Molecular Vision 2020; 26:1-13 <http://www.molvis.org/molvis/v26/1>
Received 05 January 2019 | Accepted 31 January 2020 | Published 01 February 2020

Role of the XIST-miR-181a-COL4A1 axis in the development and progression of keratoconus

Rui Tian, Lufei Wang, He Zou, Meijiao Song, Lu Liu, Hui Zhang

Department of Ophthalmology, the Second Hospital of Jilin University, Changchun 130000, Jilin Province, China

Correspondence to: Hui Zhang, Department of Ophthalmology, the Second Hospital of Jilin University, No. 218 Ziqiang Street, Nan Guan District, Changchun 130000, Jilin Province, China; Phone: +86-0430-81136545; FAX: +86-0430-81136545; email: zhanghui_m04@163.com

Abstract

Background: As a disorder occurs in the eyes, keratoconus (KC) is induced by the thinning of the corneal stroma. This study was designed to reveal the key long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and mRNAs involved in the mechanisms of KC.

Methods: Transcriptome RNA-seq data set GSE112155 was acquired from the Gene Expression Omnibus database, which contained 10 KC samples and 10 myopic control samples. Using the edgeR package, the differentially expressed (DE)-mRNAs between KC and control samples were screened. The DE-lncRNAs and DE-miRNAs in this data set were identified using the HUGO Gene Nomenclature Committee (HGNC). Using the pheatmap package, bidirectional hierarchical clustering of the DE-RNAs was conducted. Then, an enrichment analysis of the DE-mRNAs was performed using the DAVID tool. Moreover, a competitive endogenous RNA (ceRNA) regulatory network was built using the Cytoscape software. After KC-associated pathways were searched within the Comparative Toxicogenomics Database, a KC-associated ceRNA regulatory network was constructed.

Results: There were 282 DE-lncRNAs (192 upregulated and 90 downregulated), 40 DE-miRNAs (29 upregulated and 11 downregulated), and 910 DE-mRNAs (554 upregulated and 356 downregulated) between the KC and control samples. A total of 34 functional terms and 9 pathways were enriched for the DE-mRNAs. In addition, 6 mRNAs (including PPARG, HLA-B, COL4A1, and COL4A2), 5 miRNAs (including miR-181a), 9 lncRNAs (including XIST), and the XIST-miR-181a-COL4A1 axis were involved in the KC-associated ceRNA regulatory network.

Conclusions: PPARG, HLA-B, COL4A1, COL4A2, miR-181a, and XIST might be correlated with the development of KC. Further, the XIST-miR-181a-COL4A1 axis might be implicated in the pathogenesis of KC.

Introduction

As a disease of the eye, keratoconus (KC) leads to the uninflammatory thinning of the corneal stroma [1]. KC usually occurs in the transition period from childhood to adulthood, and it is more common in Asian populations [2]. KC may induce the symptoms of double vision, nearsightedness, blurry vision, light sensitivity, and astigmatism [3]. KC patients may need special contact lenses or even corneal transplantation [4,5].

KC is caused by genetic, hormonal, and environmental factors [6], and genetic and environmental factors contribute to the pathogenesis and development of KC. Previous studies have shown that the alteration of inflammatory factors and genetic molecules is associated with KC [7-9]. For instance, Pahuja et al. [8] and Shetty et al. [10] showed that the levels of inflammatory factors such as tumor-necrosis factor (TNF)-α, interleukin (IL)-6, and matrix metalloproteinase 9 (MMP-9) were elevated in the epithelium of KC patients. Long non-coding RNA (lncRNA), microRNAs (miRNAs), and several pathways—including transforming growth factor β (TGF-β), phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt), and Wnt signaling pathways—may function in the development and progression of KC [7,9,11]. The expression of miR-184 in normal cornea samples is higher than that of miR-205, and miR-184 may act in cornea development and corneal diseases [12,13]. Calpastatin plays a role in genetic susceptibility to KC, and the differential modulation of calpain–calpastatin complex may influence the functional defect of the cornea [14]. Downregulated β-actin may be correlated with decreased human antigen R (HuR) in the corneal stroma of KC patients, which may also serve as a risk factor for the occurrence and development of KC [15]. However, the key RNAs involved in the pathogenesis of KC have not been comprehensively identified, and the pathogenesis of KC remains unclear.

In recent years, more and more evidence has shown that the mutual regulation models between lncRNA and miRNA and their downstream target genes are closely related to the occurrence and development of diseases [16,17]. As an important factor in post-transcriptional regulation, miRNA activity can be regulated by lncRNA through sponge adsorption [18]. As competitive endogenous RNA (ceRNA), lncRNA competitively binds to miRNA to regulate the protein level of the coding gene and participates in the regulation of cell biologic behaviors [19]. Therefore, in-depth research into the control mechanism of ceRNA will help us better understand the occurrence and development of diseases.

By analyzing the gene expression profile of KC, the differentially expressed (DE)-RNAs between KC samples and myopic control samples were screened. Following this, a ceRNA regulatory network was built to select the key RNAs affecting the development of KC from those in myopia. This study might help to reveal the ceRNA regulatory mechanisms correlated with the pathogenesis of KC.

Methods

Data preprocessing and differential expression analysis

Transcriptome RNA-seq data set GSE112155 was downloaded from the Gene Expression Omnibus (GEO) database. There were 20 cornea epithelial tissue samples in GSE112155, including 10 KC samples (10 males) and 10 myopic control samples (6 males and 4 females). Female control samples were included in our study to avoid a gender bias, as reported by You [20]. The expression profile data related to read count level was normalized using R package preprocessCore (version 1.40.0, Berkeley, CA) [21].

The lncRNAs, miRNAs, and mRNAs in GSE112155 were annotated and identified in the HUGO Gene Nomenclature Committee (HGNC) database [22], which includes 3,979 lncRNAs, 1,932 miRNAs and 19,197 recognized protein-coding genes. Using R package edgeR (version 3.22.5) [23], the DE-lncRNAs, DE-miRNAs, and DE-mRNAs between the KC and control samples were analyzed. A |log2 fold change (FC)| of >1 and false discovery rate (FDR) of <0.05 were selected as the thresholds of significant differential expression.

Hierarchical clustering analysis and enrichment analysis

For the screened DE-RNAs, bidirectional hierarchical clustering based on a correlation algorithm was performed using the R package pheatmap (version 1.0.8) [24]. In addition, using the DAVID tool (version 6.8, Frederick, MD) [25], the DE-mRNAs were analyzed though the Gene Ontology (GO) enrichment analysis, which included the Biology Process (BP), Molecular Function (MF), and Cellular Component (CC) categories [26], and through the Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis [27]. A p value of <0.05 was taken as the threshold of enrichment significance.

Regulatory network analysis of ceRNA

As a kind of ceRNA within miRNA, lncRNA is involved in the expression regulation of target genes and plays an important role in the occurrence and development of diseases [28]. The lncRNA–miRNA regulatory pairs in the miRecode (version 11, Gothenburg, Sweden) [29] and starBase (version 2.0, Guangzhou, China) [30] databases were merged, and the regulatory pairs between DE-lncRNAs and DE-miRNAs were then screened from them. Using the Cytoscape software (version 3.6.1, Bethesda, MD) [31], the lncRNA–miRNA regulatory network was visualized.

Using the starBase database [30], the target genes were predicted for the DE-miRNAs involved in the identified lncRNA–miRNA pairs. The starBase database integrates target prediction information from the targetScan, picTar, RNA22, PITA, and miRanda databases [30]. In this study, miRNA-target regulatory pairs included in at least one of the five databases were selected. Subsequently, the DE-mRNAs were correlated with the predicted target genes, and only the miRNA-target regulatory pairs involved the DE-miRNAs and DE-mRNAs with opposite expression change directions were retained. Moreover, the miRNA–mRNA regulatory network was built using the Cytoscape software [31].

Along with the identified lncRNA–miRNA and miRNA–mRNA regulatory pairs, the ceRNA regulatory network was constructed using the Cytoscape software [31]. In addition, DAVID tool [25] was used to enrich the KEGG pathways for the mRNAs implicated in the ceRNA regulatory network, with a p value of <0.05 as the significance threshold.

Construction of KC-associated ceRNA regulatory network

Using “keratoconus” as the keyword, the KEGG pathways directly correlated with KC were searched in the Comparative Toxicogenomics Database (Raleigh, NC) [32]. To obtain the mRNAs participating in the KC-associated pathways, the searched pathways were compared with the pathways enriched for the mRNAs implicated in the ceRNA regulatory network. Finally, the regulatory pairs involving the mRNAs enriched in the KC-associated pathways were extracted from the ceRNA regulatory network to construct the KC-associated ceRNA regulatory network.

Results

Data preprocessing and differential expression analysis

A total of 22,281 RNAs were detected in transcriptome RNA-seq data set GSE112155. Based on the HGNC database, 3,261 lncRNAs, 789 miRNAs, and 18,231 mRNAs were identified. The expression distribution curves of the identified lncRNAs, miRNAs, and mRNAs are shown in Figure 1A. There were 282 DE-lncRNAs (192 upregulated and 90 downregulated), 40 DE-miRNAs (29 upregulated and 11 downregulated), and 910 DE-mRNAs (554 upregulated and 356 downregulated) between the KC and control samples. A scatter diagram of the screened DE-RNAs is presented in Figure 1B.

Hierarchical clustering analysis and enrichment analysis

A clustering heatmap showed that the expression values of the screened DE-RNAs could separate different types of samples (Figure 1C,D). The result suggested that the DE-RNAs had expression characteristics in the samples. Enrichment analysis showed that 13 GO_BP terms (such as ion transport), 10 GO_CC terms (such as extracellular region), 11 GO_MF terms (such as channel activity), and 9 KEGG pathways (such as neuroactive ligand-receptor interaction) were enriched for the DE-mRNAs (Table 1).

Analysis of the ceRNA regulatory network

After 66 eligible lncRNA–miRNA pairs were obtained, the lncRNA–miRNA regulatory network (involving 31 lncRNAs and 8 miRNAs) was built (Figure 2). Afterwards, 116 miRNA–mRNA regulatory pairs were acquired to construct the miRNA–mRNA regulatory network (involving 7 miRNAs and 92 mRNAs; Figure 3).

Then, lncRNA–miRNA–mRNA regulatory pairs were then identified, and the ceRNA regulatory network was constructed (Figure 4). In the ceRNA regulatory network, there were 131 nodes (including 32 lncRNAs, 7 miRNAs, and 92 mRNAs) and 182 edges (including 66 lncRNA–miRNA pairs and 116 miRNA–mRNA pairs). Additionally, 8 KEGG pathways (such as focal adhesion) were enriched for the mRNAs implicated in the ceRNA regulatory network (Table 2).

Construction of KC-associated ceRNA regulatory network

Only 12 KEGG pathways directly correlated with KC were found in the Comparative Toxicogenomics Database. After comparing the searched pathways with the pathways enriched for the mRNAs implicated in the ceRNA regulatory network, two overlapping pathways (type I diabetes mellitus, and pathways in cancer) were obtained as KC-associated pathways. Subsequently, the KC-associated ceRNA regulatory network was built (Figure 5). Meanwhile, the RNAs correlated with KC were obtained, including 6 mRNAs (peroxisome proliferator-activated receptor gamma [PPARG]; human leukocyte antigen-B [HLA-B]; runt-related transcription factor 1 [RUNX1T1]; carboxypeptidase E [CPE]; collagen, type IV, alpha 1 [COL4A1]; and collagen, type IV, alpha 2 [COL4A2]), 5 miRNAs (miR-301a, miR-181a, miR-222, miR-98, and miR-128), and 9 lncRNAs (X-inactive specific transcript [XIST]; ST7-AS2; LINC00309; LINC00299; LINC00261; LINC00276; LINC00355; rhabdomyosarcoma 2-associated transcript [RMST]; LINC00520; and prostate cancer antigen 3 [PCA3]; Figure 6). The XIST-miR-181a-COL4A1 axis was particularly involved in the KC-associated ceRNA regulatory network.

Discussion

Previous studies have suggested that sex and ethnicity may affect morbidity, gene expression, and episode age in KC [20,33,34]. Gender consistency in gene expression, however, will generally explain the pathogenesis of KC. In this study, a total of 282 DE-lncRNAs (192 upregulated and 90 downregulated), 40 DE-miRNAs (29 upregulated and 11 downregulated), and 910 DE-mRNAs (554 upregulated and 356 downregulated) were screened for KC, relative to control samples with no gender bias. For the DE-mRNAs, 13 GO_BP terms, 10 GO_CC terms, 11 GO_MF terms, and 9 KEGG pathways were enriched. In the KC-associated ceRNA regulatory network, there were 6 mRNAs (including PPARG, HLA-B, COL4A1, and COL4A2), 5 miRNAs (including miR-181a), and 9 lncRNAs (including XIST). The number of DE-mRNAs (910) between the KC and control samples was higher than 13 and lower than 1422 without and with gender bias, respectively, in You [20]. The number of DE-mRNAs (910) with strict criteria (edgeR, |log2FC| >1, and FDR <0.05) suggested that the result of the gene expression profile was credible.

The elevated inflammatory factors, including IL-6, TNF-α, and IL-1 receptors, have been identified in KC epithelium, despite KC having been defined as a non-inflammatory condition [8,35-37]. Multiple studies have shown conflicting results about inflammation in KC. Among the reported inflammatory factors in KC, IL-6 and IL-6 receptor (IL-6R) play roles in glaucomatous optic nerve and retina damage, and their abnormal single nucleotide polymorphisms (SNPs) are involved in the development and progression of primary open-angle glaucoma (POAG) [38]. In addition, IL-6 assumes important roles in herpes simplex virus (HSV) type I infection-induced corneal nerve degeneration [39] and in ocular inflammation and angiogenesis in the cornea [40]. The roles of PPARG in inflammation have been widely reported [41-43]. PPARG regulates the redox balance in macrophages [41]. A previous study explored the functions of PPARG agonist rosiglitazone on retinoblastoma cells and found that rosiglitazone plays an antitumor role via the suppression of cell growth, metastasis, and invasion and via the promotion of cell apoptosis [44]. HLA-A26, HLA-B40, and HLA-DR9 are frequently found in older Japanese populations and may be related to KC in young people [45]. HLA-G contributes to establishing immune tolerance in allograft, which may also help to maintain the immune-privileged status of the cornea [46]. Our present study, however, identified the downregulation of IL-6, PPARG and HLA in epithelium from patients with KC. We speculated the participation of IL-6, PPARG and HLA in KC pathogenesis. However, there were conflicting results between our study and others reporting the elevation of IL-6 in KC epithelium compared with controls [8,35-37].

In addition to conflicting inflammatory conditions in KC, histopathological changes in collagen decomposition or fibrosis are associated with KC induction [47,48]. Transcription factor 8 (TCF8) plays a role in about half of posterior polymorphous corneal dystrophy (PPCD) cases, and its target, COL4A3, is critical in both Alport syndrome and PPCD [49,50]. Enhancement during collagen decomposition is responsible for the damage of pathological tissues in KC corneas, which is still retained in initial cultures of KC fibroblasts [51]. In KC corneas, type I, III, and V collagens have no difference in distribution, while the distribution of type IV collagen is disruptive and excrescent in the corneal basement membrane [47,52]. KC corneas have decreased collagen protein levels, and collagen type IV functions as a candidate gene in the development of KC [53]. Our present study suggested that COL4A1 and COL4A2 were dysregulated in the KC epithelium relative to controls, suggesting the crucial role of collagen decomposition in the progression of KC.

In addition to the DE-mRNAs, we also identified the DE-miRNAs and lncRNAs in KC samples compared with controls. The miR-181a, miR-21, and Smad signaling coordinately regulate the expression of TGF-β-induced gene (TGFBI) protein (TGFBIp) in corneal fibroblasts, and their pharmacologic modulation may be applied to treat TGFBI-correlated corneal dystrophy [54]. The overexpression of miR-181b can be induced by hypoxia, which further promotes the angiogenesis of retinoblastoma cells by mediating GATA-binding protein 6 (GATA6) and programmed cell death 10 (PDCD10) [55]. Upregulated lncRNA XIST is positively related to an advanced stage and late differentiation state of retinoblastoma, and XIST may accelerate retinoblastoma progression via regulating the miR-124/signal transducer and activator of transcription 3 (STAT3) axis [56]. The XIST-miR-181a-COL4A1 axis was involved in the KC-associated ceRNA regulatory network, indicating that XIST and miR-181a might be correlated with the pathogenesis of KC through the XIST-miR-181a-COL4A1 axis.

In conclusion, 282 DE-lncRNAs, 40 DE-miRNAs, and 910 DE-mRNAs were identified between the KC and control samples. These DE-RNAs were identified without gender bias. Further, PPARG, HLA-B, COL4A1, COL4A2, miR-181a, and XIST might be involved in the development and progression of KC in both females and males. Moreover, the XIST-miR-181a-COL4A1 axis might function in the mechanisms of KC. However, the specific roles of these RNAs in KC should be further explored and supported by experiments.

Acknowledgments

This study is by the Norman Bethune Program of Jilin University (2015327); the Postdoctoral research project of Jilin province (801171060413); the Health service capacity improvement project (2017) of Jilin province (3D517EC63429); the youth program of National natural science foundation (2018) of China (81802998); 2019 basic Research Natural Science Foundation (20190201150JC). Conception and design of the research: R. Tian and H. Zhang. Acquisition, analysis and interpretation of data: R. Tian, H. Zou, L.F. Wang and L. Liu. Drafting the manuscript: R. Tian. Revision of manuscript for important intellectual content: H. Zhang, L. Liu and M.J. Song. All authors have read and approved the manuscript. Compliance with Ethical Standards: This article does not contain any studies with human participants or animals performed by any of the authors, the authors have no ethical conflicts to disclose.

References

  1. Sherwin T, Brookes NH. Morphological changes in keratoconus: pathology or pathogenesis. Clin Experiment Ophthalmol. 2004; 32:211-7. [PMID: 15068441]
  2. Léoni-Mesplié S, Mortemousque B, Mesplié N, Touboul D, Praud D, Malet F, Colin J. Epidemiological aspects of keratoconus in children. J Fr Ophtalmol. 2012; 35:776-85. [PMID: 22981526]
  3. Naderan M, Shoar S, Rezagholizadeh F, Zolfaghari M. Characteristics and associations of keratoconus patients. Cont Lens Anterior Eye. 2015; 182:199-205. [PMID: 25707930]
  4. Lehtosalo J, Uusitalo RJ, Mianowizc J. Epikeratophakia for treatment of keratoconus. Acta Ophthalmol Suppl. 1987; 65S182:74-7. [PMID: 2837066]
  5. Espandar L, Meyer J. Keratoconus: overview and update on treatment. Middle East Afr J Ophthalmol. 2010; 17:15-20. [PMID: 20543932]
  6. Grünauer-kloevekorn C, Duncker GI. Keratoconus: epidemiology, risk factors and diagnosis. Klin Monatsbl Augenheilkd. 2006; 223:493-502. [PMID: 16804819]
  7. Khaled ML, Bykhovskaya Y, Yablonski SE, Li H, Drewry MD, Aboobakar IF, Estes A, Gao X, Stamer WD, Xu H. Differential Expression of Coding and Long Noncoding RNAs in Keratoconus-Affected Corneas. Invest Ophthalmol Vis Sci. 2018; 59:2717-28. [PMID: 29860458]
  8. Pahuja N, Kumar NR, Shroff R, Shetty R, Nuijts RM, Ghosh A, Sinha-Roy A, Chaurasia SS, Mohan RR. Differential Molecular Expression of Extracellular Matrix and Inflammatory Genes at the Corneal Cone Apex Drives Focal Weakening in Keratoconus. Invest Ophthalmol Vis Sci. 2016; 57:5372-82. [PMID: 27732724]
  9. Ionescu C, Corbu CG, Tanase C, Jonescu-Cuypers C, Nicula C, Dascalescu D, Cristea M, Voinea LM. Inflammatory Biomarkers Profile as Microenvironmental Expression in Keratoconus. Dis Markers. 2016; 2016:1-8. [PMID: 27563164]
  10. Shetty R, Anuprita G, Lim RR, Murali S, Krina M, Reshma AR, Ashwini R, Sriharsha N, Nuijts RMMA, Roger B. Elevated expression of matrix metalloproteinase-9 and inflammatory cytokines in keratoconus patients is inhibited by cyclosporine A. Invest Ophthalmol Vis Sci. 2015; 56:738-50. [PMID: 25648341]
  11. Karolak JA, Polakowski P, Szaflik J, Szaflik JP, Gajecka M. Molecular Screening of Keratoconus Susceptibility Sequence Variants in VSX1, TGFBI, DOCK9, STK24, and IPO5 Genes in Polish Patients and Novel TGFBI Variant Identification. Ophthalmic Paediatr Genet. 2016; 37:37-43. [PMID: 24940934]
  12. Abu-Amero KK, Helwa I, Almuammar A, Strickland S, Hauser MA, Allingham RR, Liu Y. Screening of the Seed Region of MIR184 in Keratoconus Patients from Saudi Arabia. BioMed Res Int. 2015; 2015:604508 [PMID: 26380287]
  13. Hughes AE, Bradley DT, Campbell M, Lechner J, Dash DP, Simpson DA, Willoughby CE. Mutation altering the miR-184 seed region causes familial keratoconus with cataract. Am J Hum Genet. 2011; 89:628-33. [PMID: 21996275]
  14. Li X, Bykhovskaya Y, Tang YG, Picornell Y, Haritunians T, Aldave AJ, Szczotka-Flynn L, Iyengar SK, Rotter JI, Taylor KD. An association between the calpastatin (CAST) gene and keratoconus. Cornea. 2013; 32:696-701. [PMID: 23449483]
  15. Joseph R, Srivastava OP, Pfister RR. Downregulation of β-actin gene and human antigen R in human keratoconus. Invest Ophthalmol Vis Sci. 2012; 53:4032 [PMID: 22562506]
  16. Mercer TR, Dinger ME, Mattick JS. Long non-coding RNAs: insights into functions. Nat Rev Genet. 2009; 10:155-9. [PMID: 19188922]
  17. Wapinski O, Chang HY. Long noncoding RNAs and human disease. Trends Cell Biol. 2011; 21:354-61. [PMID: 21550244]
  18. Paraskevopoulou MD, Hatzigeorgiou AG. Analyzing MiRNA–LncRNA Interactions. Methods Mol Biol. 2016; 1402:271-86. [PMID: 26721498]
  19. Ergun S, Oztuzcu S. Oncocers: ceRNA-mediated cross-talk by sponging miRNAs in oncogenic pathways. Tumour Biol. 2015; 36:1-8. [PMID: 25809705]
  20. You J, Corley SM, Wen L, Hodge C, Höllhumer R, Madigan MC, Wilkins MR, Sutton G. RNA-Seq analysis and comparison of corneal epithelium in keratoconus and myopia patients. Sci Rep. 2018; 8:389 [PMID: 29321650]
  21. Null RCTR, Team R, Null RCT, Core Writing T, Null R, Team R. Null RDCT, Core R, Team R, Team RDC. R: A language and environment for statistical computing. Computing. 2009; 1:12-21.
  22. Povey S, Lovering R, Bruford E, Wright M, Lush M, Wain H. The HUGO Gene Nomenclature Committee (HGNC). Hum Genet. 2001; 109:678-80. [PMID: 11810281]
  23. Nikolayeva O, Robinson MD. edgeR for differential RNA-seq and ChIP-seq analysis: an application to stem cell biology. Methods Mol Biol. 2014; 1150:45-79. [PMID: 24743990]
  24. Kolde R. Pretty Heatmaps. R package. 2015. https://cran.r-project.org/web/packages/pheatmap/index.html.
  25. Glynn Dennis J, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC, Lempicki RA. DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol. 2003; 4:3 [PMID: 12734009]
  26. Young MD, Wakefield MJ, Smyth GK, Oshlack A. Gene ontology analysis for RNA-seq: accounting for selection bias. Genome Biol. 2010; 11:R14
  27. Zhang T, Jiang M, Chen L, Niu B, Cai Y. Prediction of gene phenotypes based on GO and KEGG pathway enrichment scores. BioMed Res Int. 2013; xxx:870795
  28. Yang C, Wu D, Gao L, Liu X, Jin Y, Wang D, Wang T, Li X. Competing endogenous RNA networks in human cancer: hypothesis, validation, and perspectives. Oncotarget. 2016; 7:13479-90. [PMID: 26872371]
  29. Jeggari A, Marks DS, Larsson E. miRcode: a map of putative microRNA target sites in the long non-coding transcriptome. Bioinformatics. 2012; 28:2062-3. [PMID: 22718787]
  30. Li JH, Liu S, Zhou H, Qu LH, Yang JH. starBase v2.0: decoding miRNA-ceRNA, miRNA-ncRNA and protein-RNA interaction networks from large-scale CLIP-Seq data. Nucleic Acids Res. 2014; 42:D92 [PMID: 24297251]
  31. Kohl M, Wiese S, Warscheid B. Cytoscape: software for visualization and analysis of biological networks. Methods Mol Biol. 2011; 696:291-303. [PMID: 21063955]
  32. Peter DA, Grondin MC, Robin J, Lay JM, Kelley LH, Cynthia SR, Daniela S, King BL, Rosenstein MC, Wiegers TC. The Comparative Toxicogenomics Database: update 2013. Nucleic Acids Res. 2013; 41:1104-14. [PMID: 23222133]
  33. Magalhaes OA, Marafon SB, Ferreira RC. Gender differences in keratoconus keratoplasty: a 25-year study in Southern Brazil and global perspective. Int Ophthalmol. 2018; 38:1627-33. [PMID: 28831627]
  34. Suzuki T, Richards SM, Liu S, Jensen RV, Sullivan DA. Influence of sex on gene expression in human corneal epithelial cells. Mol Vis. 2009; 15:2554-69. [PMID: 20011627]
  35. Krachmer JH, Feder RS, Belin MW. Keratoconus and related noninflammatory corneal thinning disorders. Surv Ophthalmol. 1984; 28:293-322. [PMID: 6230745]
  36. Galvis V, Sherwin T, Tello A, Merayo J, Barrera R, Acera A. Keratoconus: an inflammatory disorder? Eye (Lond). 2015; 29:843-59. [PMID: 25931166]
  37. Rohit S, Anuprita G, Lim RR, Murali S, Krina M, Reshma AR, Ashwini R, Sriharsha N, Nuijts RMMA, Roger B. Elevated expression of matrix metalloproteinase-9 and inflammatory cytokines in keratoconus patients is inhibited by cyclosporine A. Invest Ophthalmol Vis Sci. 2015; 56:738-50. [PMID: 25648341]
  38. Zhou G, Liu B, Yang YF. Single nucleotide polymorphism analysis of multi-loci and -genes in primary open-angle glaucoma with pathological myopia. Int J Ophthalmol. 2007; 3:36-42
  39. Chucair-Elliott AJ, Jinkins J, Carr MM, Carr DJJ. IL-6 Contributes to Corneal Nerve Degeneration after Herpes Simplex Virus Type I Infection. Am J Pathol. 2016; 186:2665-78. [PMID: 27497323]
  40. Ghasemi H. Roles of IL-6 in Ocular Inflammation: A Review. Ocul Immunol Inflamm. 2018; 26:37-50. [PMID: 28146368]
  41. Shua Y. yang N, Bei Y, Shen P. PPARγ phosphorylation at Ser186 regulates metabolic inflammation via modulating redox balance in adipocyte tissue macrophages. Free Radic Biol Med. 2018; 120:s156
  42. Sardella C, Winkler C, Quignodon L, Hardman JA, Toffoli B, Gmp GA, Hundt JE, Michalik L, Vinson CR, Paus R. Delayed Hair Follicle Morphogenesis and Hair Follicle Dystrophy in a Lipoatrophy Mouse Model of Pparg Total Deletion. J Invest Dermatol. 2017; 138:xxx
  43. Nobs SP, Natali S, Pohlmeier L, Okreglicka K, Schneider C, Kurrer M, Sallusto F, Kopf M. PPARγ in dendritic cells and T cells drives pathogenic type-2 effector responses in lung inflammation. J Exp Med. 2017; 214:3015-35. [PMID: 28798029]
  44. Cao X, Lin H, Li Y. Effects of PPARγ agonistrosiglitazone on human retinoblastoma cell in vitro and in vivo. Int J Clin Exp Pathol. 2015; 8:12549-56. [PMID: 26722443]
  45. Adachi W, Mitsuishi Y, Terai K, Nakayama C, Hyakutake Y, Yokoyama J, Mochida C, Kinoshita S. The association of HLA with young-onset keratoconus in Japan. Am J Ophthalmol. 2002; 133:557-9. [PMID: 11931792]
  46. Le DM, Moreau PP, Legeais JM, Carosella ED. Expression of HLA-G in human cornea, an immune-privileged tissue. Hum Immunol. 2003; 64:1039-44. [PMID: 14602233]
  47. Kenney MC, Brown DJ. The Cascade Hypothesis of Keratoconus. Cont Lens Anterior Eye. 2003; 26:139-46. [PMID: 16303509]
  48. Smolek MK, Beekhuis WH. Collagen fibril orientation in the human corneal stroma and its implications in keratoconus. Invest Ophthalmol Vis Sci. 1997; 38:1289 [PMID: 9191590]
  49. Krafchak Charles M, Pawar H, Moroi Sayoko E, Sugar A, Lichter Paul R, Mackey David A, Mian S, Nairus T, Elner V, Schteingart Miriam T. Mutations in TCF8 Cause Posterior Polymorphous Corneal Dystrophy and Ectopic Expression of COL4A3 by Corneal Endothelial Cells. Am J Hum Genet. 2005; 77:694-708. [PMID: 16252232]
  50. Kokolakis NS, Maria G, Chatziralli IP, Chryssanthi K, Zisis G, Peponis VG, Droutsas KD, Christos K, Nicholas A, Dimitrios M. Polymorphism analysis of COL4A3 and COL4A4 genes in Greek patients with keratoconus. Ophthalmic Genet. 2015; 36:213-7. [PMID: 24099280]
  51. Ihalainen A, Salo T, Forsius H, Peltonen L. Increase in type I and type IV collagenolytic activity in primary cultures of keratoconus cornea. Eur J Clin Invest. 2010; 16:78-84. [PMID: 3009200]
  52. Nakayasu K, Tanaka M, Konomi H, Hayashi T. Distribution of types I, II, III, IV and V collagen in normal and keratoconus corneas. Ophthalmic Res. 2009; 18:1-10. [PMID: 3513078]
  53. Saravani R, Hasanian-Langroudi F, Validad MH, Yari D, Bahari G, Faramarzi M, Khateri M, Bahadoram S. Evaluation of Possible Relationship Between COL4A4 Gene Polymorphisms and Risk of Keratoconus. Cornea. 2015; 34:318-22. [PMID: 25651396]
  54. Choi SI, Jin JY, Maeng YS, Kim TI, Kim EK. TGF-β regulates TGFBIp expression in corneal fibroblasts via miR-21, miR-181a, and Smad signaling. Biochem Biophys Res Commun. 2016; 472:150-5. [PMID: 26915797]
  55. Xu X, Ge S, Jia R, Zhou Y, Song X, Zhang H, Fan X. Hypoxia-induced miR-181b enhances angiogenesis of retinoblastoma cells by targeting PDCD10 and GATA6. Oncol Rep. 2015; 33:2789-96. [PMID: 25872572]
  56. Hu C, Liu S, Han M, Wang Y, Xu C. Knockdown of lncRNA XIST inhibits retinoblastoma progression by modulating the miR-124/STAT3 axis. Biomed Pharmacother. 2018; 107:547-54.