a receptor 1 −1.46246
173 KRAS Kirsten rat sarcoma viral oncogene homolog −1.45816
174 HTR7 5-hydroxytryptamine (serotonin) receptor 7, adenylate cyclase-
coupled
−1.45507
175 CIITA class II, major histocompatibility complex, transactivator −1.45446
176 BHLHE41 basic helix-loop-helix family member e41 −1.45152
177 PTK2 protein tyrosine kinase 2 −1.44795
178 FAM60A family with sequence similarity 60 member A −1.44001
179 ID4 inhibitor of DNA binding 4, dominant negative helix-loop-helix
protein
−1.43998
180 TCF7L2 transcription factor 7-like 2 (T-cell specific, HMG-box) −1.43844
181 MESP1 mesoderm posterior bHLH transcription factor 1 −1.43834
182 TGFA transforming growth factor alpha −1.4379
183 CNN1 calponin 1, basic, smooth muscle −1.4304
184 TFPI tissue factor pathway inhibitor −1.42959
185 ETV4 ets variant 4 −1.42898
186 HMGA2 high mobility group AT-hook 2 −1.42812
187 PELI1 pellino E3 ubiquitin protein ligase 1 −1.42779
188 LCP2 lymphocyte cytosolic protein 2 −1.42448
189 CTNNA2 catenin alpha 2 −1.42091
190 TYR tyrosinase −1.42076
191 TPM1 tropomyosin 1 (alpha) −1.42072
192 SULT2B1 sulfotransferase family 2B member 1 −1.42007
193 ADAM12 ADAM metallopeptidase domain 12 −1.42003
194 SELL selectin L −1.41915
195 HAS3 hyaluronan synthase 3 −1.41793
196 RIPK2 receptor interacting serine/threonine kinase 2 −1.41689
197 SLC2A1 solute carrier family 2 (facilitated glucose transporter), member 1 −1.41412
198 AQP1 aquaporin 1 (Colton blood group) −1.4135
199 CATSPERD catsper channel auxiliary subunit delta −1.41101
200 ETS1 v-ets avian erythroblastosis virus E26 oncogene homolog 1 −1.411
201 SCN9A sodium channel, voltage gated, type IX alpha subunit −1.4108
202 SMAD7 SMAD family member 7 −1.40998
203 ICAM1 intercellular adhesion molecule 1 −1.40688
204 HGF hepatocyte growth factor (hepapoietin A; scatter factor) −1.40635
205 IGF1R insulin like growth factor 1 receptor −1.40444
206 SYNM synemin −1.40255
207 PODXL podocalyxin-like −1.40063
208 CLEC7A C-type lectin domain family 7 member A −1.40008
209 STK35 serine/threonine kinase 35 −1.3991
210 ADORA3 adenosine A3 receptor −1.39827
211 NR1D1 nuclear receptor subfamily 1 group D member 1 −1.39569
212 ZDHHC20 zinc finger, DHHC-type containing 20 −1.39423
213 FOXP1 forkhead box P1 −1.39132
214 CAPN12 calpain 12 −1.39041
215 ATP1A4 ATPase, Na+/K+ transporting, alpha 4 polypeptide −1.38932
216 CXCL2 chemokine (C-X-C motif) ligand 2 −1.38857
217 JAG1 jagged 1 −1.388
218 RUNX1 runt-related transcription factor 1 −1.38576
219 RASSF5 Ras association (RalGDS/AF-6) domain family member 5 −1.38344
220 MYH11 myosin, heavy chain 11, smooth muscle −1.37585
221 PPIF peptidylprolyl isomerase F −1.37223
222 AJAP1 adherens junctions associated protein 1 −1.37108
223 IGHG1 immunoglobulin heavy constant gamma 1 (G1m marker) −1.37108
224 SNAI1 snail family zinc finger 1 −1.36983
225 EIF4E eukaryotic translation initiation factor 4E −1.36964
226 RASGRF1 Ras protein specific guanine nucleotide releasing factor 1 −1.36863
227 PTPRK protein tyrosine phosphatase, receptor type K −1.36543
228 CDC42EP1 CDC42 effector protein 1 −1.36535
229 GCNT2 glucosaminyl (N-acetyl) transferase 2, I-branching enzyme (I blood
group)
−1.36516
230 DEFB103A/D
EFB103B
defensin beta 103B −1.36313
231 VDR vitamin D (1,25- dihydroxyvitamin D3) receptor −1.36297
232 PARD6B par-6 family cell polarity regulator beta −1.36193
233 IER2 immediate early response 2 −1.36131
234 DLL1 delta-like 1 (Drosophila) −1.36055
235 PTPRR protein tyrosine phosphatase, receptor type R −1.35893
236 CATSPER1 cation channel, sperm associated 1 −1.3582
237 WNT4 wingless-type MMTV integration site family member 4 −1.34903
238 PTP4A1 protein tyrosine phosphatase type IVA, member 1 −1.34726
239 TNFRSF10A tumor necrosis factor receptor superfamily member 10a −1.34601
240 EPHB1 EPH receptor B1 −1.34462
241 TNFRSF12A tumor necrosis factor receptor superfamily member 12A −1.34406
242 GEMIN5 gem nuclear organelle associated protein 5 −1.34324
243 LMO4 LIM domain only 4 −1.34176
244 FUT8 fucosyltransferase 8 (alpha (1,6) fucosyltransferase) −1.3409
245 MYF5 myogenic factor 5 −1.33933
246 NAA15 N(alpha)-acetyltransferase 15, NatA auxiliary subunit −1.33862
247 DNAJB6 DnaJ heat shock protein family (Hsp40) member B6 −1.33628
248 AGTR2 angiotensin II receptor type 2 −1.33574
249 MITF microphthalmia-associated transcription factor −1.33566
250 CHL1 cell adhesion molecule L1-like −1.33449
251 WISP2 WNT1 inducible signaling pathway protein 2 −1.33436
252 TLR3 toll-like receptor 3 −1.33216
253 PRSS27 protease, serine 27 −1.32726
254 FUT3 fucosyltransferase 3 (Lewis blood group) −1.32708
255 GLRX glutaredoxin −1.3242
256 TGFBI transforming growth factor beta induced −1.32276
257 KNG1 kininogen 1 −1.32215
258 ONECUT2 one cut homeobox 2 −1.32092
259 RBFOX2 RNA binding protein, fox-1 homolog (C. elegans) 2 −1.3207
260 TSHR thyroid stimulating hormone receptor −1.32046
261 HSP90AB1 heat shock protein 90kDa alpha family class B member 1 −1.31987
262 GAD1 glutamate decarboxylase 1 −1.31719
263 SPAG9 sperm associated antigen 9 −1.31697
264 FGF2 fibroblast growth factor 2 (basic) −1.31569
265 SOCS4 suppressor of cytokine signaling 4 −1.31507
266 IL17RB interleukin 17 receptor B −1.3143
267 PIK3CG phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit
gamma
−1.31388
268 MNX1 motor neuron and pancreas homeobox 1 −1.31094
269 ASPH aspartate beta-hydroxylase −1.31058
270 PDGFB platelet derived growth factor subunit B −1.30629
271 SYNJ2BP synaptojanin 2 binding protein −1.30419
272 VAV1 vav guanine nucleotide exchange factor 1 −1.30245
273 PLPP3 phospholipid phosphatase 3 −1.30085
274 RELB v-rel avian reticuloendotheliosis viral oncogene homolog B −1.29866
275 YWHAQ tyrosine 3-monooxygenase/tryptophan 5-monooxygenase
activation protein, theta
−1.29533
276 NDST1 N-deacetylase/N-sulfotransferase (heparan glucosaminyl) 1 −1.28877
277 DYX1C1 dyslexia susceptibility 1 candidate 1 −1.28795
278 CLEC5A C-type lectin domain family 5 member A −1.28692
279 POMK protein-O-mannose kinase −1.28647
280 ZFAND5 zinc finger, AN1-type domain 5 −1.28575
281 SMN1/SMN2 survival of motor neuron 1, telomeric −1.28521
282 EFNA1 ephrin-A1 −1.28027
283 VTCN1 V-set domain containing T cell activation inhibitor 1 −1.27926
284 AMD1 adenosylmethionine decarboxylase 1 −1.27848
285 RND3 Rho family GTPase 3 −1.27788
286 DCBLD2 discoidin, CUB and LCCL domain containing 2 −1.27761
287 GJA1 gap junction protein alpha 1 −1.27588
288 HOXA2 homeobox A2 −1.27472
289 TWIST2 twist family bHLH transcription factor 2 −1.273
290 ADGRG1 adhesion G protein-coupled receptor G1 −1.26945
291 PIK3C2B phosphatidylinositol-4-phosphate 3-kinase catalytic subunit type 2
beta
−1.26945
292 NAMPT nicotinamide phosphoribosyltransferase −1.26749
293 PRMT6 protein arginine methyltransferase 6 −1.26295
294 TRAF3 TNF receptor associated factor 3 −1.26024
295 EZR ezrin −1.25889
296 PPP1R15A protein phosphatase 1 regulatory subunit 15A −1.25828
297 DRAM1 DNA damage regulated autophagy modulator 1 −1.25703
298 SNCA synuclein alpha −1.25233
299 OCLN occludin −1.25226
300 KCNK5 potassium channel, two pore domain subfamily K, member 5 −1.25156
301 PRLR prolactin receptor −1.2486
302 BCAR1 breast cancer anti-estrogen resistance 1 −1.24785
303 NFAT5 nuclear factor of activated T-cells 5, tonicity-responsive −1.24658
304 RAC2 ras-related C3 botulinum toxin substrate 2 (rho family, small GTP
binding protein Rac2)
−1.24535
305 MALT1 MALT1 paracaspase −1.24468
306 HHEX hematopoietically expressed homeobox −1.24069
307 DNAJA1 DnaJ heat shock protein family (Hsp40) member A1 −1.23969
308 NET1 neuroepithelial cell transforming 1 −1.23934
309 TCAF1 TRPM8 channel-associated factor 1 −1.239
310 ARHGAP19 Rho GTPase activating protein 19 −1.23845
311 ZNF652 zinc finger protein 652 −1.23649
312 CCL26 chemokine (C-C motif) ligand 26 −1.23552
313 MIEN1 migration and invasion enhancer 1 −1.2349
314 SMG1 SMG1 phosphatidylinositol 3-kinase-related kinase −1.23249
315 MYO5B myosin VB −1.23164
316 RAG1 recombination activating gene 1 −1.2303
317 CGB3
(includes
others)
chorionic gonadotropin beta subunit 3 −1.22918
318 RALGAPA2 Ral GTPase activating protein, alpha subunit 2 (catalytic) −1.22615
319 B4GALT5 UDP-Gal:betaGlcNAc beta 1,4- galactosyltransferase, polypeptide
5
−1.22529
320 GSK3B glycogen synthase kinase 3 beta −1.22451
321 CDK6 cyclin-dependent kinase 6 −1.22402
322 CTNNAL1 catenin alpha-like 1 −1.22048
323 CD58 CD58 molecule −1.21835
324 CYR61 cysteine rich angiogenic inducer 61 −1.21306
325 ZNF24 zinc finger protein 24 −1.21218
326 DGKE diacylglycerol kinase epsilon −1.21008
327 PTEN phosphatase and tensin homolog −1.20912
328 WWC1 WW and C2 domain containing 1 −1.20909
329 SFRP1 secreted frizzled-related protein 1 −1.20491
330 ABHD6 abhydrolase domain containing 6 −1.20477
331 NEU1 neuraminidase 1 (lysosomal sialidase) −1.19821
332 UBE2I ubiquitin conjugating enzyme E2I −1.19721
333 EBI3 Epstein-Barr virus induced 3 −1.19684
334 ZEB2 zinc finger E-box binding homeobox 2 −1.19545
335 MMP16 matrix metallopeptidase 16 −1.19347
336 CD93 CD93 molecule −1.19321
337 ANXA1 annexin A1 −1.19311
338 P4HA2 prolyl 4-hydroxylase, alpha polypeptide II −1.19140
339 ATP6V1C1 ATPase, H+ transporting, lysosomal 42kDa, V1 subunit C1 −1.19127
340 BBS4 Bardet-Biedl syndrome 4 −1.19104
341 SRPX2 sushi-repeat containing protein, X-linked 2 −1.19022
342 CXCR6 chemokine (C-X-C motif) receptor 6 −1.19013
343 TAC1 tachykinin precursor 1 −1.18419
344 FAM188A family with sequence similarity 188 member A −1.18353
345 NFKB1 nuclear factor of kappa light polypeptide gene enhancer in B-cells
1
−1.18152
346 BAG1 BCL2 associated athanogene 1 −1.17465
347 EHD1 EH domain containing 1 −1.17454
348 LIMA1 LIM domain and actin binding 1 −1.17448
349 MOV10L1 Mov10 RISC complex RNA helicase like 1 −1.17430
350 ADGRE5 adhesion G protein-coupled receptor E5 −1.17397
351 COPB2 coatomer protein complex subunit beta 2 (beta prime) −1.17383
352 NR3C1 nuclear receptor subfamily 3 group C member 1 −1.16742
353 CLEC11A C-type lectin domain family 11 member A −1.16735
354 PI4KB phosphatidylinositol 4-kinase, catalytic, beta −1.16700
355 ACP1 acid phosphatase 1, soluble −1.16609
356 C1QBP complement component 1, q subcomponent binding protein −1.16280
357 PTTG1 pituitary tumor-transforming 1 −1.16060
358 PRRX1 paired related homeobox 1 −1.15954
359 SMAD1 SMAD family member 1 −1.15936
360 ADM adrenomedullin −1.15071
361 NDRG2 NDRG family member 2 −1.14999
362 REST RE1-silencing transcription factor −1.14731
363 NINJ1 ninjurin 1 −1.14059
364 CDKN3 cyclin-dependent kinase inhibitor 3 −1.13962
365 CIB1 calcium and integrin binding 1 −1.13924
366 HSPD1 heat shock protein family D (Hsp60) member 1 −1.13652
367 MS4A4A membrane-spanning 4-domains subfamily A member 4A −1.13410
368 LGALS8 lectin, galactoside-binding, soluble, 8 −1.12928
369 VPS28 vacuolar protein sorting 28 homolog (S. cerevisiae) −1.12857
370 PODN podocan −1.12805
371 IL25 interleukin 25 −1.12250
372 TMPRSS4 transmembrane protease, serine 4 −1.12108
373 GRB2 growth factor receptor bound protein 2 −1.11159
374 PEX13 peroxisomal biogenesis factor 13 −1.11146
375 PLCL1 phospholipase C like 1 −1.10743
376 CD48 CD48 molecule −1.10057
377 ASTN1 astrotactin 1 −1.09980
378 IQUB IQ motif and ubiquitin domain containing −1.09258
379 RNASE2 ribonuclease, RNase A family, 2 (liver, eosinophil-derived
neurotoxin)
−1.08789
380 AQP4 aquaporin 4 −1.08643
381 PRKCB protein kinase C, beta −1.08474
382 SATB2 SATB homeobox 2 −1.07941
383 GRIA3 glutamate receptor, ionotropic, AMPA 3 −1.06982
384 CCDC39 coiled-coil domain containing 39 −1.06883
385 PECAM1 platelet/endothelial cell adhesion molecule 1 −1.06057

During the process of wound healing, cells at the wounded edge remodel their cytoskeleton to form polarized membrane protrusions such as lamellipodia and filopodia and move towards closing the wound (27,28). The effect of dexamethasone on corneal cell lamellipodia and filopodia has not been clearly defined. Ingenuity Pathway Analysis identified that 45 genes associated with lamellipodia were regulated by dexamethasone treatment (Figure 3E and S.Table1). Out of these 45 genes, dexamethasone treatment upregulated 19 genes (42.2%) and 26 genes (57.8%) were repressed. Lamellipodia gene network generated using Ingenuity Pathway Analysis suggested Epidermal Growth Factor Receptor (EGFR) as the most important regulator of lamellipodia formation in the presence of glucocorticoids. Cell surface glycoprotein CD44 and a guanine-nucleotide-exchange factor VAV1 are also suggested to be playing an important role in glucocorticoid-mediated changes to lamellipodia. In addition, we also found that 55 genes associated with filopodia were regulated by dexamethasone treatment (Figure 3F and S.Table2). Of these 55 genes, dexamethasone treatment upregulated 21 genes (38.2%) and 34 genes (61.8%) were repressed. Filopodia gene network suggested that several growth factors such as Vascular Endothelial Growth Factor (VEGF), Fibroblast Growth Factor 2 (FGF2) and Connective Tissue Growth Factor (CTGF) played a role in glucocorticoid-mediated changes to the filopodia. Also a part of wound healing is reestablishing epithelial integrity to maintain corneal epithelial barrier function. Therefore, we searched for genes involved in permeability using Ingenuity Pathway Analysis. Fifty genes involved in diseases and functions associated with permeability were regulated by dexamethasone (Figure 3G and S.Table3). Of these 50 genes, 16 genes were upregulated (32%) and 34 genes (68%) were repressed by dexamethasone. According to the Permeability Gene Network, the glucocorticoid receptor appears to be serving as the most active hub in regulating a large cohort of genes involved in permeability. Thus, glucocorticoid signaling is critical in regulating the genes associated in cell migration, cytoskeletal remodeling and permeability in human corneal epithelial cells.

3.4 Independent Validation of genes from the microarray

To independently validate the changes in gene expression observed by microarray, we measured glucocorticoid-regulated expression of four genes- Tumor necrosis factor receptor super family 11b (TNFRSF11b), Brain derived neurotropic factor (BDNF), Epiregulin (EREG) and Nerve Growth Factor (NGF) by real-time RT-PCR from HCE RNA samples that came from experiments independent from those employed in the microarray studies. For this purpose, HCE cells were treated for 6 hours with vehicle, dexamethasone (100nM) and/or RU486 (1000nM) (Figure 4). IPA identified these four genes to be involved in cell movement (Figure 3D). NGF was identified to be playing a role not only in migration of cells but also in regulating cytoskeleton and epithelial integrity (Figure 3 D-G). Consistent with the microarray results, TNFRSF11b, BDNF, EREG and NGF were repressed by glucocorticoids and this repression was blunted upon treating the cells with a combination of glucocorticoids and RU486 or with RU486 alone. These findings illustrate some of the ways by which glucocorticoids regulate corneal wound healing is by repressing the expression of genes involved in regulating cell movement, cytoskeleton rearrangement and maintenance of epithelial integrity.

Figure 4.

Figure 4

Validation of microarray results by RT-PCR. TNFRSF11b, BDNF, EREG and NGF mRNA levels measured by RT-PCR and normalized to PPIB mRNA level in HCE cells treated with vehicle (white bars) or 100 nM dexamethasone (light grey bars) or a combination of 100nM dexamethasone and 1000nM RU486 (dark grey bars) or 1000nM RU486 (black bars). n = 3 or 4 biological replicates; *p<0.05.

3.5 Glucocorticoids delay in vitro wound healing in HCE cells

To determine if the biological processes identified to be regulated by glucocorticoid by microarray analysis are functions involved in wound healing of HCE cells, we performed real-time wound healing scratch assays. Confluent monolayers of HCE cells were treated overnight with vehicle, dexamethasone (1000nM) or RU486 (10uM) or a combination of dexamethasone and RU486. Treated cells were scratched and images of the healing edges were taken every 30 mins for up to 30 hours. Wound closure was delayed with dexamethasone treatment (Figure 5A and Supplemental movie). Treatment with RU486 not only inhibited glucocorticoid-mediated delay, but also accelerated wound closure (Figure 5A and Supplemental movie). All treatment conditions, except dexamethasone revealed complete wound closure, which is represented in the images showing time-projection over 18 hours (Figure 5B). Quantification of the distance migrated by the wounded monolayer revealed that dexamethasone-treated monolayer migrated the least distance when compared to the other treatment conditions (Figure 5C). Consistently, the percent of the area of wound closure was decreased in dexamethasone treated HCE monolayer at the end of 18 hours (Figure 5D). Interestingly, proliferation and viability of HCE cells were not affected by glucocorticoid treatment (Supplemental Figure 1). These observations are consistent with the IPA analysis and demonstrate that glucocorticoid treatment indeed delays in vitro wound healing of HCE cell monolayer. Our findings indicate that this effect of delayed migration is mediated by the glucocorticoid receptor since this function can be rescued by treatment with glucocorticoid receptor antagonist RU486.

Figure 5.

Figure 5

Glucocorticoids delay in vitro wound healing of HCE cells. Scratch assay was performed on confluent monolayers of cells pre-treated overnight alone or in combination with vehicle, DEX (1000nM), or RU486 (10uM) in charcoal-stripped serum containing medium. Real-time analysis of cell migration was performed by imaging the every 30 minutes for 18 hours post-scratch. A) Representative images of wound healing kinetics in all the 4 conditions-vehicle, dexamethasone (1000nM), combination of dexamethasone (1000nM) and RU486 (10uM) and RU 486 (10uM) alone. Scratch width and time are on the X- and Y- axes, respectively. Yellow dots indicate the edge of the scratched monolayer. B) Representative images showing the time-projection of wound healing over a period of 18 hours in all four conditions. Time-projection is indicated with t0 in white and t18hrs in red. C) Quantification of the extent of wound healing measured from time-lapse images taken over a period of 30 hours. Average of four individual experiments is represented here (*p<0.001). D) Average area of wound closure in 18 hours represented in percentage from four individual experiments (*p<0.05). E) Representative images showing the net change in the area of the lamellipodia in vehicle and dexamethasone (1000nM) treated cells at 30 minutes after scratching the monolayer. Red represents lamellipodia that are moving forward to close the wound, green represents retraction of the lamellipodia and yellow represents no change in net movement of the lamellipodia over a period of approximately 6 minutes. Arrows are pointing to the filopodia. F) Average of the change in the area of the lamellipodia per minute in 10 minutes after creating a scratch wound in the monolayer in HCE cells treated overnight with either vehicle or 1000nM dexamethasone. Results from three experiments were averaged and are shown here. G) Average number of filopodia formed at the wounded monolayer between 10-65 minutes of wound healing. An average of three individual experiments is represented here.

3.6 Glucocorticoid treatment of HCE monolayer alters lamellipodia and filopodia formation

To understand if glucocorticoid-mediated regulation of cytoskeleton of HCE cells is contributing to the delay in migration of dexamethasone-treated cells, we evaluated the activity of lamellipodia along the wounded monolayer by quantifying the change in lamellipodia area in response to dexamethasone treatment. The data demonstrate that dexamethasone treatment decreases the activity of lamellipodia as seen by decrease in the change in lamellipodia area (less region in red in Figure 5E) compared to the vehicle treated cells (Figure 5 E and F). Additionally, quantification of the number of filopodia generated by “leader cells” along the wounded monolayer of HCE cells treated with glucocorticoids revealed fewer filopodia compared to the vehicle-treated cells (arrows in Figure 5E, and Figure 5G). These changes observed in the cytoskeleton of glucocorticoid-pre-treated cells within minutes after creating a scratch wound are indicative of a slowly migrating monolayer.

3.7 Glucocorticoids improve tight-junction protein organization and enhance barrier function

Based on the effects of glucocorticoids on migration of HCE cells, we wished to determine the role of GR on basal epithelial permeability and barrier function. We treated subconfluent cultures of HCE cells with vehicle or 100nM dexamethasone for either 6 hours or 24 hours and stained fixed cells for zonula occludens 1 (ZO-1), a protein that associates with the tight junctions on epithelial cells. Glucocorticoid treatment for as little as 6 hours had a strong recruitment of ZO-1 along the plasma membrane compared to the vehicle-treated cells. Subsequently, 24hour treatment of subconfluent cultures with dexamethasone resulted in greater ZO-1 distribution along the plasma membrane, compared to the vehicle treatment but not as robust as seen in cells treated with dexamethasone for 6 hours. The explanation for this observation is possibly due to the subconfluent culture continuing to establish cell-to-cell connections by reorganizing junctional proteins while growing to reach maximum confluency. A representative plasma membrane profile of ZO-1 staining intensity shows a peak in the intensity in the dexamethasone treated cells at both 6hr and 24hr treatment conditions (Figure 6A). Consistent with glucocorticoids influencing ZO-1 localization to the plasma membrane to form organized tight-junctions, the results from the permeability assay indicated that dexamethasone-treated cells formed a tighter epithelial barrier, thus allowing significantly lower amount of FITC dextran to permeate through the monolayer than the vehicle-treated cells (Figure 6B). These data indicate that glucocorticoids regulate epithelial tight junction proteins in subconfluent as well as confluent cultures to enhance epithelial barrier function.

Figure 6.

Figure 6

Glucocorticoids regulate ZO-1 distribution and epithelial permeability. A) Immunofluorescence of subconfluent cultures of HCE cells exhibiting ZO-1 distribution in vehicle or 1uM dexamethasone for either 6 hours or 24 hours. Asterisks indicate site of disorganized ZO-1 staining. Images are maximum intensity projections of Z-stacks imaged through the entire depth of the cell membrane. Images are a merge of Hoechst staining (blue) and ZO-1 staining (green). A representative plasma membrane profile of ZO-1 staining intensity shows a peak in the intensity in the dexamethasone treated cells (black line) at both 6hr and 24hr treatment conditions. B) Relative changes in permeability were determined by measuring the fluorescence unit of FITC dextran that permeated through the epithelial monolayer in cells treated for 24hours either with vehicle or 100nM dexamethasone. An average of three independent experiments is represented here. **p value < 0.01.

4. DISCUSSION

Corticosteroids are widely used by ophthalmologists to treat various conditions of the cornea, but very little is known about their cell type specific actions in this tissue. In this study, we determined the expression pattern of the glucocorticoid receptor expression in an adult mouse cornea and we characterized the glucocorticoid receptor system using a human corneal epithelial cell line. Whole genome array results reveal an intricate dialogue between dexamethasone and HCE cells. We provide comprehensive analyses of the cellular and biological processes in corneal epithelial cells mediated by glucocorticoid treatment. The glucocorticoid transcriptome in HCE cells can serve as an important resource to the research community where it can be used to identify the targets to maximize the benefits and minimize the adverse affects of corticosteroid therapy. It is very important to note that our results were obtained using an immortalized human corneal epithelial cell line, which has been a tool widely used in ophthalmology for many years. However, recent studies have raised concerns regarding the differences between these immortalized HCE cells and primary human corneal epithelial cells in factors including inflammatory response (29), expression of atypical cytokeratins (30), purity of cell population (31), and the genomic content (31,32). Therefore, results from our in vitro studies may not precisely reflect the actions of glucocorticoids in primary human corneal epithelial cells.

Several of the genes altered in the microarray dataset have not been previously known to regulate cell movement in the various cell types of ocular tissues. For example, TSC22D3 or GILZ is one of the most upregulated genes in the microarray dataset that have been previously reported to inhibit migration of leukocytes (33). GILZ gene expression has been shown to be induced by dexamethasone in the whole eye of a mouse (34), and in cultured primary human lens epithelial cells (35), but its expression or its role in the cornea has not been explored. Tumor necrosis factor receptor superfamily, member 11b (TNFRSF11b), a promoter of migration (36), is one of the most-repressed genes in this microarray dataset, which was reported to be expressed in corneal stroma (37), however its regulation by glucocorticoids in the cornea has never been reported. Comparison of the number of upregulated genes versus the downregulated genes from the microarray dataset suggested that glucocorticoid regulation of wound healing skewed towards downregulation of genes involved in promoting migration. An analysis using HCE cells to validate microarray results independent from the cells used for the global gene expression studies reveals that mRNA of TNFRSF11b, BDNF, EREG and NGF were indeed repressed by glucocorticoids and this repression was abolished by antagonism of the glucocorticoid receptor by the GR antagonist-RU486. TNFRSF11b (also known as Osteoprotegerin), a member of the TNF receptor super family is a secreted decoy receptor that has been associated with increase in bone density as a result of decrease in osteoclast-mediated bone resorption (38). Brain derived neurotropic factor (BDNF) is a member of the neurotrophin gene family with established roles in neuronal development and survival (39). Although BDNF’s role in neurogenesis and cell survival has been well characterized (40), there are only a few studies investigating the function of BDNF in the cornea. For example, the presence of BDNF mRNA in the human cornea is potentially associated with proliferation of corneal epithelial cells, suggesting an important role for BDNF in corneal function (41). Repression of BDNF gene expression is perhaps a novel mechanism exerted by glucocorticoids to regulate cell migration. Epiregulin (EREG) is a recently identified member of the epidermal growth factor family of ligands with functions in inflammation and wound healing (42). In mice, EREG is also known to play a critical role in corneal wound healing (43). A potential therapeutic strategy of co-administering glucocorticoids and epiregulin might offer a beneficial outcome in cases of corneal injury. Nerve growth factor (NGF) is the first-discovered member of the neurotrophin gene family, which is involved in the growth and survival of nerves (44). NGF in the cornea has been thought to play a role in promoting corneal nerve regeneration (45). In the context of wound healing, NGF is known to accelerate wound healing by promoting cell cycle progression and by increasing cell migration (46,47). A more recent study reports that NGF may have a negative impact on wound recovery by inhibiting regression of corneal lymphatic vessels and increasing the nerve density, thereby increasing sensitivity to pain (48). Glucocorticoids suppressing NGF during corneal wound healing could lead to be a better recovery of the wound, and with less pain. Interestingly, IPA identified NGF to be playing a role in cell movement, cytoskeletal reorganization and maintenance of epithelial barrier suggesting that glucocorticoids can modulate multiple biological processes driving wound healing by altering the expression of a single gene.

Corneal injuries are the most common cases of eye injury. Corneal wound healing is a complex physiological process that is modulated by a number of signaling pathways. Maintaining a protective barrier is one of the crucial functions of the corneal epithelium. We have investigated the effect of glucocorticoids on corneal wound healing in an in vitro model. This study demonstrates that glucocorticoids inhibit wound healing of human corneal epithelial cells by altering the activity of membrane lamellipodia and filopodia, together blunting the rate of migration. We also demonstrate that glucocorticoids enhance epithelial integrity by altering tight-junction protein distribution. Our data indicate that glucocorticoid-induced improvement of barrier function in subconfluent cultures can be relevant to the remodeling of a wounded epithelium. The results suggest that wound healing in the presence of glucocorticoids may result in a wound that heals at a slower rate, with improved epithelial integrity. Because epithelial integrity is an essential function in maintaining corneal homeostasis, delayed wound healing as an adverse outcome of glucocorticoid therapy does not seem entirely adverse since glucocorticoids promote improved tight junction integrity and thereby the enhanced epithelial barrier function.

Supplementary Material

1

Supplemental Figure 1: HCE cells treated with vehicle or dexamethasone (100nM and 1000nM) for 24, 48 or 72 hours and cells were counted at the end of each time-point either by hemocytometer or by flow cytometry. A) Viable cells in vehicle treated and dexamethasone (Dex) treated samples at the indicated time-points. B) Dead cells (trypan blue positive) in vehicle-treated and dexamethasone treated samples at the indicated time-points. Vehicle (light grey circles), 100nM (dark grey circles) and 1000nM (black circles) of dexamethasone; Average of 4 independent experiments is represented here. C) Percent of viable cells, D) percent of propidium iodide (PI) positive cells determined by flow cytometry; Vehicle (light grey bar), 100nM (dark grey bar) and 1000nM (black bar) of dexamethasone; Average of 3 independent experiments is represented here.

2
Download video file (3.4MB, mp4)

HIGHLIGHTS.

  • Functional glucocorticoid receptors are expressed in mouse corneas.

  • Glucocorticoids regulated over 4000 genes in human corneal epithelial cells.

  • Glucocorticoids enriched genes associated in wound healing processes.

  • Glucocorticoids decreased cell migration rate but enhanced epithelial integrity.

ACKNOWLEDGMENT

We thank Jeff Tucker from the NIEHS Fluorescence Microscopy and Imaging Center for his assistance in the real-time wound healing studies. We also thank the NIEHS Flow Cytometry Center. We thank Drs. Robert Oakley, Xiaoling Li and Harriet Kinyamu for their critical reading of the manuscript. This work was funded by the NIEHS Intramural Research Program.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Disclosure Statement: The authors have nothing to disclose.

REFERENCES

  • 1.Ren R, Oakley RH, Cruz-Topete D, Cidlowski JA. Dual role for glucocorticoids in cardiomyocyte hypertrophy and apoptosis. Endocrinology. 2012;153:5346–5360. doi: 10.1210/en.2012-1563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Gruver-Yates AL, Quinn MA, Cidlowski JA. Analysis of glucocorticoid receptors and their apoptotic response to dexamethasone in male murine B cells during development. Endocrinology. 2014;155:463–474. doi: 10.1210/en.2013-1473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Chang JH, Gabison EE, Kato T, Azar DT. Corneal neovascularization. Current opinion in ophthalmology. 2001;12:242–249. doi: 10.1097/00055735-200108000-00002. [DOI] [PubMed] [Google Scholar]
  • 4.Lambiase A, Abdolrahimzadeh S, Recupero SM. An update on intravitreal implants in use for eye disorders. Drugs of today. 2014;50:239–249. doi: 10.1358/dot.2014.50.3.2103755. [DOI] [PubMed] [Google Scholar]
  • 5.Xie X, Yan X, Lin Z, Jin X. Differential effects of low- and high-dose glucocorticoids on the innate immunity of corneal epithelium in vitro. Ocul Immunol Inflamm. 2011;19:275–281. doi: 10.3109/09273948.2011.569110. [DOI] [PubMed] [Google Scholar]
  • 6.Susarla R, Liu L, Walker EA, Bujalska IJ, Alsalem J, Williams GP, Sreekantam S, Taylor AE, Tallouzi M, Southworth HS, Murray PI, Wallace GR, Rauz S. Cortisol biosynthesis in the human ocular surface innate immune response. PLoS One. 2014;9:e94913. doi: 10.1371/journal.pone.0094913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Mirabelli P, Peebo BB, Xeroudaki M, Koulikovska M, Lagali N. Early effects of dexamethasone and anti-VEGF therapy in an inflammatory corneal neovascularization model. Experimental eye research. 2014;125:118–127. doi: 10.1016/j.exer.2014.06.006. [DOI] [PubMed] [Google Scholar]
  • 8.Cardoso SS, Ferreira AL. Effect of adrenalectomy and of dexamethasone upon circadian distribution of mitosis in the cornea of rats. I. Proc Soc Exp Biol Med. 1967;125:1254–1259. doi: 10.3181/00379727-125-32329. [DOI] [PubMed] [Google Scholar]
  • 9.Pezuk P, Mohawk JA, Wang LA, Menaker M. Glucocorticoids as entraining signals for peripheral circadian oscillators. Endocrinology. 2012;153:4775–4783. doi: 10.1210/en.2012-1486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ellenberg D, Azar DT, Hallak JA, Tobaigy F, Han KY, Jain S, Zhou Z, Chang JH. Novel aspects of corneal angiogenic and lymphangiogenic privilege. Progress in retinal and eye research. 2010;29:208–248. doi: 10.1016/j.preteyeres.2010.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Steele MM, Kelley PM, Schieler AM, Tempero RM. Glucocorticoids suppress corneal lymphangiogenesis. Cornea. 2011;30:1442–1447. doi: 10.1097/ICO.0b013e318213f39f. [DOI] [PubMed] [Google Scholar]
  • 12.Saud EE, Moraes HV, Jr., Marculino LG, Gomes JA, Allodi S, Miguel NC. Clinical and histopathological outcomes of subconjunctival triamcinolone injection for the treatment of acute ocular alkali burn in rabbits. Cornea. 2012;31:181–187. doi: 10.1097/ICO.0b013e318221ce99. [DOI] [PubMed] [Google Scholar]
  • 13.A double-masked, placebo-controlled evaluation of 0.5% loteprednol etabonate in the treatment of postoperative inflammation. The Loteprednol Etabonate Postoperative Inflammation Study Group 2. Ophthalmology. 1998;105:1780–1786. doi: 10.1016/s0161-6420(98)99054-6. [DOI] [PubMed] [Google Scholar]
  • 14.Bron A, Denis P, Hoang-Xuan TC, Boureau-Andrieux C, Crozafon P, Hachet E, Medhorn E, Akingbehin A. The effects of Rimexolone 1% in postoperative inflammation after cataract extraction. A double-masked placebo-controlled study. European journal of ophthalmology. 1998;8:16–21. doi: 10.1177/112067219800800105. [DOI] [PubMed] [Google Scholar]
  • 15.Bielory L. Ocular allergy treatment. Immunology and allergy clinics of North America. 2008;28:189–224. doi: 10.1016/j.iac.2007.12.001. vii. [DOI] [PubMed] [Google Scholar]
  • 16.Kimura K, Teranishi S, Kawamoto K, Nishida T. Protective effect of dexamethasone against hypoxia-induced disruption of barrier function in human corneal epithelial cells. Experimental eye research. 2011;92:388–393. doi: 10.1016/j.exer.2011.02.013. [DOI] [PubMed] [Google Scholar]
  • 17.Petroutsos G, Guimaraes R, Giraud JP, Pouliquen Y. Corticosteroids and corneal epithelial wound healing. The British journal of ophthalmology. 1982;66:705–708. doi: 10.1136/bjo.66.11.705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Araki-Sasaki K, Ohashi Y, Sasabe T, Hayashi K, Watanabe H, Tano Y, Handa H. An SV40-immortalized human corneal epithelial cell line and its characterization. Investigative ophthalmology & visual science. 1995;36:614–621. [PubMed] [Google Scholar]
  • 19.Cidlowski JA, Bellingham DL, Powell-Oliver FE, Lubahn DB, Sar M. Novel antipeptide antibodies to the human glucocorticoid receptor: recognition of multiple receptor forms in vitro and distinct localization of cytoplasmic and nuclear receptors. Molecular endocrinology. 1990;4:1427–1437. doi: 10.1210/mend-4-10-1427. [DOI] [PubMed] [Google Scholar]
  • 20.Seo KY, Chung SH, Lee JH, Park MY, Kim EK. Regulation of membrane-associated mucins in the human corneal epithelial cells by dexamethasone. Cornea. 2007;26:709–714. doi: 10.1097/ICO.0b013e31804f5a09. [DOI] [PubMed] [Google Scholar]
  • 21.Xie X, Yan X, Lin Z, Jin X. Differential effects of low- and high-dose glucocorticoids on the innate immunity of corneal epithelium in vitro. Ocular immunology and inflammation. 2011;19:275–281. doi: 10.3109/09273948.2011.569110. [DOI] [PubMed] [Google Scholar]
  • 22.Duma D, Collins JB, Chou JW, Cidlowski JA. Sexually dimorphic actions of glucocorticoids provide a link to inflammatory diseases with gender differences in prevalence. Science signaling. 2010;3:ra74. doi: 10.1126/scisignal.2001077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Oakley RH, Ren R, Cruz-Topete D, Bird GS, Myers PH, Boyle MC, Schneider MD, Willis MS, Cidlowski JA. Essential role of stress hormone signaling in cardiomyocytes for the prevention of heart disease. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:17035–17040. doi: 10.1073/pnas.1302546110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Revollo JR, Oakley RH, Lu NZ, Kadmiel M, Gandhavadi M, Cidlowski JA. HES1 is a master regulator of glucocorticoid receptor-dependent gene expression. Science signaling. 2013;6:ra103. doi: 10.1126/scisignal.2004389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Liu L, Walker EA, Kissane S, Khan I, Murray PI, Rauz S, Wallace GR. Gene expression and miR profiles of human corneal fibroblasts in response to dexamethasone. Investigative ophthalmology & visual science. 2011;52:7282–7288. doi: 10.1167/iovs.11-7463. [DOI] [PubMed] [Google Scholar]
  • 26.McGwin G, Jr., Owsley C. Incidence of emergency department-treated eye injury in the United States. Archives of ophthalmology. 2005;123:662–666. doi: 10.1001/archopht.123.5.662. [DOI] [PubMed] [Google Scholar]
  • 27.Poujade M, Grasland-Mongrain E, Hertzog A, Jouanneau J, Chavrier P, Ladoux B, Buguin A, Silberzan P. Collective migration of an epithelial monolayer in response to a model wound. Proceedings of the National Academy of Sciences of the United States of America. 2007;104:15988–15993. doi: 10.1073/pnas.0705062104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Omelchenko T, Vasiliev JM, Gelfand IM, Feder HH, Bonder EM. Rho-dependent formation of epithelial "leader" cells during wound healing. Proceedings of the National Academy of Sciences of the United States of America. 2003;100:10788–10793. doi: 10.1073/pnas.1834401100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Suzuki T, Sullivan DA. Comparative effects of estrogen on matrix metalloproteinases and cytokines in immortalized and primary human corneal epithelial cell cultures. Cornea. 2006;25:454–459. doi: 10.1097/01.ico.0000220777.70981.46. [DOI] [PubMed] [Google Scholar]
  • 30.Huhtala A, Nurmi SK, Tahti H, Salminen L, Alajuuma P, Rantala I, Helin H, Uusitalo H. The immunohistochemical characterisation of an SV40-immortalised human corneal epithelial cell line. Altern Lab Anim. 2003;31:409–417. doi: 10.1177/026119290303100407. [DOI] [PubMed] [Google Scholar]
  • 31.Yamasaki K, Kawasaki S, Young RD, Fukuoka H, Tanioka H, Nakatsukasa M, Quantock AJ, Kinoshita S. Genomic aberrations and cellular heterogeneity in SV40-immortalized human corneal epithelial cells. Invest Ophthalmol Vis Sci. 2009;50:604–613. doi: 10.1167/iovs.08-2239. [DOI] [PubMed] [Google Scholar]
  • 32.Greco D, Vellonen KS, Turner HC, Hakli M, Tervo T, Auvinen P, Wolosin JM, Urtti A. Gene expression analysis in SV-40 immortalized human corneal epithelial cells cultured with an air-liquid interface. Mol Vis. 2010;16:2109–2120. [PMC free article] [PubMed] [Google Scholar]
  • 33.Mazzon E, Bruscoli S, Galuppo M, Biagioli M, Sorcini D, Bereshchenko O, Fiorucci C, Migliorati G, Bramanti P, Riccardi C. Glucocorticoid-induced leucine zipper (GILZ) controls inflammation and tissue damage after spinal cord injury. CNS neuroscience & therapeutics. 2014;20:973–981. doi: 10.1111/cns.12315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Cari L, Ricci E, Gentili M, Petrillo MG, Ayroldi E, Ronchetti S, Nocentini G, Riccardi C. A focused Real Time PCR strategy to determine GILZ expression in mouse tissues. Results Immunol. 2015;5:37–42. doi: 10.1016/j.rinim.2015.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Gupta V, Galante A, Soteropoulos P, Guo S, Wagner BJ. Global gene profiling reveals novel glucocorticoid induced changes in gene expression of human lens epithelial cells. Mol Vis. 2005;11:1018–1040. [PubMed] [Google Scholar]
  • 36.Kobayashi-Sakamoto M, Isogai E, Hirose K, Chiba I. Role of alphav integrin in osteoprotegerin-induced endothelial cell migration and proliferation. Microvascular research. 2008;76:139–144. doi: 10.1016/j.mvr.2008.06.004. [DOI] [PubMed] [Google Scholar]
  • 37.Wilson SE, Mohan RR, Netto M, Perez V, Possin D, Huang J, Kwon R, Alekseev A, Rodriguez-Perez JP. RANK, RANKL, OPG, and M-CSF expression in stromal cells during corneal wound healing. Investigative ophthalmology & visual science. 2004;45:2201–2211. doi: 10.1167/iovs.03-1162. [DOI] [PubMed] [Google Scholar]
  • 38.Simonet WS, Lacey DL, Dunstan CR, Kelley M, Chang MS, Luthy R, Nguyen HQ, Wooden S, Bennett L, Boone T, Shimamoto G, DeRose M, Elliott R, Colombero A, Tan HL, Trail G, Sullivan J, Davy E, Bucay N, Renshaw-Gegg L, Hughes TM, Hill D, Pattison W, Campbell P, Sander S, Van G, Tarpley J, Derby P, Lee R, Boyle WJ. Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell. 1997;89:309–319. doi: 10.1016/s0092-8674(00)80209-3. [DOI] [PubMed] [Google Scholar]
  • 39.Bhave SV, Ghoda L, Hoffman PL. Brain-derived neurotrophic factor mediates the anti-apoptotic effect of NMDA in cerebellar granule neurons: signal transduction cascades and site of ethanol action. J Neurosci. 1999;19:3277–3286. doi: 10.1523/JNEUROSCI.19-09-03277.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Chao MV, Rajagopal R, Lee FS. Neurotrophin signalling in health and disease. Clinical science. 2006;110:167–173. doi: 10.1042/CS20050163. [DOI] [PubMed] [Google Scholar]
  • 41.You L, Kruse FE, Volcker HE. Neurotrophic factors in the human cornea. Investigative ophthalmology & visual science. 2000;41:692–702. [PubMed] [Google Scholar]
  • 42.Riese DJ, 2nd, Cullum RL. Epiregulin: roles in normal physiology and cancer. Semin Cell Dev Biol. 2014;28:49–56. doi: 10.1016/j.semcdb.2014.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Zhang Y, Kobayashi T, Hayashi Y, Yoshioka R, Shiraishi A, Shirasawa S, Higashiyama S, Ohashi Y. Important role of epiregulin in inflammatory responses during corneal epithelial wound healing. Investigative ophthalmology & visual science. 2012;53:2414–2423. doi: 10.1167/iovs.11-8869. [DOI] [PubMed] [Google Scholar]
  • 44.Cohen S, Levi-Montalcini R, Hamburger V. A Nerve Growth-Stimulating Factor Isolated from Sarcom as 37 and 180. Proc Natl Acad Sci U S A. 1954;40:1014–1018. doi: 10.1073/pnas.40.10.1014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Sarkar J, Chaudhary S, Jassim SH, Ozturk O, Chamon W, Ganesh B, Tibrewal S, Gandhi S, Byun YS, Hallak J, Mahmud DL, Mahmud N, Rondelli D, Jain S. CD11b+GR1+ myeloid cells secrete NGF and promote trigeminal ganglion neurite growth: implications for corneal nerve regeneration. Invest Ophthalmol Vis Sci. 2013;54:5920–5936. doi: 10.1167/iovs.13-12237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hong J, Qian T, Le Q, Sun X, Wu J, Chen J, Yu X, Xu J. NGF promotes cell cycle progression by regulating D-type cyclins via PI3K/Akt and MAPK/Erk activation in human corneal epithelial cells. Mol Vis. 2012;18:758–764. [PMC free article] [PubMed] [Google Scholar]
  • 47.Blanco-Mezquita T, Martinez-Garcia C, Proenca R, Zieske JD, Bonini S, Lambiase A, Merayo-Lloves J. Nerve growth factor promotes corneal epithelial migration by enhancing expression of matrix metalloprotease-9. Invest Ophthalmol Vis Sci. 2013;54:3880–3890. doi: 10.1167/iovs.12-10816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Fink DM, Connor AL, Kelley PM, Steele MM, Hollingsworth MA, Tempero RM. Nerve growth factor regulates neurolymphatic remodeling during corneal inflammation and resolution. PLoS One. 2014;9:e112737. doi: 10.1371/journal.pone.0112737. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

Supplemental Figure 1: HCE cells treated with vehicle or dexamethasone (100nM and 1000nM) for 24, 48 or 72 hours and cells were counted at the end of each time-point either by hemocytometer or by flow cytometry. A) Viable cells in vehicle treated and dexamethasone (Dex) treated samples at the indicated time-points. B) Dead cells (trypan blue positive) in vehicle-treated and dexamethasone treated samples at the indicated time-points. Vehicle (light grey circles), 100nM (dark grey circles) and 1000nM (black circles) of dexamethasone; Average of 4 independent experiments is represented here. C) Percent of viable cells, D) percent of propidium iodide (PI) positive cells determined by flow cytometry; Vehicle (light grey bar), 100nM (dark grey bar) and 1000nM (black bar) of dexamethasone; Average of 3 independent experiments is represented here.

2
Download video file (3.4MB, mp4)

RESOURCES