Identification of pathogenic genes and transcription factors in glaucoma

  • Authors:
    • Jie Feng
    • Jing Xu
  • View Affiliations

  • Published online on: May 14, 2019     https://doi.org/10.3892/mmr.2019.10236
  • Pages: 216-224
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Glaucoma is a group of eye diseases characterized by alterations in the contour of the optic nerve head, with corresponding visual field defects and progressive loss of retinal ganglion cells. The present study aimed to identify the key genes and upstream regulators in glaucoma. To screen the pathogenic genes involved in glaucoma, an integrated analysis was performed by using the microarray datasets in glaucoma derived from the Gene Expression Omnibus (GEO) database. The functional annotation and potential pathways of differentially expressed genes (DEGs) were additionally examined by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. A glaucoma‑specific transcriptional regulatory network was constructed to identify crucial transcriptional factors that target the DEGs in glaucoma. From two GEO datasets, 1,935 DEGs (951 upregulated and 984 downregulated genes) between glaucoma and normal controls were identified. GO and KEGG analyses identified that ‘eye development’ [false discovery rate (FDR)=0.00415533] and ‘visual perception’ (FDR=0.00713283) were significantly enriched pathways for DEGs. The expression of lipocalin 2 (LCN2), monoamine oxidase A (MAOA), hemoglobin subunit β (HBB), paired box 6 (PAX6), fibronectin (FN1) and cAMP responsive element binding protein 1 (CREB1) were demonstrated to be involved in the pathogenesis of glaucoma. In conclusion, LCN2, MAOA, HBB, PAX6, FN1 and CREB1 may serve roles in glaucoma, regulated by PAX4, solute carrier family 22 member 1, hepatocyte nuclear factor 4 α and ELK1, ETS transcription factor. These data may contribute to the development of novel potential biomarkers, reveal the underlying pathogenesis and additionally identify novel therapeutic targets for glaucoma.

Introduction

Glaucoma is a widely known, multi-factorial disease, which may result in apoptosis of retinal ganglion cells. According to the World Health Organization, glaucoma is the second principal cause of blindness and the most common cause of irreversible blindness in the world (1,2). In glaucoma, the anterior and posterior segments of the eye are affected, and serious damage may be detected in the trabecular meshwork (3). Oxidative stress is considered to be responsible for the molecular damage in the anterior chamber. Primary open-angle glaucoma (POAG) is the most common type of glaucoma, accounting for 60–70% all glaucoma (4). A candidate protein that may be associated with POAG is myocilin (MYOC), encoded by the MYOC gene. MYOC mutations are common in patients with POAG with high levels of intraocular pressure (IOP) (5,6). Additionally, mutations in optineurin were identified in patients with POAG (7). Previous studies suggested that an abnormal expression of serine/threonine-protein kinase TBK1 is a cause of normal-tension glaucoma (810). Furthermore, a previous study suggested that the calcium load-activated calcium channel was involved in glaucoma and that cyclin-dependent kinase 4 inhibitor B antisense RNA 1 was upregulated in the retina of a rat model of glaucoma (11).

However, even substantial decreases in IOP are not able to prevent the development and progression of glaucoma in a number of clinical cases (12). Glaucoma-associated cell death is primarily caused by apoptosis, which is triggered by oxidative stress via mitochondrial damage, inflammation, endothelial dysregulation and dysfunction, and hypoxia (13). In general, glaucoma is not preventable; however, the vast majority of patients may maintain useful visual function for life if they have early detection and appropriate treatment (14). Therefore, for the prevention of glaucoma, emphasis must be placed on early detection, and early diagnosis and treatment.

The rapid development and application of high-throughput sequencing technology has provided a comprehensive and rapid analytical method for the study of the pathogenesis of glaucoma, and provide novel ideas for the future treatment of glaucoma (15). The present study aimed to analyze high-throughput transcriptome data from tissue samples of patients with glaucoma and a normal control group. The data was used in bioinformatics analyses to identify key transcription factors (TFs) associated with glaucoma, to examine the pathogenesis of glaucoma and provide a basis for the diagnosis of glaucoma and drug development.

Materials and methods

Microarray expression profiling in Gene Expression Omnibus (GEO)

The GEO is the largest database of high-throughput gene expression data that was developed and is maintained by the National Center for Biotechnology Information (16). The GEO was searched to obtain gene expression profiling studies of glaucoma subjects. The following key search terms were used: [‘glaucoma’ (Medical subject headings Terms) OR ‘glaucoma’ (All Fields)] AND ‘Homo sapiens’ (porgn) AND ‘gse’ (Filter). The selection criteria were as follows: i) The selected dataset must include genome-wide mRNA transcriptome data; ii) the data was obtained from the trabecular meshwork tissue samples of glaucoma and normal control trabecular meshwork tissue samples; and iii) normalized and raw datasets were considered. Following selection, two sets of GSE27276 (17) and GSE4316 (18) glaucoma mRNA data were obtained (19,20).

Identification of differentially expressed genes (DEGs) in glaucoma compared with normal controls

Background correction was performed on the raw data. The normalization was performed using the Linear Models for Microarray (Limma version 3.30.13) Data package in R (21). Subsequently, two-tailed Student's t-tests were performed to calculate individual P-values. Stouffer's test was used to merge individual P-values, and multiple comparison correction was performed using the Benjamini and Hochberg method to obtain the false discovery rate (FDR) (22). Genes with FDR<0.001 were selected as DEGs. Finally, the DEGs in glaucoma vs. normal were identified.

Functional annotation of DEGs

Gene Ontology (GO) (23) and Kyoto Encyclopedia of Genes and Genomes (KEGG) (24) pathway enrichment analysis were performed to detect the biological functions and potential pathways associated with DEGs using GeneCoDis3 (http://genecodis.cnb.csic.es/analysis) as previously described (19). The GO functions of the DEGs were determined according to the three categories of: ‘Biological process’; ‘molecular functions’; and ‘cellular component’. Pathway enrichment analysis was based on the KEGG database, as previously described (25).

Protein-protein interaction (PPI) network construction

In order to identify candidate genes involved in the formation of glaucoma, PPI networks of significant DEGs were constructed, according to the data from Biological General Repository for Interaction Datasets (BioGRID; http://thebiogrid.org/). Based on the existing data of protein interaction in the BioGRID database, Cytoscape (www.cytoscape.org/; (version 3.5.0) was used to search the top 100 upregulated and downregulated mRNAs. The PPI network interaction map was generated subsequent to the exclusion of genes that were not differentially expressed.

Screening for TFs of the top 20 DEGs and construction of TF regulatory network

For the top 20 DEGs, the 2 kb upstream promoter regions were downloaded from the University of California Santa Cruz (UCSC) Genome Browser website (genome.ucsc.edu). The TRANScription FACtor (TRANSFAC) website match tool (gene-regulation.com/pub/databases.html) was subsequently used to analyze TFs capable of binding to the promoter region of the DEGs. TFs that exhibited altered expression in glaucoma with FDR<0.001 were selected. Position Weight Matrix scanning was used to scan the human genome sequence to obtain the protein-coding genes that were regulated by the differentially expressed TFs. Following removal of redundant information, the glaucoma-specific transcriptional regulatory network was constructed using Cytoscape software.

In silico validation of DEGs using GEO

The GEO database (GSE9944) was used to validate the expression of selected glaucoma DEGs. The expression levels of these genes were compared between the glaucoma cases and the normal group. The expression of five genes, cAMP responsive element binding protein 1 (CREB1), fibronectin 1 (FN1), keratin 19 (KRT19), lipocalin 2 (LCN2) and paired box 6 (PAX6) was detected, with the difference of expression levels presented as box-plots.

Results

Differential expression analysis of genes in glaucoma

The probes corresponding to multiple genes were removed, and the average gene expression to which multiple probes corresponded with was calculated. Finally, the intersection of 15,757 genes was obtained.

A total of two gene expression microarray datasets (GSE27276 and GSE4316) were used for the analysis. Compared with the normal controls, 1,935 DEGs in glaucoma were obtained (P<0.05); among these, 951 genes were upregulated and 984 genes were downregulated. The top 40 most significantly up- or downregulated genes are summarized in Table I. Among which, PAX6 (26), LCN2 (27), and MAOA (28) were downregulated and were associated with glaucoma. The DEGs were screened for clustering analysis. The heatmap produced by cluster analysis of the two sets of cDNA microarray data is presented in Fig. 1.

Table I.

Top 40 differentially expressed mRNAs.

Table I.

Top 40 differentially expressed mRNAs.

A, Upregulated

IDSymbolCombined.ESP-valueFDR
3043HBB5.1601833700
9415FADS22.35332790 1.40×10−9 3.68×10−6
56172ANKH2.33974344 7.72×10−9 1.11×10−5
3113HLA-DPA12.22895504 1.17×10−8 1.41×10−5
5538PPT12.27584322 1.72×10−8 1.69×10−5
83643CCDC32.09336726 2.56×10−8 2.32×10−5
81618ITM2C2.10210833 5.11×10−8 3.81×10−5
8857FCGBP2.00251174 1.07×10−7 6.25×10−5
4166CHST61.94047822 1.48×10−7 7.07×10−5
4753NELL21.97462732 2.45×10−7 1.07×10−4
8718TNFRSF251.80276729 8.11×10−7 2.84×10−4
81552VOPP11.82118066 8.70×10−7 2.91×10−4
2191FAP1.74786612 1.01×10−6 3.09×10−4
187APLNR1.74800187 1.02×10−6 3.09×10−4
51196PLCE11.77349072 1.23×10−6 3.61×10−4
23676SMPX1.76299253 1.37×10−6 3.78×10−4
51226COPZ21.72176939 1.72×10−6 4.24×10−4
1290COL5A21.71261435 1.78×10−6 4.31×10−4
57559STAMBPL11.69907540 1.81×10−6 4.31×10−4
2331FMOD1.75952553 1.83×10−6 4.31×10−4

B, Downregulated

IDSymbol Combined.ESP-valueFDR

3880KRT19−2.54377891 3.23×10−10 1.70×10−6
116039OSR2−2.44873456 7.08×10−10 2.23×10−6
5648MASP1−2.39197514 2.01×10−9 4.52×10−6
79845RNF122−2.31970767 3.00×10−9 5.91×10−6
3934LCN2−2.20821475 8.44×10−9 1.11×10−6
57801HES4−2.22824712 1.65×10−8 1.69×10−5
10232MSLN−2.09546485 2.65×10−8 2.32×10−5
9245GCNT3−2.11365461 4.07×10−8 3.38×10−5
84525HOPX−2.01200085 5.57×10−8 3.81×10−5
4128MAOA−2.03058309 7.95×10−8 5.22×10−5
5080PAX6−2.09500899 8.92×10−8 5.62×10−5
64073C19orf33−2.02417662 1.26×10−7 6.86×10−5
3866KRT15−1.96990194 1.40×10−7 6.92×10−5
7148TNXB−1.92147744 1.40×10−7 6.92×10−5
163732CITED4−1.94673475 1.94×10−7 9.01×10−5
1675CFD−1.97823285 2.68×10−7 1.14×10−4
6510SLC1A5−1.95192053 2.77×10−7 1.15×10−4
1638DCT−1.84682776 3.22×10−7 1.30×10−4
3861KRT14−1.77674842 7.12×10−7 2.61×10−4
10053AP1M2−1.77498212 8.52×10−7 2.92×10−4

[i] FDR, false discovery rate.

Functional annotation

In Fig. 2, GO enrichment demonstrated that the DEGs were significantly enriched the ‘Biological processes’ categories: ‘eye development’ (FDR=4.15×10−3); ‘visual perception’ (FDR=7.13×10−3); ‘negative regulation of insulin receptor signaling pathway’ (FDR=1.47×10−2); the ‘Cellular components’ categories: ‘membrane’ (FDR=3.41×10−52); ‘endoplasmic reticulum’ (FDR=3.76×10−12); ‘cytoplasm’ (FDR=1.73×10−63); and ‘Molecular functions’ categories: ‘nucleotide binding’ (FDR=8.50×10−16); ‘hydrolase activity’ (FDR=8.50×10−16); and ‘protein binding’ (FDR=6.15×10−60).

Furthermore, as presented in Fig. 3, the results of the KEGG pathway enrichment demonstrate that DEGs were enriched in ‘Staphylococcus aureus infection’ (FDR=6.15×10−60); ‘Pathways in cancer’ (FDR=8.66×10−5); ‘Systemic lupus erythematosus’; and ‘Type I diabetes mellitus’ (FDR=0.84×10−3).

PPI network construction

Following removal of the non-DEGs, the PPI network was established. The results are presented in Fig. 4. The network consisted of nodes and edges. The nodes in the network represent the proteins and the edges represent the interactions between them. There were 290 nodes and 290 edges identified. Among them, the genes with higher degrees were: FN1 (degree=92); KRT15 (degree=19); major histocompatibility complex, class II, DP α 1 (HLA-DPA1; degree=12); CREB1 (degree=11); KRT19 (degree=9); histone cluster 2 H2B family member e (degree=9); cell division cycle associated 7 like (degree=7); PAX6 (degree=7); FOS like 2, AP-1 transcription factor subunit (degree=7); minichromosome maintenance complex component 2 (degree=6); palmitoyl-protein thioesterase 1 (degree=6); and lysine demethylase 5B (degree=6). The hub proteins were FN1 (degree=92), KRT15 (degree=19) and HLA-DPA1 (degree=12).

TFs of the top 20 DEGs and TF regulation network

A total of 250 TF binding associations were identified, including multiple binding sites of the same TF in a gene. Among these, 36 TFs were involved. In Table II, the top six TFs with the most downstream genes (the top 20 differentially expressed genes) were included: PAX4; solute carrier family 22 member 1 (1-Oct); hepatocyte nuclear factor 4 α (HNF-4); NK2 homeobox 5 (Nkx2-5); PAX6; and ELK1, ETS transcription factor (Elk-1).

Table II.

Top six TFs with the highest number of downstream regulatory genes and their target genes.

Table II.

Top six TFs with the highest number of downstream regulatory genes and their target genes.

TF nameNumber of regulated genesRegulated genes
Pax-411HES4, ITM2C, ANKH, HLA-DPA1, RNF122, LCN2, CCDC3, NELL2, MSLN, CHST6, KRT19
1-Oct11ITM2C, MASP1, HOPX, ANKH, HLA-DPA1, OSR2, RNF122, CCDC3, HBB, NELL2, MSLN
HNF-411ITM2C, MASP1, HLA-DPA1, LCN2, MAOA, HBB, FADS2, NELL2, MSLN, CHST6, KRT19
Nkx2-5  9PPT1, HOPX, RNF122, CCDC3, FADS2, NELL2, MSLN, CHST6, KRT19
Pax-6  8ITM2C, MASP1, ANKH, OSR2, CCDC3, NELL2, GCNT3, KRT19
Elk-1  5ITM2C, MASP1, ANKH, LCN2, CCDC3, FADS2, CHST6

[i] TF, transcription factor.

Cytoscape software was used to identify the TFs and the differentially expressed regulatory network of their target genes (Fig. 5). There were 61 nodes, including 44 transcription factors and 20 differentially expressed target genes and 132 edges identified. The eight target genes with the highest degrees were: Coiled-coil domain containing 3 (degree=14); neural tissue-specific epidermal growth factor-like repeat domain-containing protein (NELL2; degree=13); hes family bHLH transcription factor 4 (degree=10); mesothelin (degree=10); integral membrane protein 2C (degree=9); fatty acid desaturase 2 (degree=9); mannan binding lectin serine peptidase 1 (degree=8); and hemoglobin subunit b (HBB; degree=8).

Validation of DEGs in GEO GSE9944 dataset

To define the key genes with key roles in glaucoma, the GEO GSE9944 repository was searched for high throughput gene expression data and hybridization arrays, chips and microarrays. As demonstrated in Fig. 6, the genes CREB1 and FN1 were upregulated, and the genes KRT19, LCN2 and PAX6 were downregulated. These results were consistent with the integrated analysis in the GSE27276 and GSE4316 datasets.

Discussion

Glaucoma is the leading cause of irreversible blindness worldwide and there is no effective treatment at present (27). In the present study, integrated analysis was performed using data obtained from the GEO database. KEGG, GO and other biological information databases, and R analysis tools were used to analyze the DEGs. A total of 1,935 DEGs in glaucoma (951 genes were upregulated, 984 genes were downregulated) were obtained. Critical signaling pathways that affected the pathogenesis of glaucoma, including ‘eye development’ (FDR=0.00415533) and ‘visual perception’ (FDR=0.00713283) were identified. In addition, based on the promoter sequence of DEGs obtained from UCSC, a TF regulatory network was constructed using the match tool of the TRANSFAC website to obtain the corresponding TFs.

LCN2, encoded by lcn2 gene, is a neutrophil gelatinase-associated lipocalin. The 25-kD LCN2 protein is able to bind small lipophilic substances, including bacteria-derived lipopolysaccharide and formylpeptides (28). The protein additionally functions as a modulator of inflammation. Khalyfa et al (29) suggested that LCN2 was highly upregulated in glaucoma. In the results of the present study, LCN2 was downregulated and the validation result in the GSE9944 dataset was consistent with the analysis. A previous study additionally demonstrated that the coadministration of a reversible monoamine oxidase A (MAOA) inhibitor with epinephrine may be useful for patients with glaucoma (28). In the results of the present study, MAOA was downregulated.

CREB1 is a transcription regulator activated in response to harmful stress stimuli, including hypoxia and oxidative stress, and is involved in the cellular defense against these stresses (30). High IOP, optic nerve damage and visual field defects are the primary pathological features of glaucoma, with high IOP being the most common (27). CREB has been demonstrated to promote neuronal survival (31), and a previous study indicated that CREB has a neuroprotective effect against hydrogen peroxide-induced retinal ganglion cell death via 2 downstream cell survival genes, including brain-derived neurotrophic factor and apoptosis regulator Bcl-2 (32). In a study by Yasuda et al (12), an in silico pathway analysis conducted in rats with axonal injury induced by transecting the right optic nerve, CREB1 was the most significant upstream regulator. In the present study, CREB1 (degree=11) was the in the top 3 genes with the highest degrees in the PPI network. In the KEGG analysis, CREB1 was enriched in the function of Huntington's disease.

FN1 is a glycoprotein within the extracellular matrix, which has a crucial role in cell adhesion, growth, migration and differentiation (33,34). Cellular FN is secreted in a soluble form by a variety of different cell types, including fibroblasts, chondrocytes, macrophages and epithelial cells, and is later assembled into an insoluble extracellular matrix (35). FN has a multimodular structure organized into functional domains that interact with multiple binding partners, including integrins, heparin sulfate proteoglycans, collagen, glycosaminoglycans, proteoglycans, heparin, fibrin and bacteria (36). A previous study by Anshu et al (2) identified that FN1 (P=0.0003) was expressed at a significantly increased level in aqueous humor samples compared with controls. In the present study, FN1 was the top protein with the highest degree in the PPI network. Additionally, in the KEGG analysis, FN1 was enriched in the function of ‘Focal adhesion’.

NELL2 is a secreted glycoprotein that is predominantly expressed in neural tissues. NELL2 is additionally involved in promoting the neuronal survival required for the formation of a sexually dimorphic nucleus of the preoptic area in male rats (37). In the TF regulation network in the present study, NELL2 (degree=13) was the in top 2 genes and was regulated by PAX4, 1-Oct, HNF-4, Nkx2-5 and PAX6. Additionally, NELL2 was among the top 40 DEGs. These results indicated that NELL2 may have a key role in the pathogenesis of glaucoma.

The OMIM database was also utilized in the present study, and it was identified that PAX6 was key gene. PAX6 is a member of the paired box gene family, which encodes a transcriptional regulator involved in oculogenesis and other developmental processes. Halder et al (38) suggested that the ectopic expression of Drosophila PAX6 induces ectopic eye development. Wawersik and Maas (39) reviewed the role of PAX6 and other genes in vertebrate and fly oculogenesis; the study demonstrated that regulators of eye development are conserved across large evolutionary distances. The expression patterns and conserved functional domains suggested that Pax6 may have important roles in vertebrate eye formation. Fujimura et al (40) demonstrated that PAX6 appears to function as a pleiotropic regulator, directing the development of ocular tissues in concert with the signaling pathway and, concomitantly, regulating the expression of structural components of the eye, including shielding pigment. In the present study, PAX6 was downregulated in the patients with glaucoma. In addition, in the PPI network, PAX6 was one of the top eight genes with the highest degrees.

In conclusion, six DEGs (LCN2, MAOA, HBB, PAX6, FN1 and CREB1) were identified to be involved in the process of glaucoma. From the two GEO datasets analyzed, 1,935 DEGs (951 upregulated and 984 downregulated genes) were identified between glaucoma and normal controls. DEG validation in the GEO GSE9944 dataset was consistent with the integrated analysis of the GSE27276 and GSE4316 datasets. These results may contribute to the elucidation of novel potential biomarkers, reveal the underlying pathogenesis and identify novel therapeutic targets for the treatment of glaucoma.

Acknowledgements

Not applicable.

Funding

The present study was supported by Science and Technology Development Plan of Jining [grant no. (2016) 56–31] and Project Medical and Health Technology Development Project of Shandong (grant no. 2017WS528).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

JF and JX conceived the study and wrote the manuscript. JX performed the data analyses. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Tham YC, Li X, Wong TY, Quigley HA, Aung T and Cheng CY: Global prevalence of glaucoma and projections of glaucoma burden through 2040: A systematic review and meta-analysis. Ophthalmology. 121:20812014. View Article : Google Scholar : PubMed/NCBI

2 

Anshu A, Price MO, Richardson MR, Segu ZM, Lai X, Yoder MC and Price FW Jr: Alterations in the aqueous humor proteome in patients with a glaucoma shunt device. Mol Vis. 17:1891–1900. 2011.PubMed/NCBI

3 

Saccà SC, Gandolfi S, Bagnis A, Manni G, Damonte G, Traverso CE and Izzotti A: From DNA damage to functional changes of the trabecular meshwork in aging and glaucoma. Ageing Res Rev. 29:26–41. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Kayange PC, Nkume HB, Feyi-Waboso A, Kalua K, Msukwa G and Schwering Schulze M: Presentation of primary open angle glaucoma (POAG) at lions sight first eye hospital in Blantyre, Malawi. Malawi Med J. 26:60–62. 2014.PubMed/NCBI

5 

Kwon YH, Fingert JH, Kuehn MH and Alward WL: Primary open-angle glaucoma. N Engl J Med. 2:1113–1124. 2009. View Article : Google Scholar

6 

Nazir S, Mukhtar M, Shahnawaz M, Farooqi S, Fatima N, Mehmood R and Sheikh N: A novel single nucleotide polymorphism in exon 3 of MYOC enhances the risk of glaucoma. PLoS One. 13:e01951572018. View Article : Google Scholar : PubMed/NCBI

7 

Huang C, Xie L, Wu Z, Cao Y, Zheng Y, Pang CP and Zhang M: Detection of mutations in MYOC, OPTN, NTF4, WDR36 and CYP1B1 in Chinese juvenile onset open-angle glaucoma using exome sequencing. Sci Rep. 8:44982018. View Article : Google Scholar : PubMed/NCBI

8 

Fingert JH, Robin AL, Stone JL, Roos BR, Davis LK, Scheetz TE, Bennett SR, Wassink TH, Kwon YH, Alward WL, et al: Copy number variations on chromosome 12q14 in patients with normal tension glaucoma. Hum Mol Genet. 20:2482–2494. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Kawase K, Allingham RR, Meguro A, Mizuki N, Roos B, Solivan-Timpe FM, Robin AL, Ritch R and Fingert JH: Confirmation of TBK1 duplication in normal tension glaucoma. Exp Eye Res. 96:178–180. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Ritch R, Darbro B, Menon G, Khanna CL, Solivan-Timpe F, Roos BR, Sarfarzi M, Kawase K, Yamamoto T, Robin AL, et al: TBK1 gene duplication and normal-tension glaucoma. JAMA Ophthalmol. 132:544–548. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Burdon KP, Macgregor S, Hewitt AW, Sharma S, Chidlow G, Mills RA, Danoy P, Casson R, Viswanathan AC, Liu JZ, et al: Genome-wide association study identifies susceptibility loci for open angle glaucoma at TMCO1 and CDKN2B-AS1. Nat Genet. 43:574–578. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Yasuda M, Tanaka Y, Omodaka K, Nishiguchi KM, Nakamura O, Tsuda S and Nakazawa T: Transcriptome profiling of the rat retina after optic nerve transection. Sci Rep. 6:287362016. View Article : Google Scholar : PubMed/NCBI

13 

Shi Z, Rudzinski M, Meerovitch K, Lebrun-Julien F, Birman E, Di Polo A and Saragovi HU: Alpha2-macroglobulin is a mediator of retinal ganglion cell death in glaucoma. J Biol Chem. 283:29156–29165. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Konstas AG, Maskaleris G, Gratsonidis S and Sardelli C: Compliance and viewpoint of glaucoma patients in greece. Eye (Lond). 14:752–756. 2000. View Article : Google Scholar : PubMed/NCBI

15 

Tajiri-Utagawa E, Hara M, Takahashi K, Watanabe M and Wakita T: Development of a rapid high-throughput method for high-resolution melting analysis for routine detection and genotyping of noroviruses. J Clin Microbiol. 47:435–440. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Chang WS, Wang YH, Zhu XT and Wu CJ: Genome-wide profiling of miRNA and mRNA expression in Alzheimer's disease. Med Sci Monit. 23:2721–2731. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Liu Y, Allingham RR, Qin X, Layfield D, Dellinger AE, Gibson J, Wheeler J, Ashley-Koch AE, Stamer WD and Hauser MA: Gene expression profile in human trabecular meshwork from patients with primary open-angle glaucoma. Invest Ophthalmol Vis Sci Sep. 54:6382–6389. 2013. View Article : Google Scholar

18 

Liton PB, Luna C, Challa P, Epstein DL and Gonzalez P: Genome-wide expression profile of human trabecular meshwork cultured cells, nonglaucomatous and primary open angle glaucoma tissue. Mol Vis. 12:774–790. 2006.PubMed/NCBI

19 

Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, Marshall KA, Phillippy KH, Sherman PM, Holko M, et al: NCBI GEO: Archive for functional genomics data sets-update. Nucleic Acids Rese 41 (Database Issue). D991–D995. 2013.

20 

Li JJ, Wang BQ, Fei Q, Yang Y and Li D: Identification of candidate genes in osteoporosis by integrated microarray analysis. Bone Joint Res. 5:594–601. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e472015. View Article : Google Scholar : PubMed/NCBI

22 

Klipper-Aurbach Y, Wasserman M, Braunspiegel-Weintrob N, Borstein D, Peleg S, Assa S, Karp M, Benjamini Y, Hochberg Y and Laron Z: Mathematical formulae for the prediction of the residual beta cell function during the first two years of disease in children and adolescents with insulin-dependent diabetes mellitus. Med Hypotheses. 45:486–490. 1995. View Article : Google Scholar : PubMed/NCBI

23 

Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, et al: Gene ontology: Tool for the unification of biology. The gene ontology consortium. Nat Genet. 25:25–29. 2000. View Article : Google Scholar : PubMed/NCBI

24 

Kanehisa M and Goto S: KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28:27–30. 2000. View Article : Google Scholar : PubMed/NCBI

25 

Wang F, Wang R, Li Q, Qu X, Hao Y, Yang J, Zhao H, Wang Q, Li G, Zhang F, et al: A transcriptome profile in hepatocellular carcinomas based on integrated analysis of microarray studies. Diagn Pathol. 12:42017. View Article : Google Scholar : PubMed/NCBI

26 

Dufier JL, Rozet JM, Kaplan J and Roche O: From congenital glaucoma to chronic open angle glaucoma in adulthood: A clinical and genetic continuum. Bull Acad Natl Med. 197:133–141. 2013.PubMed/NCBI

27 

Wang X, Huai G, Wang H, Liu Y, Qi P, Shi W, Peng J, Yang H, Deng S and Wang Y: Mutual regulation of the Hippo/Wnt/LPA/TGF-β signaling pathways and their roles in glaucoma (review). Int J Mol Med. 41:1201–1212. 2018.PubMed/NCBI

28 

Alpízar-Alpízar W, Laerum OD, Illemann M, Ramírez JA, Arias A, Malespín-Bendaña W, Ramírez V, Lund LR, Borregaard N and Nielsen BS: Neutrophil gelatinase-associated lipocalin (NGAL/Lcn2) is upregulated in gastric mucosa infected with Helicobacter pylori. Virchows Archiv. 455:225–233. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Khalyfa A, Chlon T, Qiang H, Agarwal N and Cooper NG: Microarray reveals complement components are regulated in the serum-deprived rat retinal ganglion cell line. Mol Vis. 13:293–308. 2007.PubMed/NCBI

30 

Maeda I, Ueda T, Koide R, Inatomi M, Fukado Y, Uchida E, Oguchi K and Yasuhara H: Ocular hypotensive effects of monoamine oxidase-a inhibitors in rabbit. Jpn J Ophthalmol. 32:211–218. 1988.PubMed/NCBI

31 

Ajmone-Cat MA, De Simone R, Nicolini A and Minghetti L: Effects of phosphatidylserine on p38 mitogen activated protein kinase, cyclic AMP responding element binding protein and nuclear factor-kappaB activation in resting and activated microglial cells. J Neurochem. 84:413–416. 2003. View Article : Google Scholar : PubMed/NCBI

32 

Walton M, Woodgate AM, Muravlev A, Xu R, During MJ and Dragunow M: CREB phosphorylation promotes nerve cell survival. J Neurochem. 73:1836–1842. 1999.PubMed/NCBI

33 

Shao Y, Yu Y, Zhou Q, Li C, Yang L and Pei CG: Inhibition of miR-134 protects against hydrogen peroxide-induced apoptosis in retinal ganglion cells. J Mol Neurosci. 56:461–471. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Pankov R and Yamada KM: Fibronectin at a glance. J Cell Sci. 115:3861–3863. 2002. View Article : Google Scholar : PubMed/NCBI

35 

Yi W, Xiao E, Ding R, Luo P and Yang Y: High expression of fibronectin is associated with poor prognosis, cell proliferation and malignancy via the NF-κB/p53-apoptosis signaling pathway in colorectal cancer. Oncol Rep. 36:3145–3153. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Jun HK, Jung YJ and Choi BK: Inflammasome activators induce fibronectin expression and release in macrophages. Cell Microbiol. 19:2017. View Article : Google Scholar : PubMed/NCBI

37 

Jeong JK, Ryu BJ, Choi J, Kim DH, Choi EJ, Park JW, Park JJ and Lee BJ: NELL2 participates in formation of the sexually dimorphic nucleus of the pre-optic area in rats. J Neurochem. 106:1604–1613. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Halder G, Callaerts P and Gehring WJ: Induction of ectopic eyes by targeted expression of the eyeless gene in Drosophila. Science. 267:1788–1792. 1995. View Article : Google Scholar : PubMed/NCBI

39 

Wawersik S and Maas RL: Vertebrate eye development as modeled in Drosophila. Hum Mol Genet. 9:917–925. 2000. View Article : Google Scholar : PubMed/NCBI

40 

Fujimura N, Klimova L, Antosova B, Smolikova J, Machon O and Kozmik Z: Genetic interaction between Pax6 and β-catenin in the developing retinal pigment epithelium. Dev Genes Evol. 225:121–128. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2019
Volume 20 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Feng J and Feng J: Identification of pathogenic genes and transcription factors in glaucoma. Mol Med Rep 20: 216-224, 2019
APA
Feng, J., & Feng, J. (2019). Identification of pathogenic genes and transcription factors in glaucoma. Molecular Medicine Reports, 20, 216-224. https://doi.org/10.3892/mmr.2019.10236
MLA
Feng, J., Xu, J."Identification of pathogenic genes and transcription factors in glaucoma". Molecular Medicine Reports 20.1 (2019): 216-224.
Chicago
Feng, J., Xu, J."Identification of pathogenic genes and transcription factors in glaucoma". Molecular Medicine Reports 20, no. 1 (2019): 216-224. https://doi.org/10.3892/mmr.2019.10236