Investigative Ophthalmology & Visual Science Cover Image for Volume 44, Issue 2
February 2003
Volume 44, Issue 2
Free
Biochemistry and Molecular Biology  |   February 2003
Tissue Differential Microarray Analysis of Dexamethasone Induction Reveals Potential Mechanisms of Steroid Glaucoma
Author Affiliations
  • Wayne R. Lo
    From the Departments of Ophthalmology and
  • Laura Leigh Rowlette
    From the Departments of Ophthalmology and
  • Montserrat Caballero
    From the Departments of Ophthalmology and
  • Ping Yang
    Departments of Ophthalmology and Visual Sciences and
  • M. Rosario Hernandez
    Departments of Ophthalmology and Visual Sciences and
    Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri.
  • Teresa Borrás
    From the Departments of Ophthalmology and
    Genetics, Duke University Medical Center, Durham, North Carolina; and the
Investigative Ophthalmology & Visual Science February 2003, Vol.44, 473-485. doi:https://doi.org/10.1167/iovs.02-0444
  • Views
  • PDF
  • Share
  • Tools
    • Alerts
      ×
      This feature is available to authenticated users only.
      Sign In or Create an Account ×
    • Get Citation

      Wayne R. Lo, Laura Leigh Rowlette, Montserrat Caballero, Ping Yang, M. Rosario Hernandez, Teresa Borrás; Tissue Differential Microarray Analysis of Dexamethasone Induction Reveals Potential Mechanisms of Steroid Glaucoma. Invest. Ophthalmol. Vis. Sci. 2003;44(2):473-485. https://doi.org/10.1167/iovs.02-0444.

      Download citation file:


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

      ×
  • Supplements
Abstract

purpose. To identify myocilin (TIGR/MYOC) properties that are specific to the human trabecular meshwork (HTM). To search for genes highly expressed in dexamethasone (DEX)-induced HTM cells that are barely expressed or absent in DEX-induced cells from other tissues.

methods. TIGR/MYOC induction by DEX (10−7 M for 8–10 days) was analyzed by Northern and Western blot analyses in HTM, human umbilical vein endothelial cells, HeLa cells, and human embryonic skeletal muscle cells and optic nerve head (ONH) astrocytes at confluence. Processing and secretion were analyzed after the cells were infected with adenoviruses overexpressing wild-type and mutant forms of TIGR/MYOC. Affymetrix U95Av2 GeneChips (n = 6) and software were used to compare paired expression profiles of HTM, HTM-DEX, ONH astrocytes, and ONH astrocytes-DEX. Identification of HTM-DEX–specific genes (compared with ONH astrocytes-DEX) was performed by selecting genes with the highest fold change values (≥20). Genes with fold change values of four or more were matched with loci linked to glaucoma, by using gene databases.

results. TIGR/MYOC induction by DEX occurred only in HTM cells. Secretory and glycosylation characteristics remained the same across cell types. Expression profile analysis revealed multiple genes differentially upregulated in HTM-DEX including, in addition to TIGR/MYOC, a serine protease inhibitor (α1-antichymotrypsin), a neuroprotective factor (pigment epithelium-derived factor), an antiangiogenesis factor (cornea-derived transcript 6), and a prostaglandin synthase (prostaglandin D2 synthase). Fifteen of the 249 genes with fold change values of four or more mapped to glaucoma-linked loci.

conclusions. The induction of TIGR/MYOC by DEX is HTM-specific, whereas its secretory and glycosylation characteristics are ubiquitous. The known functions of HTM-DEX–specific genes reveal the presence of protective and damaging mechanisms for regulation of IOP during DEX treatment. Besides TIGR/MYOC, other HTM-DEX–specific genes may be good candidates for linkage to glaucoma.

The glaucomas are a group of diseases characterized by progressive degeneration of the optic nerve, usually accompanied by elevated intraocular pressure (IOP). They affect 70 million people worldwide and are the leading cause of blindness among African Americans. 1 Recently, mutations in TIGR/MYOC, also known as trabecular meshwork inducible glucocorticoid response protein (TIGR), have been linked to several types of glaucoma. 2 3 4 Although the function of this gene is currently unknown, genetic linkage analysis has demonstrated the presence of more than 25 distinct mutations in families with the most prevalent form of glaucoma, primary open-angle glaucoma (POAG), and in those with a rare, early-onset form, juvenile open-angle glaucoma (JOAG). 5  
TIGR/MYOC was identified as a protein upregulated in the HTM after long-term treatment with DEX 6 7 8 and was independently discovered in ciliary body 9 and normal human retina. 10 Some investigators believe its induction by DEX correlates with the time course and development of steroid-induced glaucoma, 8 11 a condition occurring in 30% to 40% of patients who are treated with steroids. 12 Because steroid-induced glaucoma mimics many aspects of POAG, it may be an important model for study of the disease. Mutations in TIGR/MYOC, however, are not implicated in causing or increasing susceptibility to steroid-induced glaucoma. 13 14  
TIGR/MYOC is a 504-amino-acid secretory protein translated from three exons. The amino terminal domain contains N- and O-glycosylation sites and a leucine zipper, and it is 25% homologous to the heavy chain of myosin. TIGR/MYOC is glycosylated 8 15 and the glycosylated and nonglycosylated protein forms migrate at approximately 57 and 55 kDa, respectively, on SDS-PAGE. 15  
Most glaucoma-linked mutations in TIGR/MYOC are located at its C-terminal end, in the region known as the olfactomedin (OLF) domain because of its 40% homology to olfactomedin, an extracellular matrix (ECM) protein first described in bullfrog olfactory epithelium. 16 Recent studies have demonstrated that many missense and nonsense mutations affecting the OLF domain impair secretion of the TIGR/MYOC mutant protein. 17 18 The impairment of secretion appears to be due to an inefficient processing of the mutant protein that results in misfolding and accumulation of aggregates inside the cell. 15 These results have lead us to postulate that the association of TIGR/MYOC mutants with glaucoma may be through a gain of deleterious function, 15 an idea supported by the findings that heterozygous and homozygous Myoc-knockout mice are viable and fertile, have normal IOP, and exhibit normal ocular morphology. 19  
The expression of TIGR/MYOC may be under the control of its 5-kb promoter region, which contains 13 predicted hormone-response elements and includes several glucocorticoid-response elements (GREs). 6 8 20 It has also been reported that multiple GRE half-sites exist within the 1900-bp region upstream of the putative translation start site. 21 Some of these half-sites have been described in systems in which there is delayed induction of glucocorticoid-induced gene expression. 22 It has been proposed that the delayed induction of TIGR/MYOC could be the result of classic GREs far upstream, the nearer GRE half-sites, or a secondary response to steroids. 21 More recent studies examining the promoter’s effects in luciferase reporter gene assay systems show that certain regions of putative GREs are not responsive to treatment with DEX 20 and that TIGR/MYOC is a delayed glucocorticoid-responsive gene in which the glucocorticoid receptor indirectly affects expression. 23 Thus, the described GREs in the promoter may not be directly responsible for upregulation of DEX, and distinct factors specifically present in DEX-induced HTM may first be required to stimulate expression of TIGR/MYOC. To date, these factors have not been described. 
TIGR/MYOC is highly expressed and secreted in the TM but is also found in many other tissues of the body, including skeletal muscle, retina, 9 and ONH astrocytes. 24 25 However, mutations in TIGR/MYOC are only linked to glaucoma and do not seem to affect the function of any other organ. This fact may be an indication that TIGR/MYOC serves TM-specific functions and/or has TM-specific properties that could be integral to glaucoma pathophysiology. In this study, we compared TIGR/MYOC expression, induction, secretion, and posttranslational processing in several selected human cell types. In an effort to identify TM glucocorticoid factors that either directly modulate IOP or affect TIGR/MYOC levels only in the TM, we used gene array technology. To select against genes upregulated by DEX in non-TM tissues, we compared the expression profile of HTM-DEX cells with another DEX-induced cell line, ONH astrocytes. Our choice of ONH astrocytes was based on their relevance in degeneration of the ONH in glaucoma, 26 the presence of detectable levels of TIGR/MYOC in vivo, 24 and our own result of the absence of DEX-induced TIGR/MYOC in these cells. In addition, because of the delayed glucocorticoid response of TIGR/MYOC, we chose to compare DEX induction after longer periods of drug treatment. 
Materials and Methods
Cell Culture
To examine the properties of TIGR/MYOC in different cell types, we chose several human cell lines based on their characteristics. HTM cells were used as a positive point of reference. Human umbilical vein endothelial cells (HUVECs) were chosen for their endothelial nature and because they are well described in the literature. Human embryonic skeletal muscle cells (SkMCs) and ONH astrocytes were chosen as representatives of other tissues with previously reported TIGR/MYOC expression. 9 27 28 HeLa cells were used as an unrelated, transformed cell line. 
The procedures in this study adhered to the provisions of the Declaration of Helsinki for research in human tissue. HTM cell lines used in these experiments were derived from two different donors with no history of glaucoma (HTM40 and HTM41). Primary cultures were established from donor eyes obtained from national eye banks within 48 hours of death. The TM from each of the donor eyes was isolated from surrounding tissue with incisions both anterior and posterior to the meshwork and treated with collagenase, as previously described. 29 Briefly, the explanted tissue was placed on 2% gelatin-coated 35-mm dishes and improved minimal essential medium (IMEM; Biofluids, Rockville, MD) supplemented with 20% fetal bovine serum (FBS) (Gibco BRL, Rockville, MD) and 50 μg/mL gentamicin (Gibco BRL). Cells that grew from digested tissue were first-passaged 1:4 at 2 to 3 weeks after explantation and subsequently propagated in the same medium with 10% serum. Each cell line survived for approximately 8 to 10 passages. For the current experiments, HTM cells were used at passages 4 to 6. HUVECs and HeLa cells were obtained from the American Type Culture Collection (ATCC, Manassas, VA). Human embryonic SkMCs were obtained from Clonetics (Walkersville, MD). HUVECs were maintained in endothelial cell medium EBM-2 basal medium with EGM-2 SingleQuots supplements and growth factors from Clonetics (Walkersville, MD). HeLa cells were maintained in IMEM supplemented with 10% FBS, 50 μg/mL gentamicin, and 100 μM nonessential amino acid solution (Gibco BRL). SkMCs were maintained in the provider’s medium (Clonetics). 
ONH astrocytes (line 00-1LLS) were grown and characterized from two normal eyes (aged 15 years) with no history of eye disease, diabetes, or neurodegenerative disease, as previously described in detail. 30 31 Briefly, four explants from each human lamina cribrosa were dissected and freed from sclera and surrounding tissues. Each explant was placed into a 25-cm2 tissue culture flask (Primaria; Falcon-BD Biosciences, Lincoln Park, NJ) and maintained in Dulbecco’s modified Eagle’s medium (DMEM)/Ham’s F-12 (Washington University Medical School Tissue Culture Support Center, St. Louis, MO) supplemented with 10% FBS (BioWhittaker, Walkersville, MD) and PSFM (10,000 U/mL penicillin, 10,000 μg/mL streptomycin, and 25 μg/mL amphotericin B; Gibco BRL). Cells were kept in a 37°C, 5% CO2 incubator. After 2 to 4 weeks, using a modified immunopanning procedure described by Mi and Barres, 32 we obtained purified primary cultures. To select ONH (type 1B) astrocytes, cell suspensions from the primary cultures were placed on a P100 panning dish coated with C5 anti-neuroepithelial antibody (1:20, gift from Ben Barres, Stanford University, Stanford, CA) and incubated at room temperature for 30 minutes. After the nonadherent cells were removed, the attached cells were grown for 1 to 2 weeks in DMEM/Ham’s F-12 with 10% FBS in the incubator. Cultured ONH astrocytes were characterized by positive immunostaining for both glial fibrillary acidic protein (GFAP) and neural cell adhesion molecule (NCAM), as described. 30 For these experiments, third-passage cells were grown and maintained in DMEM/Ham’s F-12 (Gibco BRL), 10% FBS, and 50-μg/mL gentamicin. 
RNA Extraction and Northern Blot Hybridization
Cell cultures were grown to confluence in 10-cm plates and treated with DEX (Sigma, St. Louis, MO) for 10 days at a concentration of 10−7 M. DEX was prepared in absolute ethanol at 0.1 mM and diluted 1000-fold into fresh complete IMEM every other day for the duration of the experiment. At the end of 10 days, both control and treated sample RNAs were extracted with a kit (RNeasy Mini Kit; Qiagen, Valencia, CA). RNA molecules selectively bound to the silica gel base were eluted with 50 μL RNase-free water. RNA from each sample was lyophilized to dryness, denatured in 50% formamide, and separated by 2.2 M formaldehyde, 1.25% agarose, 0.05 M 3-[N-morpholino] propane sulfonic acid (MOPS), and 1 mM EDTA gel electrophoresis. After electrophoresis, gels were washed with dH20 for 30 minutes and transferred to a nitrocellulose membrane (Schleicher & Schuell, Keene, NH) overnight by capillary action with 10× sodium saline citrate (SSC). After the transfer, UV cross-linked blots were prehybridized at 42°C for 6 to 12 hours in a buffer containing 50% formamide, 5× SSC, 5× Denhardt’s, 50 mM NaP04 (pH 7.4), 0.1% sodium dodecyl sulfate (SDS) with 50 μg/mL sheared salmon sperm DNA (Research Genetics, Huntsville, AL). The filters were then placed in fresh buffer, hybridized overnight to 1 to 5 × 106 cpm/mL of random-primer–labeled TIGR/MYOC cDNA (Roche Molecular Biosystems, Indianapolis, IN) at the same temperature. The TIGR/MYOC cDNA probe used in the hybridization experiments contained 1603 base pairs and was obtained by PCR amplification of pMC2 15 with primers 5′-AGCTTTGTTTAAACGCCTCACCAAGCCTCTGCAA-3′ (59–78 nucleotides [nt] TIGR/MYOC cDNA) and 5′-GGCGGATCCTGCCATTGCCTGTACAGCTTGGAG-3′ (1597–1621 nt TIGR/MYOC cDNA) followed by gel purification of the fragment with a PCR purification kit (QIAquick; Qiagen). After hybridization, filters were washed five times (15–20 minutes each): four times in 2× SSC-1% SDS (two at room temperature [RT] and two at 52°C) and once in 2× SSC at 52°C. Exposure was conducted using x-ray film (BioMax MR; Scientific Imaging Systems, Eastman Kodak, New Haven, CT) at −80°C with intensifying screens. To monitor RNA degradation and loading, filters were subsequently rehybridized to 2 × 106 cpm/mL of 28S oligonucleotide at 42°C for 2 hours, washed twice in 2× SSC-1% SDS (one at RT, one at 37°C) and exposed at RT. 33 TIGR/MYOC nucleotide number is according to Adam et al. 34  
Protein Extraction and Western Blot Analysis
Cell cultures were grown to confluence in 35-mm wells and treated with fresh 10−7 M DEX every other day. Control and DEX-treated cultures were collected at time points ranging from 1 to 10 days. For each cell culture, medium and cells from each 35-mm well were collected separately. The medium was centrifuged at 41,000g for 12 minutes to remove cellular debris. Cleared medium containing secreted cell products was then concentrated 20× on a filter (Centricon C-30; Amicon-Millipore, Bedford, MA) followed by buffer exchange to 0.01 M Tris-HCl [pH 7.4] and storage at −20°C. After the medium was removed, the cell monolayer from each well was washed twice with phosphate-buffered saline (PBS; Gibco BRL) followed by the addition of 2× loading buffer (100 mM Tris-HCl, 200 mM dithiothreitol [DTT], 4% SDS, 20% glycerol, and 0.02% bromophenol blue) to a concentration of 3 × 107 cells/mL. Proteins were extracted by boiling samples for 10 minutes, followed by centrifugation at 16,000g and storage at −20°C. 
Protein extracts were separated by 10% SDS-PAGE and electrotransferred to a polyvinylidene difluoride (PVDF) membrane (Bio-Rad, Hercules, CA). After blocking with 5% nonfat dry milk in PBS-0.2% Tween 20 (Sigma) for 2 hours, the membranes were incubated overnight at 4°C with rabbit polyclonal TIGR/MYOC antibodies. 35 Immunoreactive bands were visualized by chemiluminescence (ECL Plus; Amersham Pharmacia Biotech, Piscataway, NJ) after treatment with anti-rabbit IgG secondary antibodies conjugated to horseradish peroxidase (1:8000; Pierce, Rockford, IL). For the loading control, blots were reprobed with a mouse monoclonal β-tubulin antibody (clone TUB 2.1, 1:1000; Sigma) and visualized with conjugated horseradish peroxidase under the same conditions. Gels were run with equivalent volumes of the total cell extract samples and their corresponding supernatants. 
Recombinant Adenoviruses and their Delivery to Tissue Culture Cells
Replication-deficient recombinant adenoviral expression vectors encoding full-length TIGR/MYOC (AdhTIG3) and a truncated mutant TIGR/MYOC (AdhTIG1, Thr345STOP) were constructed and purified as previously described. 15 For gene delivery, cell cultures were grown to confluence in 35-mm wells, washed twice with PBS, and exposed to 25 to 100 plaque forming units (pfu)/cell of either AdhTIG3 or AdhTIG1 for 1 hour in 1 mL serum-free medium. After exposure to the virus, 1 mL IMEM-5% FBS was added to the medium and incubation continued for 48 hours. Proteins from the medium and cell monolayer were then extracted and analyzed as described earlier. Control dishes were treated with the same volume of viral vehicle under identical conditions. 
Gene Microarray Analysis
Early passages of HTM cell lines HTM-40 and HTM-41 and two separate dishes of 00-1LLS ONH astrocytes were treated with 10−7 M DEX for 6 to 8 days. Total RNA from each of the four experiments was extracted as described earlier. The targets for the DNA microarray analysis were prepared according to the manufacturer’s instructions (Human Genome U95Av2 GeneChip microarrays, n = 6; Affymetrix, Santa Clara, CA) at the Duke University Microarray Facility. These microarrays contain oligonucleotide probes representing 12,627 genes, in which each gene is represented by a probe set consisting of 16 to 20 probes. Arrays were hybridized to the targets at 45°C for 16 hours and then washed and stained, by using the staining station according to the manufacturer’s instructions (GeneChip Fluidics; Affymetrix). DNA chips were scanned (GeneChip Scanner; Affymetrix), and signals obtained by scanning were processed by the accompanying software (Microarray Suite, ver.4.0; Affymetrix). The absolute analysis determines whether transcripts represented on the array are detected in the sample (transcripts present, marginal, or absent). It uses a variety of metrics, including background and noise calculations, based on the hybridization intensities measured by the scanner. The comparison analysis determines the relative change in abundance for each transcript between a baseline and an experimental sample (fold change). It also uses a variety of metrics, including normalization and scaling factors, based on the intensity differences between each baseline and experimental probe set. The program normalizes expression values against the internal control and eliminates genes absent in both chips from the comparisons. Listed fold change values of ±2 are significant. 
To search for genes located at chromosomal regions linked to glaucoma, each of the selected genes was chromosome mapped using the Locus Link database (http://www.ncbi.nlm.nih.gov/LocusLink/). Each of the obtained chromosomal regions was then manually matched for glaucoma by using known glaucoma-associated loci acquired in the OMIM Morbid Map database (http://www.ncbi.nlm.nih.gov/Omim/searchmorbid/, provided in the public domain by the National Center for Biotechnology Information, Bethesda, MD). 
Relative Quantitative Reverse Transcription–Polymerase Chain Reaction (RQ RT-PCR)
Reverse transcription (RT) reactions were performed with a kit (Retroscript; Ambion). One microgram total RNA was mixed with 2 μL of 50 μM random decamers and nuclease-free water in a total of 12 μL, heated at 75°C for 2 minutes, and cooled on ice. The reaction was continued in a 20-μL reaction mixture containing 10 μL of 10× RT buffer (500 mM Tris-HCl [pH8.3], 750 mM KCl, 30 mM MgCl2, and 50 mM DTT), 4 μL dNTPs (2.5 mM each), 1 μL RNase inhibitor (10 U), and 1 μL Moloney murine leukemia virus (M-MuLV) reverse transcriptase (100 U). The RT reactions were incubated at 42°C for 60 minutes and terminated at 92°C for 2 minutes. A 2.5-μL aliquot of RT reaction was used for amplification by PCR, which was performed in a 50-μL reaction mixture containing 5 μL 10× PCR buffer (400 mM Tricine-KOH [pH 9.2], 150 mM KOAc, 35 mM Mg(OAc)2, and 37 μg/mL bovine serum albumin), 4 μL dNTPs (2.5 mM each), 4 μL of the gene primer-specific mix (5 μM each forward and reverse primer) and 1 μL cDNA polymerase mix (Advantage; Clontech, Palo Alto, CA). The PCR mixture was split into four tubes, and amplification was performed as follows: 94°C, 4 minutes and cycles of 94°C for 30 seconds, 60°C for 30 seconds, and 72°C for 60 seconds. For each primer pair, the linear range of the PCR reaction was determined by varying the number of cycles in each of the four tubes. RT-PCR products were electrophoresed on 2% agarose gels containing 25 ng/mL ethidium bromide. To eliminate the possibility of amplifying genomic DNA, primer pairs were chosen to span intron–exon splice boundaries, RT reactions were conducted in parallel omitting the reverse transcriptase enzyme, or both. An additional control was performed by incubating an aliquot of the PCR mixture without the RT template. 
Quantification of the PCR products was accomplished by the use of 18S rRNA internal standards. 18S rRNA was amplified with a primer-competimer set (QuantumRNA; Ambion) yielding a band of 489 bp. Because 18S rRNA is far more abundant than the mRNA of any gene under study, the 18S amplification reaction is reduced by the addition of competimers. These primers are modified to block extension by DNA polymerase. When combined with the functional primers, they compete for annealing to the 18S rRNA and allow amplification of a reduced number of 18S rRNA molecules. To determine the correct ratio of primers to competimers to use with a given gene, a 2.5-μL aliquot of the RT reaction was used in a 40-μL reaction mixture containing 5 μL 10× PCR buffer (400 mM Tricine-KOH [pH 9.2], 150 mM KOAc, 35 mM Mg(OAc)2, and 37 μg/mL bovine serum albumin), 4 μL dNTPs (2.5 mM each), and 1 μL cDNA polymerase mix (Advantage; Clontech). The mixture was split into five 8-μL tubes each containing 1 μL of gene-specific primers (5 μM each) and 1 μL of different 18S primer-competimer ratios (usually 1:9, 2:8, and 3:7) plus controls. PCR amplification was performed at the determined linear cycle for the gene under study in the same conditions, to yield multiplex PCR products and to allow selection of the linear range of the 18S rRNA. 
For relative quantification, reactions for each gene were repeated in triplicate in conditions in which the multiplex PCR products were all in the linear range. Band intensities were captured (ChemiDoc System, including a Chemi-cooled charge-coupled device camera, PCI digitizing image acquisition board, EpiChemi II Darkroom with transilluminator, and LabWorks Software; all from UVP, Upland, CA), and transferred to a spreadsheet (Excel, Microsoft, Redmond, WA) for calculation of averages and standard errors. 
Results
TIGR/MYOC Induction by DEX Is HTM Specific
Total RNAs from control and DEX-treated cells corresponding to the different tissue types were analyzed by Northern blot hybridization with full-coding TIGR/MYOC cDNA and 28S control probes. Figure 1A shows the photograph of an autoradiogram from representative Northern blots containing control and DEX-treated samples. Hybridization to control, 10-day confluent HTM cells revealed the presence of a single TIGR/MYOC mRNA band of approximately 2.0 kb (Fig. 1A) . HTM treatment with 10−7 M DEX for the same length of time showed a marked increase in TIGR/MYOC signal in the two previously described mRNA bands. 36 However, hybridization of total RNA from embryonic SkMCs, HeLa cells, and HUVECs revealed no detectable levels of TIGR/MYOC mRNA, either with or without DEX treatment. Reprobing of the blots with 28S showed that the lack of detection of TIGR/MYOC mRNA was not due to a breakdown of total RNA. Because of the poor RNA recovery of the DEX-treated HeLa cells, the noninduction of TIGR/MYOC in this cell line was reconfirmed using RT-PCR (not shown). TIGR/MYOC RNA was not detected in primary ONH astrocytes by microarray analysis. The absolute values for this gene in both uninduced and DEX-treated ONH astrocyte GeneChips resulted in “absent” calls. 
Western blot analyses of protein extracts from control and DEX-treated cells were performed using TIGR/MYOC and β-tubulin antibodies. Results are shown in Figure 1B . With TIGR/MYOC antibody, blots from HTM protein extracts showed a 55-kDa doublet protein, corresponding to glycosylated and nonglycosylated forms of TIGR/MYOC. 15 Treatment with 10−7 M DEX for 10 days resulted in a significant increase of TIGR/MYOC in HTM cells. However, analysis of protein extracts from control and DEX induced HeLa cells, HUVECs, and SkMCs, as well as ONH astrocytes, revealed faint TIGR/MYOC cross-reacting bands. In ONH astrocytes, the TIGR/MYOC protein band appears to be less intense in the DEX-treated cells. The cross-reacting band in the SkMC extracts had a slightly faster electrophoretic mobility than TIGR/MYOC. Although it is possible that some of the faint bands observed in the nonHTM cell extracts are nonspecific, a positive control HTM-DEX sample that was run in all gels showed only TIGR/MYOC at the corresponding size. Reprobing of the blots with β-tubulin antibody controlled the loading of the samples. These observations demonstrate that the TIGR/MYOC gene is induced with DEX only in the HTM cells. 
Processing and Secretion of TIGR/MYOC in the Different Cell Types
Regardless of a tissue’s ability to produce TIGR/MYOC natively, we wanted to determine whether the cell lines in which TIGR/MYOC was not induced by DEX possessed the intrinsic processing and trafficking machinery necessary for the posttranslational modification and secretion of TIGR/MYOC. HeLa cells, HUVECs, and SkMCs were grown to 70% to 80% confluence and infected with replication-deficient adenoviral expression vectors for both wild-type TIGR/MYOC (AdhTIG3) 15 and a truncated mutant TIGR/MYOC (AdhTIG1) 17 previously shown to be retained intracellularly in HTM cells. 17 Western blot analysis of equivalent fractions of infected pellets and supernatants revealed that in all cases, the wild-type form of TIGR/MYOC was secreted, whereas the mutant was not (Fig. 2) . As seen previously in HTM cells, 15 17 18 the wild-type infected cultures showed a doublet of 55 kDa in all cell types, both intracellularly and secreted into the supernatant. Similarly, when infected with truncated mutant TIGR/MYOC expression vector, all cell lines retained the protein intracellularly. The observation that the mutant TIGR/MYOC is not secreted may indicate that this pathologic mutation interrupts or cripples a common processing pathway. 17 18 Together, these results indicate that although the DEX induction of TIGR/MYOC is not present in other cell types, the glycosylation and secretion mechanisms for this protein are ubiquitous among the different body tissues studied. 
Microarray Comparison of Genes Present in DEX-Induced ONH Astrocytes and HTM
From our RNA and protein studies we found that the DEX response of TIGR/MYOC appears to be TM tissue-specific. To identify other TM factors that could play a role in the TIGR/MYOC secondary glucocorticoid response and/or be involved in the DEX-triggered increase of resistance, we used gene microarrays. We compared expression profiles of DEX-induced ONH astrocytes (baseline) versus DEX-induced HTM (experimental) using microarrays (U95Av2 GeneChips, n = 6; Affymetrix). We searched for genes overexpressed in the HTM-DEX in comparison with ONH astrocytes-DEX. These HTM-overexpressed genes included genes from three groups: those upregulated by DEX in HTM and not upregulated (or less upregulated) in ONH astrocytes; those that were present in HTM and not in ONH astrocytes (tissue specific genes), and genes that were downregulated by DEX in ONH astrocytes and not downregulated (or less downregulated) in HTM cells. 
Four different experiments—two using one human ONH astrocytes cell-cultured line (00-1LLS; denoted A1DEX and A2DEX) and two using two HTM cell-cultured lines (HTM40 and HTM41; denoted H1DEX and H2DEX)—were treated with 10−7 M DEX for 6 to 8 days and their RNA extracted. Each of the four treated RNAs and two untreated controls (A1 and H1) were then hybridized to individual microarrays (n = 6) and analyzed as described in the Materials and Methods section. Individual outputs from each of the gene chips were compared in sets of two. To identify genes that were specifically overexpressed in the HTM-DEX sample, ONH-DEX astrocytes were used as a baseline. A total of four paired comparisons were performed: A1DEX versus H1DEX, A1DEX versus H2DEX, A2DEX versus H1DEX, and A2DEX versus H2DEX. Additional comparisons using the untreated control (H1 versus H1DEX and A1 versus A1DEX) were also performed. 
The number of genes with intensities in the chips that resulted in absolute values classified as present (P) by the manufacturer’s algorithms (Affymetrix) were calculated for the A1DEX and H1DEX chips. From the 12,627 genes measured on each microarray, we found that 6099 genes were expressed in the A1DEX cell line, whereas 4751 genes were expressed in the H1DEX. Within the set of genes expressed in H1DEX-treated cells, 458 genes were not expressed at any detectable levels in A1DEX-treated cells. We called these genes “turned on” in H1DEX. Similarly, 1806 genes that showed expression in the A1DEX astrocytes were not detected in H1DEX and were designated “turned off” in H1DEX. A total of 4292 genes showed expression in both induced cell types. 
Genes with Expression Levels Changing More Than 20-fold in Comparing ONH Astrocytes-DEX with HTM-DEX
To identify those genes with expression levels that were the most changed (either most increased or most decreased) in HTM-DEX compared with ONH astrocytes-DEX cells, we used fold change analysis. Fold change values were calculated as the ratio of the mean expression levels of each gene between HTM and ONH astrocyte DEX-treated cell cultures. We identified 503 genes that were overexpressed and 1227 genes that were underexpressed in the HTM-DEX cells. Those genes with expression levels that changed by more than 20-fold in the HTM-DEX cells compared with ONH astrocyte-DEX are shown in Table 1 . All but one of the 40 genes showing a more than 20-fold increase were genes of known function. Four of the 18 genes with expression decreased more than 20-fold were unknown expressed sequence tags (ESTs). Very few if any of these genes were housekeeping genes and at least one fourth of them seemed to be involved in extracellular functions. 
To reduce the number of genes for further analysis, we focused on those with the most increased expression. To do so, we searched for those genes showing the highest fold change values not only in one, but in all four ONH-DEX (baseline) versus HTM-DEX (experimental) comparisons (as described earlier). Because of the wide variability of fold change values among the four comparisons, we established a stringent criterion. We selected only those genes with fold change values of five or higher in at least three of the four comparisons (fold change values of 2 are already significant). By implementing this filter, we identified 15 genes. These 15 genes along with the fold change values in each of the four DEX comparisons were designated The Most Overexpressed HTM-DEX–Specific Genes and are listed in Table 2
Classification of the Most Overexpressed HTM-DEX–Specific Genes
The genes identified on Table 2 were categorized into three groups defined as follows: genes induced by DEX in HTM and not induced (or less induced) in ONH astrocytes (group a), genes present in HTM and not present in ONH astrocytes (group b, tissue specific genes) and genes downregulated by DEX in ONH astrocytes and not downregulated (or less downregulated) in HTM (group c). 
To categorize the genes from Table 2 into these three groups, we made use of the comparison analysis obtained with the microarrays from HTM uninduced versus HTM-DEX, ONH astrocytes uninduced versus ONH-DEX, and HTM uninduced versus ONH astrocytes uninduced (H1 versus H1DEX; A1 versus A1DEX; A1 versus HTM). We found that most genes (12 out of 15) belonged to group a. Of these, nine were induced by DEX in HTM but not in ONH astrocytes, and three (IGFBP2, AKR1C3, and AP1G1) were induced by DEX in both cell lines (higher in HTM). Two genes belonged to group b. Of these, one (AKR1C1), was overexpressed in uninduced HTM versus uninduced ONH astrocytes (tissue specific) and further overexpressed by DEX in the HTM cells but not in ONH astrocytes; the second gene (ASS) was only HTM tissue–specific and its expression was not induced by DEX. Only one gene belonged to group c. This gene, PCOLCE, was severely downregulated in ONH astrocytes treated with DEX but showed no change with DEX in HTM cells. 
In addition to TIGR/MYOC, 8 of the 15 genes differentially overexpressed in HTM-DEX encode secreted glycoproteins. These include: α1-antichymotrypsin (ACT) a serine protease inhibitor (serpin) involved in neurodegenerative diseases 37 ; pigment epithelium-derived factor (PEDF), an RPE-secreted glycoprotein with a neuroprotective role 38 ; type I procollagen C-proteinase enhancer (PCOLCE), a procollagen binding protein that affects synthesis of collagen type I 39 ; human cartilage protein GP-39, a secretory product of chondrocytes, highly abundant in cartilage and a marker for joint disease 40 ; cornea-derived transcript 6 (CDT6), involved in increased ECM deposition and antiangiogenesis 41 ; prostaglandin D2 synthase (PGD2S), responsible for the conversion of prostaglandin H2 into prostaglandin D2 (PGD2), a hormone that inhibits release of nitric oxide (NO) and induces vasodilatation 42 ; aldoketoreductase 3 (AKR1C3), an enzyme that catalyzes the conversion of PGD2 to prostaglandin F2 (PGF2); 43 and finally, apolipoprotein D (APOD), a lipocalin shown to accumulate in the cerebrospinal fluid of patients with neurologic diseases, such as Alzheimer, multiple sclerosis, and schizophrenia. 44 45  
The nonsecreted proteins identified by our criteria include metabolic enzymes, (AKR1C1 and ASS), an actin (ACTG2), and proteins involved in mitochondrial transport (CTP) and trafficking (AP1G1). 
RQ RT-PCR Confirmation of Differentially Expressed Genes
Five selected genes identified as being differentially expressed according to the parameters of the microarray system (Affymetrix) were further analyzed by RQ RT-PCR, using H1DEX and A1DEX total RNA. The primers used for the amplifications are shown in Table 3 , and the amplified products in Figure 3 . For each of the genes measured by RQ RT-PCR, the mRNA levels were shown to change in the same direction as the changes measured by the microarray detection. However, the fold change values showed a higher dynamic range, a phenomenon also known to occur between relative quantitative PCR and microarrays. 46 The expression of TIGR/MYOC and PGD2S was not detected by PCR in ONH astrocytes-DEX, agreeing with the absence call obtained in the arrays. In the TIGR/MYOC panel, the TIGR/MYOC reaction loaded in the gel was diluted 100×. Assuming a minimal number for densitometry detection of the A1DEX band, the change calculation would result in a TIGR/MYOC H1DEX overexpression of approximately 315-fold. Overexpression of α1-antichymotrypsin was calculated to be 54 ± 1.1 higher. In the PEDF panel, the PEDF reaction loaded in the gel was diluted 100×, resulting in a calculated change multiple of overexpression in the HTM of 111 ± 3.5. Last, in the PCOLCE panel, the overexpression of this gene by this method was found to be 1.5 ± 0.1. 
HTM-DEX–Specific Genes Located in Regions Linked to Glaucoma
Because glaucoma develops in some patients treated with corticosteroids, we were interested to know how many of the specific HTM-DEX–upregulated genes would map to chromosomal regions linked to glaucoma. Comparison of A1DEX versus H1DEX revealed that 249 genes were differentially overexpressed in HTM-DEX with fold change values higher than 4. Locus Link database mapping of each of those genes, together with examination of known glaucoma-linked regions listed in OMIM (as of January 24, 2002), produced an overlap of 15 genes (Table 4) . Of these 15 genes, only the TIGR/MYOC gene has thus far been linked to glaucoma. 2 Two additional genes, angiopoietin like-factor CTD6 and type I membrane associated glycoprotein T1A-2 are significantly induced by DEX only in the HTM. Optineurin, (labeled as FIP2 in the microarray chip; Affymetrix) a gene recently linked to glaucoma, was not overexpressed in H1DEX versus A1DEX. 
Discussion
Although TIGR/MYOC is expressed in many different tissues in the body, mutations in TIGR/MYOC have thus far been associated only with glaucoma. How mutations in this widely expressed protein cause disease only in the eye is not yet understood. 
Our results showing that altered processing and secretion of the mutant occurring across cell types are an indication that these characteristics, per se, are not responsible for the disease. Current hypotheses on the pathologic nature of TIGR/MYOC focus on the abnormal intracellular accumulation of the mutant protein in the TM cells 15 and thus in a gain of function. 15 19 An intracellular accumulation of TIGR/MYOC mutant protein resulting in a pathologic condition in the TM and not in other tissues could be caused by a tissue-specific overexpression of a gene in response to certain insults. Similarly, the disease could be triggered by the specific induction of a particular cellular trait that would make these cells less tolerable to defective protein accumulation. Studying the differential induction of TIGR/MYOC in the TM could then provide an insight as to what causes glaucoma to develop in patients with mutated TIGR/MYOC. 
In this study, we report that the distinguishing feature of TIGR/MYOC in the TM is its marked induction with glucocorticoid treatment. TIGR/MYOC RNA and protein levels increase in HTM after 7 to 10 days of DEX treatment but do not increase in any of the other tested cells. TIGR/MYOC was also reported not to be induced by DEX on porcine astrocytes. 25 We believe this TIGR/MYOC tissue-specific property may be able to elucidate the linkage of this protein to glaucoma only. 
Having identified the specific TIGR/MYOC induction, we set out to find other TM factors that were present in DEX-induced HTM cells and barely present or absent in other tested DEX-induced cells, including ONH astrocytes, a key cell type involved in ONH remodeling in glaucoma. 26 We reasoned that their specific presence in HTM-DEX could be involved in overexpression of TIGR/MYOC or be independently relevant in steroid-induced elevated IOP. Using microarray technology (GeneChip; Affymetrix), we compared expression profiles of HTM cells and ONH astrocytes induced with DEX. 
In addition to confirming our TIGR/MYOC findings, the microarray studies further identified approximately 600 genes with expression levels in HTM-DEX in comparison with ONH astrocytes-DEX had fold change values higher than plus four or lower than minus four. To narrow our selection, we implemented filters on the fold change expression. Our focus was, rather than to define an actual fold change value, to select those genes that exhibited the highest overexpression. Because of the variability of fold change values observed among the compared cell lines, we applied a stringent criterion and selected only genes with values higher that fivefold (values of twofold are significant) in three of four comparisons. We think this observed variability is mostly due to the distinct response of a given gene to insults in different individuals. 36 Our criteria identified 15 genes, most of which encode secreted glycoproteins. Although we do not know the particular role of these proteins in the TM, their reported known functions appear to have potential relevance to the regulation and maintenance of IOP. 
Because of the important role of the abundance and composition of ECM in the physiology of TM, 47 48 an induction of functional serine protease inhibitors such as α1-antichymotrypsin would result in increased ECM deposition and decreased degradation in the TM. Furthermore, α1-antichymotrypsin is associated with formation of amyloid plaques 37 in aged and Alzheimer-affected brains. 49 50 It seems possible that the presence of plaques or extracellular aggregates observed in the HTM of steroid-induced glaucoma 51 is influenced by the HTM DEX-induced expression of this powerful serpin. 
Because of their clear role in other tissues, two of the other glycoproteins identified, CDT6 and human cartilage GP39, may also influence the composition of the ECM in the DEX-induced TM. It is well established that the TM from human anterior segments perfused with DEX, as well as that of patients diagnosed with corticosteroid-induced glaucoma, has a distinct morphology, with thickened trabecular beams, decreased intratrabecular spaces and an increased amount of extracellular materials. 51 52 It is further intriguing that CDT6, a relatively new corneal protein, maps to chromosomal region 1p36, where the GLC3B region has been associated with a recessive form of congenital glaucoma. 53 We had previously reported GP-39 as one of the most abundant clones in an HTM library from a normal individual. 54 Its specific induction with DEX reinforces the relevance of this protein in the TM. 
The specific induction of PEDF in the TM is intriguing. Its reported protection of neurons from apoptosis, glutamate toxicity, and especially hydrogen peroxide 38 55 56 leads us to speculate that perhaps this protein serves a similar protective role in the TM. 
One of the two reported functions of PGD2 synthase is the formation of PGD2. A synthetic analogue of PGD2 that has been shown to reduce IOP in humans. 57 PGD2 synthase has been detected in several ocular tissues and in aqueous humor. 58 Recently, the same enzyme has been reported to be overexpressed in ONH astrocytes cultured from glaucomatous eyes. 59 It is interesting that the expression of this gene is altered in two glaucoma-implicated cell types and under two distinct glaucomatous conditions. 
In our study, two other genes encoding enzymes that regulate steroid hormone metabolism, AKR1C1 and AKR1C3, were also markedly overexpressed in HTM-DEX cultures. The role of these enzymes, also known as 3α-hydrosteroid dehydrogenases (3α-HSD), 60 in the HTM have been studied in detail in relation to steroid-induced elevated IOP. 61 Abnormal activity of 3α-HSD in the HTM results in abnormal accumulation of 5α- and 5β-dihydrocortisol, which causes elevated IOP in rabbits, whereas their normal product, tetrahydrocortisol, was inactive. 62 63 64 More recently, it has been shown that primary cultures of glaucomatous ONH astrocytes exhibit extraordinary upregulation of AKR1C1 and AKR1C3 mRNA, suggesting active steroid metabolism by glaucomatous astrocytes. 59 Our current results in HTM-DEX, together with these findings, suggest for the first time a potential association between abnormal steroid metabolism in the HTM, elevated IOP, and glaucomatous optic neuropathy. 
Although this study focused primarily on genes overexpressed in the TM, we have also listed those genes with expression that is most reduced in HTM-DEX, compared with ONH astrocytes-DEX. Of particular interest is the lower expression of elastin in the HTM, further confirmed by the presence of two probe sets in the microarray. This difference appears to be due to a higher abundance of the elastin gene in ONH astrocytes rather than a down- or upregulation of the gene by DEX. Using immunohistochemistry, earlier studies reported no detection of elastin in the HTM cells, with only a moderate induction of DEX. 65 In contrast, ONH astrocytes express detectable elastin mRNA in vitro and in vivo. 66 Although it has been reported that DEX suppresses elastin gene expression in skin fibroblasts in culture, 67 it has also been shown that the elastin promoter contains glucocorticoid response elements that upregulate the expression of reporter genes in transgenic mice. 68 69 Other genes that are downregulated in DEX-HTM are associated with the central nervous system; therefore, they are more abundant in astrocytes. Among these genes are STAT1, P311, Pentraxin 1, and Serpin B2. 
Most of the proteins identified in this study are secreted. As such, they have the potential to affect significantly the ECM of the TM. It is interesting to note that the known functions of these proteins point to mechanisms that could have different, possibly opposite, effects on outflow facility. The overexpression of proteins, such as α1-antichymotrypsin, CDT6, and cartilage GP39, seems to suggest a direct increase in ECM deposition and decreased degradation—thus, a risk for potential obstruction of outflow channels. Stimulation of other types of proteins such a PEDF, with an extensive history of its protective role; TIGR/MYOC, a protein potentially involved in protecting TM against elevated IOP 36 ; and perhaps PGD2S, an enzyme involved in the synthesis of an IOP-lowering prostaglandin, points to the induction of different types of defense mechanisms against increased resistance. 
TIGR/MYOC was first identified as a gene upregulated in HTM cells with long-term steroid treatment in culture. The initial excitement surrounding this discovery was the prospect that this gene could be responsible for steroid-induced glaucoma. Establishing the connection between TIGR/MYOC function and pressure regulation has been difficult and remains the object of intense research and controversy. Our data show that in the cell types studied, TIGR/MYOC was significantly induced by DEX in HTM only. By comparing expression profiles, we have identified additional genes that share this tissue-specific induction. This set of genes, though far from complete, confirms at the molecular level the primacy of the role of the ECM in the development of steroid-induced glaucoma. Most important, these genes reveal the presence of new, potentially protective mechanisms that may also become activated in the cells of the HTM after treatment with DEX. Elucidation of which of those mechanisms TIGR/MYOC serves in the meshwork and of the relevance of the genes identified in this analysis remains an important challenge. 
 
Figure 1.
 
Induction of TIGR/MYOC by dexamethasone (DEX) in different cell types after 10 days of no treatment (lane C) or 10−7 M DEX treatment (lane D). (A) Northern blot analysis of total RNA from the indicated cells. Each blot was hybridized sequentially to TIGR/MYOC and 28S probes to control for RNA integrity and equal loading. (B) Western blot analysis of proteins from the indicated cells. Protein extracts were separated on 10% SDS-polyacrylamide gels and transferred to PVDF membrane. Specific proteins were detected with a TIGR/MYOC polyclonal antibody 29 and visualized by chemiluminescence. Blots were reprobed with a β-tubulin antibody for control loading. TIGR/MYOC was induced with DEX only in HTM cells. Induction occurred at transcription and translation levels. ONH-Astr, human ONH astrocytes.
Figure 1.
 
Induction of TIGR/MYOC by dexamethasone (DEX) in different cell types after 10 days of no treatment (lane C) or 10−7 M DEX treatment (lane D). (A) Northern blot analysis of total RNA from the indicated cells. Each blot was hybridized sequentially to TIGR/MYOC and 28S probes to control for RNA integrity and equal loading. (B) Western blot analysis of proteins from the indicated cells. Protein extracts were separated on 10% SDS-polyacrylamide gels and transferred to PVDF membrane. Specific proteins were detected with a TIGR/MYOC polyclonal antibody 29 and visualized by chemiluminescence. Blots were reprobed with a β-tubulin antibody for control loading. TIGR/MYOC was induced with DEX only in HTM cells. Induction occurred at transcription and translation levels. ONH-Astr, human ONH astrocytes.
Figure 2.
 
Processing and secretion of TIGR/MYOC in the different cell types. Cell lines were infected with either wild-type (AdhTIG3) or mutant (AdhTIG1, Thr345STOP) TIGR/MYOC adenovirus expression vectors at 25 to 200 pfu/cell. Proteins were extracted from supernatants (Sup) and cellular fractions (Cell) at 48 hours after infection. Equivalent volumes of cell extracts and their corresponding supernatants were run on 10% SDS-PAGE and transferred to PVDF membrane. Specific proteins were detected with a TIGR/MYOC polyclonal antibody 29 and visualized by chemiluminescence. Wild-type TIGR/MYOC (molecular mass doublet at 55–57 kDa) was secreted in every cell type. Mutant truncated TIGR/MYOC (mass ∼43 kDa) was found to be retained intracellularly in every cell type.
Figure 2.
 
Processing and secretion of TIGR/MYOC in the different cell types. Cell lines were infected with either wild-type (AdhTIG3) or mutant (AdhTIG1, Thr345STOP) TIGR/MYOC adenovirus expression vectors at 25 to 200 pfu/cell. Proteins were extracted from supernatants (Sup) and cellular fractions (Cell) at 48 hours after infection. Equivalent volumes of cell extracts and their corresponding supernatants were run on 10% SDS-PAGE and transferred to PVDF membrane. Specific proteins were detected with a TIGR/MYOC polyclonal antibody 29 and visualized by chemiluminescence. Wild-type TIGR/MYOC (molecular mass doublet at 55–57 kDa) was secreted in every cell type. Mutant truncated TIGR/MYOC (mass ∼43 kDa) was found to be retained intracellularly in every cell type.
Table 1.
 
Genes with Expression Levels That Changed More Than 20-fold, when A1DEX Astrocytes were compared with H1DEX Trabecular Meshwork Cells
Table 1.
 
Genes with Expression Levels That Changed More Than 20-fold, when A1DEX Astrocytes were compared with H1DEX Trabecular Meshwork Cells
GenBank Accession No. UniGene Gene Name Fold Change* Gene Function
Overexpressed in H1DEX
 AF089747 Hs.234726 α1-antichymotrypsin (SERPINA3) 478 Serine protease inhibitor
 U05861 Hs.201967 Hepatic dihydrodiol dehydrogenase (AKR1C1) 354 NADH/NADPH aldoketoreductase
 U37100 Hs.116724 Aldose reductase-like peptide (AKR1B10) 175 NADPH reductase
 Z97171 Hs.78454 Trabecular meshwork inducible gluco. resp. (MYOC) 148 Secreted glycoprotein
 D17793 Hs.78183 Aldo-ketoreductase (AKR1C3) 132 Prostaglandin D2 metabolism
 M28130 Hs.624 Interleukin 8 (IL8) 100 Chemokine, cell motility, signaling
 U29953 Hs.173594 Pigment epithelium-derived factor (PEDF) 87 Serine protease inhibitor, serpin
 AF037335 Hs.5338 Carbonic anhydrase XII (CA12) 85 Aqueous humor formation/calcification
 L33799 Hs.202097 Procollagen C-proteinase enhancer protein (PCOLCE) 79 Collagen processing
 AL050025 Hs.5344 γ1-adaptin (AP1G1) 55 Formation of clathrin pits and vesicles
 J02611 Hs.75736 Apolipoprotein D (APOD) 50 Lipocalin, secr glycpr binds hydroph mol
 M98539 Hs.8272 Prostaglandin D2 synthase (PTGDS) 49 Prostaglandin biosynthesis
 D31628 Hs.2899 4-Hydroxyphenylpyruvic acid dioxygenase (HPD) 49 Tyrosine catabolism
 L25879 Hs.89649 Epoxide hydrolase 1 (EPHX1) 43 Detoxification exogenous chemicals
 AF017786 Hs.239756 Phosphatidic acid phosphatase (PPAP2B) 41 Phosphatidylinositol (PI) turnover
 D00654 Hs.78045 γ2-actin, enteric smooth muscle (ACTG2) 40 Cytoskeletal network
 AF015926 Hs.184276 Solute carrier family 9 (SLC9A3R1) 39 Sodium/hydrog exchange/actin organization
 A1445461 Hs.351316 Transmembrane 4 superfamily member 1 (TM4SF1) 39 Tetraspanin, regulates basic cell processes
 M90657 Hs.351316 Tumor-associated antigen L6 (TAAL6) 39 Tetraspanin, regulates basic cell processes
 AL049653 Hs.146559 Angiopoietin-like factor (CDT6) 37 Anti-angiogenesis/ECM deposition
 M17017 Hs.624 Interleukin 8 (IL8) 32 Chemokine, cell motility
 U09937 Hs.179657 Plasminogen activator urokinase receptor (PLAUR) 31 Signal transduction/ECM remodeling
 Y18483 Hs.22891 Solute carrier family 7 (SLC7A8) 31 Amino acid transporter
 M25915 Hs.75106 Clusterin (CLU) 29 Plasma glycoprotein
 M76665 Hs.37012 Hydroxysteroid dehydrogenase (HSD11B1) 27 Conversion of cortisol to cortisone
 L26232 Hs.154854 Phospholipid transfer protein (PLTP) 26 Plasma lipid transfer
 X01630 Hs.160786 Argininosuccinate synthetase (ASS) 26 Nitric oxide synthesis/urea cycle
 L38486 Hs.118223 Microfibril-associated glycoprotein 4 (MFAP4) 26 Integrin ligand, extracellular matrix
 AI936826 Hs.85339 G protein-coupled receptor 39 (GPR39) 25 Signal transduction
 AF096870 Hs.194540 Estrogen-responsive B box protein (EBBP) 24 Zinc finger protein
 L13698 Hs.65029 Growth arrest-specific 1 (GAS1) 24 Cell growth inhibition
 J04164 Hs.146360 Interferon induced transmembrane protein 1 (IFITM1) 22 Cell growth inhibition
 U96750 Hs.194695 Ras homologue gene family member 1 (ARH1) 22 Cell growth inhibition
 AB003184 Hs.102171 Immunoglobulin superfamily leucine-rich (ISLR) 22 Protein binding, cell adhesion
 X14885 Hs.2025 Transforming growth factor β3 (TGFB3) 21 Cell proliferation and differenciation
 X04470 Hs.251754 Secretory leukocyte protease inhibitor (SLPI) 21 Protease (chymotrypsin) inhibitor
 AF060567 Hs.126782 Sushi repeat protein (SRPUL) 21 Deleted in RP, unknown function
 L13698 Hs.65029 Growth arrest-specific 1 (GAS1) 21 Cell growth inhibition
 AF027734 Hs.6090 Deleted in bladder cancer chromosomal region candidate 1 (DBCCR1) 21 Tumor suppressor
 AI381790 Hs.74120 Adipose-specific 2 (APM2) 21 Unknown/specific to adipose tissue
Underexpressed in H1DEX
 AB018301 Hs.22039 KIAA0758 protein (KIAA0758) −112 Unknown
 X52896 Hs.9295 Elastin (ELN) −110 Elastic fiber component/connective tissue
 U61849 Hs.84154 Neuronal pentraxin 1 (NPTX1) −68 Snake venom-binding protein
 Z24680 Hs.151641 Glycoprotein A repetitions predominant (GARP) −63 Unknown
 M18533 Hs.169470 Dystrophin (DMD) −42 Anchoring cytoskeleton to membrane
 U30521 Hs.142827 P311 protein (P311) −41 Cell transformation and motility
 AF045800 Hs.40098 Gremlin/bone morphogenetic protein antagonist1 (CKTSF1B1) −39 Secreted/blocks BMP signaling
 AB023194 Hs.300855 KIAA0977 protein (KIAA0977) −37 Unknown
 D83402 Hs.302085 Prostacyclin synthase (PTGIS) −35 Potent vasodilator/P450 superfamily
 M26326 Hs.65114 Keratin 18 (KRT18) −35 Intermediate filament component
 W27472 Hs.24391 Hypothetical protein FLJ13612 (FLJ13612) −33 Unknown
 Y00630 Hs.75716 Plasminogen activator-inhibitor (SERPINB2) −30 Inhibitor extracellular proteolysis
 M97936 Hs.21486 Transcription factor (STAT1) (ISGF-3) −29 Activator of transcription
 X15998 Hs.81800 Chondroitin sulfate proteoglycan 2 (CSPG2) −29 ECM glycoprotein/binds to hyaluronan
 AL031058 Hs.74316 Desmoplakin I (DPI) −26 Cytoskeleton/intercellular adhesion
 L06139 Hs.89640 Receptor tyrosine kinase (TEK) −25 Cells of endothelial lineage
 X15998 Hs.81800 Chondroitin sulphate proteoglycan 2 (CSPG2) −25 ECM glycoprotein/binds to hyaluronan
 M36860 Hs.9295 Elastin (ELN) −23 Elastic fiber component/connective tissue
Table 2.
 
The Most Overexpressed HTM-DEX–Specific Genes
Table 2.
 
The Most Overexpressed HTM-DEX–Specific Genes
GenBank Accession No. UniGene Fold Change Gene Name Gene Group* Gene Function
A1DEX vs. H1DEX A1DEX vs. H2DEX A2DEX vs. H1DEX A2DEX vs. H2DEX Average
AF089747 Hs.234726 478 125 42 13 165 α1-antichymotrypsin (SERPINA3) a Serine protease inhibitor
U05861 Hs.201967 354 69 66 13 126 Hepatic dihydrodiol dehydrogenase (AKR1C1) b NADH/NADPH aldoketoreductase
BC004312 Hs.162 6 5 113 102 57 Insulin-like growth factor binding protein-2 (IGFBP2) a Modulates IGF function
Z97171 Hs.78454 148 18 30 5 50 Trabecular Meshwork inducible gluco. resp. (MYOC) a Secreted glycoprotein
U29953 Hs.173594 87 59 20 18 46 Pigment epithelium-derived factor (PEDF) a Neuron survival factor
L33799 Hs.202097 79 60 18 17 44 Procollagen C-proteinase enhancer (PCOLCE) c Collagen processing
D17793 Hs.78183 132 27 12 3 43 Aldo-keto reductase (AKR1C3) a Prostaglandin D2 metabolism
M98539 Hs.8272 49 65 13 25 38 Prostaglandin D2 synthase (PTGDS) a Prostaglandin biosynthesis
Y08374 Hs.75184 12 91 3 24 33 Cartilage GP-39 (CHI3L1) a Extracellular matrix remodeling
D00654 Hs.78045 40 57 9 15 30 γ2-actin, enteric smooth muscle (ACTG2) a Cytoskeletal network
AL049653 Hs.146559 37 49 10 17 28 Angiopoietin-like factor (CDT6) a Antiangiogenesis/ECM deposition
X01630 Hs.160786 26 58 8 18 28 Argininosuccinate synthetase (ASS) b Nitric oxide synthesis/urea cycle enzyme
X96924 Hs.111024 31 42 8 12 23 Citrate transport protein (CTP) a Citrate mitochondrial transmembrane transport
J02611 Hs.75736 50 20 12 5 22 Apolipoprotein D (APOD) a Lipocalin/secreted glycoprotein binds hydrophobic molecules
AL050025 Hs.5344 55 7 21 3 21 γ1-adaptin (AP1G1) a Formation of clathrin pits and vesicles, protein sorting, and traffic
Table 3.
 
Primers and Amplification Products Used in RQ RT-PCR Analysis
Table 3.
 
Primers and Amplification Products Used in RQ RT-PCR Analysis
Gene Name Accession No. Sense Primer Antisense Primer Size (bp)
TIGR/MYOC NM_000261 5′-CTG GAG GAA GAG AAG AAG CGA CTA A-3′ 5′-CTG TGT CAT AAG CAA AGT TGA CGG TA-3′ 912
α1-antichymotrypsin AF089747 5′-AAG CTC ATC AAC GAC TAC GTG A-3′ 5′-ATT CCA AGT TCC TTA CTG AGA GCC-3′ 969
PEDF M76979 5′-GGC TGT CTC CAA CTT CGG CTA-3′ 5′-TGG TGA CTT CGC CTT CGT AAC T-3′ 811
Prostaglandin D2 synthase BC005939 5′-CCA ACT TCC AGC AGG ACA AGT TCC-3′ 5′-CTT CCG GAG TTT ATT GTG CAB GAT-3′ 633
PCOLCE L33799 5′-CTA ATA AGG AGT GCA TCT GGA CCA TAA-3′ 5′-CTG AAG GAG ATT CCT CTG TTT TCT C-3′ 691
Figure 3.
 
Confirmation of the differential expression of selected genes in DEX-treated HTM cells and ONH astrocytes by RQ RT-PCR. Amplification of the five selected genes was performed with their specific primers (Table 3) and their predetermined exponential number of cycles (shown underneath each panel). Multiplex PCR reactions for each gene were performed together with an 1:15 primer/competimer mix of 18S RNA in conditions under which the multiplex products were in the linear range. Band intensities were then captured and analyzed. Amplification of each gene was repeated three times. H1DEX cDNA in the first and third panels was diluted 100× because of the high abundance of the target gene. Lane M: ΦX174 RF DNA/HaeIII fragments. The level of expression of all selected genes changed in the same direction as the changes measured by the microarrays.
Figure 3.
 
Confirmation of the differential expression of selected genes in DEX-treated HTM cells and ONH astrocytes by RQ RT-PCR. Amplification of the five selected genes was performed with their specific primers (Table 3) and their predetermined exponential number of cycles (shown underneath each panel). Multiplex PCR reactions for each gene were performed together with an 1:15 primer/competimer mix of 18S RNA in conditions under which the multiplex products were in the linear range. Band intensities were then captured and analyzed. Amplification of each gene was repeated three times. H1DEX cDNA in the first and third panels was diluted 100× because of the high abundance of the target gene. Lane M: ΦX174 RF DNA/HaeIII fragments. The level of expression of all selected genes changed in the same direction as the changes measured by the microarrays.
Table 4.
 
H1DEX Overexpressed Specific Genes Located in Chromosomal Regions Linked to Glaucoma
Table 4.
 
H1DEX Overexpressed Specific Genes Located in Chromosomal Regions Linked to Glaucoma
GenBank Accession No. UniGene Gene Name Fold Change Glaucoma-Linked Chromosomal Region
Z97171 Hs.78454 Trab Mesh inducible gluco. resp. (MYOC)* 148 1q24.3-q25.2
AI445461 Hs.351316 Transmembr 4 superfamily1 (TM4SF1) 39 3q21-q25
M90657 Hs.351316 Tumor-associated antigen L6 (TAAL6) 39 3q21-q25
AL049653 Hs.146559 Angiopoietin-like factor (CTD6)* 37 1p36
U30255 Hs.75888 Phosphogluconate dehydrogenase (PGD) 19 1p36.2-p36.13
AI660929 Hs.135150 Type-I membrane-asso glycoprotein (T1A-2)* 12 1p36
AF030428 Hs.135150 Type-I membrane-asso glycoprotein (T1A-2)* 11 1p36
D83492 Hs.3796 Ephrin (kinase) receptor protein (EPHB6) 9 7q33-q35
X15414 Hs.75313 Aldose reductase (AR) 9 7q35
D28124 Hs.76307 Human mRNA for unknown product 6 1p36.3-p36.2
M94345 Hs.82422 Capping protein (actin filament) (CAPG) 5 2cen-q24
AF044253 Hs.298184 Potassium channel (KCNAB2) 5 1p36.3
U50648 Hs.274382 Protein kinase, interferon inducible (PRKR) 5 2p22-p21
M64231 Hs.76244 Spermidine synthase (SRM) 5 1p36-p22
M55914 Hs.254105 Enolase 1 (ENO1) 4 1p36.3-p36.2
The authors thank Holly Dressman, Duke University Microarray Facility, and Eric Porterfield for invaluable assistance with the computer analysis of the expression profiles and Jason Vittitow for critical evaluation of the manuscript. 
Leske, MC. (1983) The epidemiology of open-angle glaucoma: a review Am J Epidemiol 118,166-191 [PubMed]
Stone, EM, Fingert, JH, Alward, WLM, et al (1997) Identification of a gene that causes primary open angle glaucoma Science 275,668-670 [CrossRef] [PubMed]
Alward, WL, Fingert, JH, Coote, MA, et al (1998) Clinical features associated with mutations in the chromosome 1 open-angle glaucoma gene (GLC1A) N Engl J Med 338,1022-1027 [CrossRef] [PubMed]
Alward, WL. (2000) The genetics of open-angle glaucoma: the story of GLC1A and myocilin Eye 14,429-436 [CrossRef] [PubMed]
Wordinger, RJ, Clark, AF. (1999) Effects of glucocorticoids on the trabecular meshwork: towards a better understanding of glaucoma Prog Retin Eye Res 18,629-667 [CrossRef] [PubMed]
Polansky, JR, Fauss, DJ, Chen, P, et al (1997) Cellular pharmacology and molecular biology of the trabecular meshwork inducible glucocorticoid response gene product Ophthalmologica 211,126-139 [CrossRef] [PubMed]
Polansky, JR. (1993) HTM cell culture model for steroid effects on IOP: overview Lütjen-Drecoll, E eds. Basic Aspects of Glaucoma Research III ,307-318 Schattauer Stuttgart, Germany.
Nguyen, TD, Chen, P, Huang, WD, Chen, H, Johnson, D, Polansky, JR. (1998) Gene structure and properties of TIGR, an olfactomedin-related glycoprotein cloned from glucocorticoid-induced trabecular meshwork cells J Biol Chem 273,6341-6350 [CrossRef] [PubMed]
Ortego, J, Escribano, J, Coca-Prados, M. (1997) Cloning and characterization of subtracted cDNAs from a human ciliary body library encoding TIGR, a protein involved in juvenile open angle glaucoma with homology to myosin and olfactomedin FEBS Lett 413,349-353 [CrossRef] [PubMed]
Kubota, R, Noda, S, Wang, Y, et al (1997) A novel myosin-like protein (myocilin) expressed in the connecting cilium of the photoreceptor: molecular cloning, tissue expression, and chromosomal mapping Genomics 41,360-369 [CrossRef] [PubMed]
Clark, AF, Steely, HT, Dickerson, JE, Jr, et al (2001) Glucocorticoid induction of the glaucoma gene MYOC in human and monkey trabecular meshwork cells and tissues Invest Ophthalmol Vis Sci 42,1769-1780 [PubMed]
Johnson, DH. (1997) Corticosteroid glaucoma Epstein, DL Allingham, RR Schuman, JS eds. Chandler and Grant’s Glaucoma ,404-411 Williams & Wilkins Baltimore, MD.
Craig, JE, Snibson, GR, Alward, WLM. (1998) Exclusion of GLC1A coding region mutations as a common cause of steroid-induced ocular hypertension [ARVO Abstract] Invest Ophthalmol Vis Sci 39(4),S687Abstract nr 3153
Fingert, JH, Clark, AF, Craig, JE, et al (2001) Evaluation of the myocilin (MYOC) glaucoma gene in monkey and human steroid-induced ocular hypertension Invest Ophthalmol Vis Sci 42,145-152 [PubMed]
Caballero, M, Borrás, T. (2001) Inefficient processing of an olfactomedin-deficient myocilin mutant: potential physiological relevance to glaucoma Biochem Biophys Res Commun 282,662-670 [CrossRef] [PubMed]
Yokoe, H, Anholt, RR. (1993) Molecular cloning of olfactomedin, an extracellular matrix protein specific to olfactory neuroepithelium Proc Natl Acad Sci USA 90,4655-4659 [CrossRef] [PubMed]
Caballero, M, Rowlette, LL, Borrás, T. (2000) Altered secretion of a TIGR/MYOC mutant lacking the olfactomedin domain Biochim Biophys Acta 1502,447-460 [CrossRef] [PubMed]
Jacobson, N, Andrews, M, Shepard, AR, et al (2001) Non-secretion of mutant proteins of the glaucoma gene myocilin in cultured trabecular meshwork cells and in aqueous humor Hum Mol Genet 10,117-125 [CrossRef] [PubMed]
Kim, BS, Savinova, OV, Reedy, MV, et al (2001) Targeted disruption of the myocilin gene (Myoc) suggests that human glaucoma-causing mutations are gain of function Mol Cell Biol 21,7707-7713 [CrossRef] [PubMed]
Kirstein, L, Cvekl, A, Chauhan, BK, Tamm, ER. (2000) Regulation of human myocilin/TIGR gene transcription in trabecular meshwork cells and astrocytes: role of upstream stimulatory factor Genes Cells 5,661-676 [CrossRef] [PubMed]
Fingert, JH, Ying, L, Swiderski, RE, et al (1998) Characterization and comparison of the human and mouse GLC1A glaucoma genes Genome Res 8,377-384 [PubMed]
Chan, GC, Hess, P, Meenakshi, T, Carlstedt-Duke, J, Gustafsson, JA, Payvar, F. (1991) Delayed secondary glucocorticoid response elements J Biol Chem 266,22634-22644 [PubMed]
Shepard, AR, Jacobson, N, Fingert, JH, Stone, EM, Sheffield, VC, Clark, AF. (2001) Delayed secondary glucocorticoid responsiveness of the glaucoma gene myocilin in human trabecular meshwork cells [ARVO Abstract] Invest Ophthalmol Vis Sci 42(4),S143Abstract nr 762
Ricard, CS, Agapova, OA, Salvador-Silva, M, Kaufman, PL, Hernandez, MR. (2001) Expression of myocilin/TIGR in normal and glaucomatous primate optic nerves Exp Eye Res 73,433-447 [CrossRef] [PubMed]
Tanaka, Y, Obazawa, M, Mashima, Y, et al (2001) Cloning and characterization of porcine MYOC in cultured porcine trabecular meshwork cells and astrocytes from optic nerve head Invest Ophthalmol Vis Sci 42(4),S830Abstract nr 4455
Hernandez, MR. (2000) The optic nerve head in glaucoma: role of astrocytes in tissue remodeling Prog Retin Eye Res 19,297-321 [CrossRef] [PubMed]
Clark, AF, Kawase, K, English-Wright, S, et al (2001) Expression of the glaucoma gene myocilin (MYOC) in the human optic nerve head FASEB J ,1251-1253
Ricard, CS, Kobayashi, S, Pena, JD, Salvador-Silva, M, Agapova, O, Hernandez, MR. (2000) Selective expression of neural cell adhesion molecule (NCAM)-180 in optic nerve head astrocytes exposed to elevated hydrostatic pressure in vitro Brain Res Mol Brain Res 81,62-79 [CrossRef] [PubMed]
Vittitow, JL, Garg, R, Rowlette, LL, Epstein, DL, O’Brien, ET, Borrás, T. (2002) Gene transfer of dominant-negative RhoA increases outflow facility in perfused human anterior segment cultures Mol Vis 8,32-44 [PubMed]
Kobayashi, S, Vidal, I, Pena, JD, Hernandez, MR. (1997) Expression of neural cell adhesion molecule (NCAM) characterizes a subpopulation of type 1 astrocytes in human optic nerve head Glia 20,262-273 [CrossRef] [PubMed]
Salvador-Silva, M, Ricard, CS, Agapova, OA, Yang, P, Hernandez, MR. (2001) Expression of small heat shock proteins and intermediate filaments in the human optic nerve head astrocytes exposed to elevated hydrostatic pressure in vitro J Neurosci Res 66,59-73 [CrossRef] [PubMed]
Mi, H, Barres, BA. (1999) Purification and characterization of astrocyte precursor cells in the developing rat optic nerve J Neurosci 19,1049-1061 [PubMed]
Barbu, V, Dautry, F. (1989) Northern blot normalization with a 28S rRNA oligonucleotide probe Nucleic Acids Res 17,7115 [CrossRef] [PubMed]
Adam, MF, Belmouden, A, Binisti, P, et al (1997) Recurrent mutations in a single exon encoding the evolutionarily conserved olfactomedin-homology domain of TIGR in familial open-angle glaucoma Hum Mol Genet 6,2091-2097 [CrossRef] [PubMed]
Stamer, WD, Roberts, BC, Howell, DN, Epstein, DL. (1998) Isolation, culture, and characterization of endothelial cells from Schlemm’s canal Invest Ophthalmol Vis Sci 39,1804-1812 [PubMed]
Borrás, T, Rowlette, LL, Tamm, ER, Gottanka, J, Epstein, DL. (2002) Effects of elevated intraocular pressure on outflow facility and TIGR/MYOC expression in perfused human anterior segments Invest Ophthalmol Vis Sci 43,33-40 [PubMed]
Abraham, CR, Shirahama, T, Potter, H. (1990) Alpha 1-antichymotrypsin is associated solely with amyloid deposits containing the beta-protein: amyloid and cell localization of alpha 1-antichymotrypsin Neurobiol Aging 11,123-129 [CrossRef] [PubMed]
Taniwaki, T, Hirashima, N, Becerra, SP, Chader, GJ, Etcheberrigaray, R, Schwartz, JP. (1997) Pigment epithelium-derived factor protects cultured cerebellar granule cells against glutamate-induced neurotoxicity J Neurochem 68,26-32 [PubMed]
Kessler, E, Mould, AP, Hulmes, DJ. (1990) Procollagen type I C-proteinase enhancer is a naturally occurring connective tissue glycoprotein Biochem Biophys Res Commun 173,81-86 [CrossRef] [PubMed]
Hakala, BE, White, C, Recklies, AD. (1993) Human cartilage gp-39, a major secretory product of articular chondrocytes and synovial cells, is a mammalian member of a chitinase protein family J Biol Chem 268,25803-25810 [PubMed]
Peek, R, Kammerer, RA, Frank, S, Otte-Holler, I, Westphal, JR. (2002) The angiopoietin-like factor cornea-derived transcript 6 is a putative morphogen for human cornea J Biol Chem 277,686-693 [CrossRef] [PubMed]
Urade, Y, Hayaishi, O. (2000) Prostaglandin D synthase: structure and function Vitam Horm 58,89-120 [PubMed]
Matsuura, K, Shiraishi, H, Hara, A, et al (1998) Identification of a principal mRNA species for human 3alpha-hydroxysteroid dehydrogenase isoform (AKR1C3) that exhibits high prostaglandin D2 11-ketoreductase activity J Biochem (Tokyo) 124,940-946 [CrossRef]
Reindl, M, Knipping, G, Wicher, I, et al (2001) Increased intrathecal production of apolipoprotein D in multiple sclerosis J Neuroimmunol 119,327-332 [CrossRef] [PubMed]
Thomas, EA, Dean, B, Pavey, G, Sutcliffe, JG. (2001) Increased CNS levels of apolipoprotein D in schizophrenic and bipolar subjects: implications for the pathophysiology of psychiatric disorders Proc Natl Acad Sci USA 98,4066-4071 [CrossRef] [PubMed]
Carmel, JB, Galante, A, Soteropoulos, P, et al (2001) Gene expression profiling of acute spinal cord injury reveals spreading inflammatory signals and neuron loss Physiol Genomics 7,201-213 [CrossRef] [PubMed]
Acott, TS. (1992) Trabecular extracellular matrix regulation Drance, SM Van Buskirk, EM Neufeld, AH eds. Pharmacology of Glaucoma ,125-157 Williams & Wilkins Baltimore.
Alexander, JP, Samples, JR, Van Buskirk, EM, Acott, TS. (1991) Expression of matrix metalloproteinases and inhibitor by human trabecular meshwork Invest Ophthalmol Vis Sci 32,172-180 [PubMed]
Abraham, CR, Selkoe, DJ, Potter, H, Price, DL, Cork, LC. (1989) Alpha 1-antichymotrypsin is present together with the beta-protein in monkey brain amyloid deposits Neuroscience 32,715-720 [CrossRef] [PubMed]
Shoji, M, Hirai, S, Yamaguchi, H, Harigaya, Y, Ishiguro, K, Matsubara, E. (1991) Alpha 1-antichymotrypsin is present in diffuse senile plaques. A comparative study of beta-protein and alpha 1-antichymotrypsin immunostaining in the Alzheimer brain Am J Pathol 138,247-257 [PubMed]
Johnson, D, Gottanka, J, Flugel, C, Hoffmann, F, Futa, R, Lütjen-Drecoll, E. (1997) Ultrastructural changes in the trabecular meshwork of human eyes treated with corticosteroids Arch Ophthalmol 115,375-383 [CrossRef] [PubMed]
Clark, AF, Wilson, K, de Kater, AW, Allingham, RR, McCartney, MD. (1995) Dexamethasone-induced ocular hypertension in perfusion-cultured human eyes Invest Ophthalmol Vis Sci 36,478-489 [PubMed]
Stoilov, I, Akarsu, AN, Sarfarazi, M. (1997) Identification of three different truncating mutations in cytochrome P4501B1 (CYP1B1) as the principal cause of primary congenital glaucoma (Buphthalmos) in families linked to the GLC3A locus on chromosome 2p21 Hum Mol Genet 6,641-647 [CrossRef] [PubMed]
Gonzalez, P, Epstein, DL, Borrás, T. (2000) Characterization of gene expression in human trabecular meshwork using single-pass sequencing of 1060 clones Invest Ophthalmol Vis Sci 41,3678-3693 [PubMed]
Araki, T, Taniwaki, T, Becerra, SP, Chader, GJ, Schwartz, JP. (1998) Pigment epithelium-derived factor (PEDF) differentially protects immature but not mature cerebellar granule cells against apoptotic cell death J Neurosci Res 53,7-15 [CrossRef] [PubMed]
Cao, W, Tombran-Tink, J, Elias, R, Sezate, S, Mrazek, D, McGinnis, JF. (2001) In vivo protection of photoreceptors from light damage by pigment epithelium-derived factor Invest Ophthalmol Vis Sci 42,1646-1652 [PubMed]
Toris, CB, Camras, CB, Yablonski, ME, Brubaker, RF. (1997) Effects of exogenous prostaglandins on aqueous humor dynamics and blood-aqueous barrier function Surv Ophthalmol 41(suppl 2),S69-S75 [CrossRef] [PubMed]
Gerashchenko, DY, Beuckmann, CT, Marcheselli, VL, et al (1998) Localization of lipocalin-type prostaglandin D synthase (beta-trace) in iris, ciliary body, and eye fluids Invest Ophthalmol Vis Sci 39,198-203 [PubMed]
Hernandez, MR, Agapova, OA, Yang, P, Salvador-Silva, M, Ricard, CS, Aoi, S. (2002) Differential gene expression in astrocytes from human normal and glaucomatous optic nerve head analyzed by cDNA microarray Glia 38,45-64 [CrossRef] [PubMed]
Penning, TM, Burczynski, ME, Jez, JM, et al (2001) Structure-function aspects and inhibitor design of type 5 17beta-hydroxysteroid dehydrogenase (AKR1C3) Mol Cell Endocrinol 171,137-149 [CrossRef] [PubMed]
Weinstein, BI, Munnangi, P, Gordon, GG, Southren, AL. (1985) Defects in cortisol-metabolizing enzymes in primary open-angle glaucoma Invest Ophthalmol Vis Sci 26,890-893 [PubMed]
Southren, AL, Wandel, T, Gordon, GG, Weinstein, BI. (1994) Treatment of glaucoma with 3 alpha, 5 beta-tetrahydrocortisol: a new therapeutic modality J Ocul Pharmacol 10,385-391 [CrossRef] [PubMed]
Southren, AL, l’Hommedieu, D, Gordon, GG, Weinstein, BI. (1987) Intraocular hypotensive effect of a topically applied cortisol metabolite: 3 alpha, 5 beta-tetrahydrocortisol Invest Ophthalmol Vis Sci 28,901-903 [PubMed]
Southren, AL, Gordon, GG, l’Hommedieu, D, Ravikumar, S, Dunn, MW, Weinstein, BI. (1985) 5 beta-Dihydrocortisol: possible mediator of the ocular hypertension in glaucoma Invest Ophthalmol Vis Sci 26,393-395 [PubMed]
Yun, AJ, Murphy, CG, Polansky, JR, Newsome, DA, Alvarado, JA. (1989) Proteins secreted by human trabecular cells: glucocorticoid and other effects Invest Ophthalmol Vis Sci 30,2012-2022 [PubMed]
Pena, JD, Roy, S, Hernandez, MR. (1996) Tropoelastin gene expression in optic nerve heads of normal and glaucomatous subjects Matrix Biol 15,323-330 [CrossRef] [PubMed]
Kahari, VM. (1994) Dexamethasone suppresses elastin gene expression in human skin fibroblasts in culture Biochem Biophys Res Commun 201,1189-1196 [CrossRef] [PubMed]
Del Monaco, M, Covello, SP, Kennedy, SH, Gilinger, G, Litwack, G, Uitto, J. (1997) Identification of novel glucocorticoid-response elements in human elastin promoter and demonstration of nucleotide sequence specificity of the receptor binding J Invest Dermatol 108,938-942 [CrossRef] [PubMed]
Ledo, I, Wu, M, Katchman, S, Brown, D, Kennedy, S, Hsu-Wong, S, Uitto, J. (1994) Glucocorticosteroids upregulate human elastin gene promoter activity in transgenic mice J Invest Dermatol 103,632-636 [CrossRef] [PubMed]
Figure 1.
 
Induction of TIGR/MYOC by dexamethasone (DEX) in different cell types after 10 days of no treatment (lane C) or 10−7 M DEX treatment (lane D). (A) Northern blot analysis of total RNA from the indicated cells. Each blot was hybridized sequentially to TIGR/MYOC and 28S probes to control for RNA integrity and equal loading. (B) Western blot analysis of proteins from the indicated cells. Protein extracts were separated on 10% SDS-polyacrylamide gels and transferred to PVDF membrane. Specific proteins were detected with a TIGR/MYOC polyclonal antibody 29 and visualized by chemiluminescence. Blots were reprobed with a β-tubulin antibody for control loading. TIGR/MYOC was induced with DEX only in HTM cells. Induction occurred at transcription and translation levels. ONH-Astr, human ONH astrocytes.
Figure 1.
 
Induction of TIGR/MYOC by dexamethasone (DEX) in different cell types after 10 days of no treatment (lane C) or 10−7 M DEX treatment (lane D). (A) Northern blot analysis of total RNA from the indicated cells. Each blot was hybridized sequentially to TIGR/MYOC and 28S probes to control for RNA integrity and equal loading. (B) Western blot analysis of proteins from the indicated cells. Protein extracts were separated on 10% SDS-polyacrylamide gels and transferred to PVDF membrane. Specific proteins were detected with a TIGR/MYOC polyclonal antibody 29 and visualized by chemiluminescence. Blots were reprobed with a β-tubulin antibody for control loading. TIGR/MYOC was induced with DEX only in HTM cells. Induction occurred at transcription and translation levels. ONH-Astr, human ONH astrocytes.
Figure 2.
 
Processing and secretion of TIGR/MYOC in the different cell types. Cell lines were infected with either wild-type (AdhTIG3) or mutant (AdhTIG1, Thr345STOP) TIGR/MYOC adenovirus expression vectors at 25 to 200 pfu/cell. Proteins were extracted from supernatants (Sup) and cellular fractions (Cell) at 48 hours after infection. Equivalent volumes of cell extracts and their corresponding supernatants were run on 10% SDS-PAGE and transferred to PVDF membrane. Specific proteins were detected with a TIGR/MYOC polyclonal antibody 29 and visualized by chemiluminescence. Wild-type TIGR/MYOC (molecular mass doublet at 55–57 kDa) was secreted in every cell type. Mutant truncated TIGR/MYOC (mass ∼43 kDa) was found to be retained intracellularly in every cell type.
Figure 2.
 
Processing and secretion of TIGR/MYOC in the different cell types. Cell lines were infected with either wild-type (AdhTIG3) or mutant (AdhTIG1, Thr345STOP) TIGR/MYOC adenovirus expression vectors at 25 to 200 pfu/cell. Proteins were extracted from supernatants (Sup) and cellular fractions (Cell) at 48 hours after infection. Equivalent volumes of cell extracts and their corresponding supernatants were run on 10% SDS-PAGE and transferred to PVDF membrane. Specific proteins were detected with a TIGR/MYOC polyclonal antibody 29 and visualized by chemiluminescence. Wild-type TIGR/MYOC (molecular mass doublet at 55–57 kDa) was secreted in every cell type. Mutant truncated TIGR/MYOC (mass ∼43 kDa) was found to be retained intracellularly in every cell type.
Figure 3.
 
Confirmation of the differential expression of selected genes in DEX-treated HTM cells and ONH astrocytes by RQ RT-PCR. Amplification of the five selected genes was performed with their specific primers (Table 3) and their predetermined exponential number of cycles (shown underneath each panel). Multiplex PCR reactions for each gene were performed together with an 1:15 primer/competimer mix of 18S RNA in conditions under which the multiplex products were in the linear range. Band intensities were then captured and analyzed. Amplification of each gene was repeated three times. H1DEX cDNA in the first and third panels was diluted 100× because of the high abundance of the target gene. Lane M: ΦX174 RF DNA/HaeIII fragments. The level of expression of all selected genes changed in the same direction as the changes measured by the microarrays.
Figure 3.
 
Confirmation of the differential expression of selected genes in DEX-treated HTM cells and ONH astrocytes by RQ RT-PCR. Amplification of the five selected genes was performed with their specific primers (Table 3) and their predetermined exponential number of cycles (shown underneath each panel). Multiplex PCR reactions for each gene were performed together with an 1:15 primer/competimer mix of 18S RNA in conditions under which the multiplex products were in the linear range. Band intensities were then captured and analyzed. Amplification of each gene was repeated three times. H1DEX cDNA in the first and third panels was diluted 100× because of the high abundance of the target gene. Lane M: ΦX174 RF DNA/HaeIII fragments. The level of expression of all selected genes changed in the same direction as the changes measured by the microarrays.
Table 1.
 
Genes with Expression Levels That Changed More Than 20-fold, when A1DEX Astrocytes were compared with H1DEX Trabecular Meshwork Cells
Table 1.
 
Genes with Expression Levels That Changed More Than 20-fold, when A1DEX Astrocytes were compared with H1DEX Trabecular Meshwork Cells
GenBank Accession No. UniGene Gene Name Fold Change* Gene Function
Overexpressed in H1DEX
 AF089747 Hs.234726 α1-antichymotrypsin (SERPINA3) 478 Serine protease inhibitor
 U05861 Hs.201967 Hepatic dihydrodiol dehydrogenase (AKR1C1) 354 NADH/NADPH aldoketoreductase
 U37100 Hs.116724 Aldose reductase-like peptide (AKR1B10) 175 NADPH reductase
 Z97171 Hs.78454 Trabecular meshwork inducible gluco. resp. (MYOC) 148 Secreted glycoprotein
 D17793 Hs.78183 Aldo-ketoreductase (AKR1C3) 132 Prostaglandin D2 metabolism
 M28130 Hs.624 Interleukin 8 (IL8) 100 Chemokine, cell motility, signaling
 U29953 Hs.173594 Pigment epithelium-derived factor (PEDF) 87 Serine protease inhibitor, serpin
 AF037335 Hs.5338 Carbonic anhydrase XII (CA12) 85 Aqueous humor formation/calcification
 L33799 Hs.202097 Procollagen C-proteinase enhancer protein (PCOLCE) 79 Collagen processing
 AL050025 Hs.5344 γ1-adaptin (AP1G1) 55 Formation of clathrin pits and vesicles
 J02611 Hs.75736 Apolipoprotein D (APOD) 50 Lipocalin, secr glycpr binds hydroph mol
 M98539 Hs.8272 Prostaglandin D2 synthase (PTGDS) 49 Prostaglandin biosynthesis
 D31628 Hs.2899 4-Hydroxyphenylpyruvic acid dioxygenase (HPD) 49 Tyrosine catabolism
 L25879 Hs.89649 Epoxide hydrolase 1 (EPHX1) 43 Detoxification exogenous chemicals
 AF017786 Hs.239756 Phosphatidic acid phosphatase (PPAP2B) 41 Phosphatidylinositol (PI) turnover
 D00654 Hs.78045 γ2-actin, enteric smooth muscle (ACTG2) 40 Cytoskeletal network
 AF015926 Hs.184276 Solute carrier family 9 (SLC9A3R1) 39 Sodium/hydrog exchange/actin organization
 A1445461 Hs.351316 Transmembrane 4 superfamily member 1 (TM4SF1) 39 Tetraspanin, regulates basic cell processes
 M90657 Hs.351316 Tumor-associated antigen L6 (TAAL6) 39 Tetraspanin, regulates basic cell processes
 AL049653 Hs.146559 Angiopoietin-like factor (CDT6) 37 Anti-angiogenesis/ECM deposition
 M17017 Hs.624 Interleukin 8 (IL8) 32 Chemokine, cell motility
 U09937 Hs.179657 Plasminogen activator urokinase receptor (PLAUR) 31 Signal transduction/ECM remodeling
 Y18483 Hs.22891 Solute carrier family 7 (SLC7A8) 31 Amino acid transporter
 M25915 Hs.75106 Clusterin (CLU) 29 Plasma glycoprotein
 M76665 Hs.37012 Hydroxysteroid dehydrogenase (HSD11B1) 27 Conversion of cortisol to cortisone
 L26232 Hs.154854 Phospholipid transfer protein (PLTP) 26 Plasma lipid transfer
 X01630 Hs.160786 Argininosuccinate synthetase (ASS) 26 Nitric oxide synthesis/urea cycle
 L38486 Hs.118223 Microfibril-associated glycoprotein 4 (MFAP4) 26 Integrin ligand, extracellular matrix
 AI936826 Hs.85339 G protein-coupled receptor 39 (GPR39) 25 Signal transduction
 AF096870 Hs.194540 Estrogen-responsive B box protein (EBBP) 24 Zinc finger protein
 L13698 Hs.65029 Growth arrest-specific 1 (GAS1) 24 Cell growth inhibition
 J04164 Hs.146360 Interferon induced transmembrane protein 1 (IFITM1) 22 Cell growth inhibition
 U96750 Hs.194695 Ras homologue gene family member 1 (ARH1) 22 Cell growth inhibition
 AB003184 Hs.102171 Immunoglobulin superfamily leucine-rich (ISLR) 22 Protein binding, cell adhesion
 X14885 Hs.2025 Transforming growth factor β3 (TGFB3) 21 Cell proliferation and differenciation
 X04470 Hs.251754 Secretory leukocyte protease inhibitor (SLPI) 21 Protease (chymotrypsin) inhibitor
 AF060567 Hs.126782 Sushi repeat protein (SRPUL) 21 Deleted in RP, unknown function
 L13698 Hs.65029 Growth arrest-specific 1 (GAS1) 21 Cell growth inhibition
 AF027734 Hs.6090 Deleted in bladder cancer chromosomal region candidate 1 (DBCCR1) 21 Tumor suppressor
 AI381790 Hs.74120 Adipose-specific 2 (APM2) 21 Unknown/specific to adipose tissue
Underexpressed in H1DEX
 AB018301 Hs.22039 KIAA0758 protein (KIAA0758) −112 Unknown
 X52896 Hs.9295 Elastin (ELN) −110 Elastic fiber component/connective tissue
 U61849 Hs.84154 Neuronal pentraxin 1 (NPTX1) −68 Snake venom-binding protein
 Z24680 Hs.151641 Glycoprotein A repetitions predominant (GARP) −63 Unknown
 M18533 Hs.169470 Dystrophin (DMD) −42 Anchoring cytoskeleton to membrane
 U30521 Hs.142827 P311 protein (P311) −41 Cell transformation and motility
 AF045800 Hs.40098 Gremlin/bone morphogenetic protein antagonist1 (CKTSF1B1) −39 Secreted/blocks BMP signaling
 AB023194 Hs.300855 KIAA0977 protein (KIAA0977) −37 Unknown
 D83402 Hs.302085 Prostacyclin synthase (PTGIS) −35 Potent vasodilator/P450 superfamily
 M26326 Hs.65114 Keratin 18 (KRT18) −35 Intermediate filament component
 W27472 Hs.24391 Hypothetical protein FLJ13612 (FLJ13612) −33 Unknown
 Y00630 Hs.75716 Plasminogen activator-inhibitor (SERPINB2) −30 Inhibitor extracellular proteolysis
 M97936 Hs.21486 Transcription factor (STAT1) (ISGF-3) −29 Activator of transcription
 X15998 Hs.81800 Chondroitin sulfate proteoglycan 2 (CSPG2) −29 ECM glycoprotein/binds to hyaluronan
 AL031058 Hs.74316 Desmoplakin I (DPI) −26 Cytoskeleton/intercellular adhesion
 L06139 Hs.89640 Receptor tyrosine kinase (TEK) −25 Cells of endothelial lineage
 X15998 Hs.81800 Chondroitin sulphate proteoglycan 2 (CSPG2) −25 ECM glycoprotein/binds to hyaluronan
 M36860 Hs.9295 Elastin (ELN) −23 Elastic fiber component/connective tissue
Table 2.
 
The Most Overexpressed HTM-DEX–Specific Genes
Table 2.
 
The Most Overexpressed HTM-DEX–Specific Genes
GenBank Accession No. UniGene Fold Change Gene Name Gene Group* Gene Function
A1DEX vs. H1DEX A1DEX vs. H2DEX A2DEX vs. H1DEX A2DEX vs. H2DEX Average
AF089747 Hs.234726 478 125 42 13 165 α1-antichymotrypsin (SERPINA3) a Serine protease inhibitor
U05861 Hs.201967 354 69 66 13 126 Hepatic dihydrodiol dehydrogenase (AKR1C1) b NADH/NADPH aldoketoreductase
BC004312 Hs.162 6 5 113 102 57 Insulin-like growth factor binding protein-2 (IGFBP2) a Modulates IGF function
Z97171 Hs.78454 148 18 30 5 50 Trabecular Meshwork inducible gluco. resp. (MYOC) a Secreted glycoprotein
U29953 Hs.173594 87 59 20 18 46 Pigment epithelium-derived factor (PEDF) a Neuron survival factor
L33799 Hs.202097 79 60 18 17 44 Procollagen C-proteinase enhancer (PCOLCE) c Collagen processing
D17793 Hs.78183 132 27 12 3 43 Aldo-keto reductase (AKR1C3) a Prostaglandin D2 metabolism
M98539 Hs.8272 49 65 13 25 38 Prostaglandin D2 synthase (PTGDS) a Prostaglandin biosynthesis
Y08374 Hs.75184 12 91 3 24 33 Cartilage GP-39 (CHI3L1) a Extracellular matrix remodeling
D00654 Hs.78045 40 57 9 15 30 γ2-actin, enteric smooth muscle (ACTG2) a Cytoskeletal network
AL049653 Hs.146559 37 49 10 17 28 Angiopoietin-like factor (CDT6) a Antiangiogenesis/ECM deposition
X01630 Hs.160786 26 58 8 18 28 Argininosuccinate synthetase (ASS) b Nitric oxide synthesis/urea cycle enzyme
X96924 Hs.111024 31 42 8 12 23 Citrate transport protein (CTP) a Citrate mitochondrial transmembrane transport
J02611 Hs.75736 50 20 12 5 22 Apolipoprotein D (APOD) a Lipocalin/secreted glycoprotein binds hydrophobic molecules
AL050025 Hs.5344 55 7 21 3 21 γ1-adaptin (AP1G1) a Formation of clathrin pits and vesicles, protein sorting, and traffic
Table 3.
 
Primers and Amplification Products Used in RQ RT-PCR Analysis
Table 3.
 
Primers and Amplification Products Used in RQ RT-PCR Analysis
Gene Name Accession No. Sense Primer Antisense Primer Size (bp)
TIGR/MYOC NM_000261 5′-CTG GAG GAA GAG AAG AAG CGA CTA A-3′ 5′-CTG TGT CAT AAG CAA AGT TGA CGG TA-3′ 912
α1-antichymotrypsin AF089747 5′-AAG CTC ATC AAC GAC TAC GTG A-3′ 5′-ATT CCA AGT TCC TTA CTG AGA GCC-3′ 969
PEDF M76979 5′-GGC TGT CTC CAA CTT CGG CTA-3′ 5′-TGG TGA CTT CGC CTT CGT AAC T-3′ 811
Prostaglandin D2 synthase BC005939 5′-CCA ACT TCC AGC AGG ACA AGT TCC-3′ 5′-CTT CCG GAG TTT ATT GTG CAB GAT-3′ 633
PCOLCE L33799 5′-CTA ATA AGG AGT GCA TCT GGA CCA TAA-3′ 5′-CTG AAG GAG ATT CCT CTG TTT TCT C-3′ 691
Table 4.
 
H1DEX Overexpressed Specific Genes Located in Chromosomal Regions Linked to Glaucoma
Table 4.
 
H1DEX Overexpressed Specific Genes Located in Chromosomal Regions Linked to Glaucoma
GenBank Accession No. UniGene Gene Name Fold Change Glaucoma-Linked Chromosomal Region
Z97171 Hs.78454 Trab Mesh inducible gluco. resp. (MYOC)* 148 1q24.3-q25.2
AI445461 Hs.351316 Transmembr 4 superfamily1 (TM4SF1) 39 3q21-q25
M90657 Hs.351316 Tumor-associated antigen L6 (TAAL6) 39 3q21-q25
AL049653 Hs.146559 Angiopoietin-like factor (CTD6)* 37 1p36
U30255 Hs.75888 Phosphogluconate dehydrogenase (PGD) 19 1p36.2-p36.13
AI660929 Hs.135150 Type-I membrane-asso glycoprotein (T1A-2)* 12 1p36
AF030428 Hs.135150 Type-I membrane-asso glycoprotein (T1A-2)* 11 1p36
D83492 Hs.3796 Ephrin (kinase) receptor protein (EPHB6) 9 7q33-q35
X15414 Hs.75313 Aldose reductase (AR) 9 7q35
D28124 Hs.76307 Human mRNA for unknown product 6 1p36.3-p36.2
M94345 Hs.82422 Capping protein (actin filament) (CAPG) 5 2cen-q24
AF044253 Hs.298184 Potassium channel (KCNAB2) 5 1p36.3
U50648 Hs.274382 Protein kinase, interferon inducible (PRKR) 5 2p22-p21
M64231 Hs.76244 Spermidine synthase (SRM) 5 1p36-p22
M55914 Hs.254105 Enolase 1 (ENO1) 4 1p36.3-p36.2
×
×

This PDF is available to Subscribers Only

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

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

×