You are on page 1of 12

Review Article

Ophthalmic Res 2022;65:481–492 Received: August 9, 2021


Accepted: February 18, 2022
DOI: 10.1159/000523878 Published online: April 11, 2022

Biomarkers in the Occurrence


and Development of Pterygium
Siying He Zhaoxia Wu
Clinical Lab, Jinhua Hospital of Zhejiang University, Jinhua, China

Downloaded from http://karger.com/ore/article-pdf/65/5/481/4021749/000523878.pdf by guest on 05 November 2023


Keywords transition (EMT) [3]. Pterygium is usually located on the
Pterygium · Biomarkers · Genetics · Epigenetics · Proteomics nasal side, affecting patients’ appearance, generating a
foreign body sensation, and even causing blindness [4].
Pterygium is widely considered a chronic inflamma-
Abstract tion and is associated with prolonged exposure to out-
Pterygium is a kind of common conjunctival degeneration. door ultraviolet (UV) light and dust [5]. Records of pte-
The pathogenesis of pterygium is complex, and various bio- rygium date back to 770 BC, and the disease is derived
markers provide new targets for treatment and prognosis. from the syndrome of wind-heat and damp-heat [6]. Pte-
Currently, the most common treatment for pterygium is sur- rygium is widely considered to be caused by joint genetic
gical excision, but it is invasive risk and has a high recurrence and environmental factors [7], such as human papilloma-
rate. Since the development of sequencing, gene chip tech- virus [8], allergens [9], and cholesterol metabolism [10].
nology, and proteomics technologies has been rapid, re- A brief history of hypotheses on the etiology of pterygium
search on the internal mechanism of disease has been facili- is presented in Figure 1.
tated. This review focuses on recent advances in the discov- Currently, the most common treatment for pterygium
ery of biomarkers from the fields of genetics, proteomics, and is surgical excision with conjunctival autografts [11].
epigenetics and their likely functional mechanisms and clini- However, postoperative use of adjuvant agents, such as
cal applications in pterygium. © 2022 The Author(s). mitomycin C and 5-flurouracil, can effectively reduce the
Published by S. Karger AG, Basel recurrence rate [12]. Nonetheless, there are still many dif-
ficulties associated with therapy, such as operative risks,
postoperative scarring, and a high postoperative recur-
Introduction rence rate [13, 14]. These problems have led many clini-
cians to devote themselves to the study of the pathogen-
Pterygium is a common ocular disease. Epidemiologi- esis and treatment of both primary and recurrent pteryg-
cal studies worldwide indicate that the prevalence rates of ium. Therefore, the identification of biomarkers for the
pterygium range from 0.3% to 37.46% [1, 2]. Pterygium occurrence and development of primary pterygium is im-
is characterized by hyperplastic, centripetally directed portant to deepen the understanding of pterygium patho-
growth of altered limbal epithelial cells accompanied by genesis and provide new biomarkers for therapeutics and
Bowman’s layer dissolution and epithelial-mesenchymal recurrence prevention.

Karger@karger.com © 2022 The Author(s). Correspondence to:


www.karger.com/ore Published by S. Karger AG, Basel Siying He, hesiying @ whu.edu.cn
This is an Open Access article licensed under the Creative Commons
Attribution-NonCommercial-4.0 International License (CC BY-NC)
(http://www.karger.com/Services/OpenAccessLicense), applicable to
the online version of the article only. Usage and distribution for com-
mercial purposes requires written permission.
Syndrome of Matrix
wind-heat and Inflammatory metalloproteinase
damp-heat response Apoptosis (MMPs)

Pax6
Vascular Oncogenic
EMT proteins: p53
dysplasia

BC770 1893 1954 1956 1966 1971 1975 1982 1984 1994 1997 1999 2000 2004 2008

Pterygium

Ultraviolet Oxidative
light damage Limbal
stem cells

Downloaded from http://karger.com/ore/article-pdf/65/5/481/4021749/000523878.pdf by guest on 05 November 2023


HPV
Hereditary Immunologic Cholesterol
mechanism metabolism

Fig. 1. The history of hypotheses for the etiology of pterygium. EMT, epithelial-mesenchymal transition; HPV,
human papillomavirus; Pax6, paired box protein 6.

Genetic Biomarkers of Pterygium p53 function offers hope for more effective pterygium
treatment.
Both gene mutations and single nucleotide polymor- Unlike p53, Kirsten rat sarcoma viral oncogene homo-
phisms involve changes in gene structure or expression logue (K-ras) is one of the most frequently mutated on-
levels. The use of next-generation sequencing and DNA cogenes in cancer. Mutated K-ras leads to continual K-ras
microarrays has provided new strategies for research on activation, and disordered signal transduction results in
the mechanism of pterygium. cell proliferation. The K-ras mutation rate at codon 12 is
The first genetic biomarker of pterygium, heparin significantly higher in patients with recurrent pterygium
binding-epidermal growth factor (HB-EGF), was identi- and young patients [20], while the K-ras codon 61 muta-
fied in 2003; its mRNA was elevated 6.8-fold at 6 h after tion frequency is significantly higher in primary and bi-
irradiation [15]. HB-EGF is a potent mitogen that is sig- lateral pterygium tissues [21]. However, the number of
nificantly induced by UV radiation in pterygium-derived specimens in both studies was too small to obtain a con-
epithelial cells, suggesting that HB-EGF is positively cor- clusion with certainty, but it may be speculated that K-ras
related with the progression of pterygium and that UV is associated with a recurrent risk of pterygium.
irradiation might participate in inducing pterygium Differentially expressed genes were first identified
pathogenesis. from the DNA microarray and further characterized by
Early research has shown that both the expression and real-time polymerase chain reaction, western blot, and
function of p53 are dysregulated in pterygium [16, 17]. immunohistochemistry. Thus, neutrophil gelatinase-as-
Mutations within exons 4–8 in p53 are detected in the sociated lipocalin and matrix metalloproteinase 9
pterygium epithelium, but only 15.7% (8 of 51) of pte- (MMP9) were upregulated, while insulin-like growth fac-
rygium have p53 mutations [17]. Mutation in the p53 tor-binding protein 3 was downregulated in pterygium
gene leads to increased protein stability, allowing more [22, 23]. Neutrophil gelatinase-associated lipocalin and
pronounced immunohistochemical detection [18]. UV MMP9 account for extracellular matrix (ECM) accumu-
light activates survivin to suppress the biological function lation and a chronic inflammatory reaction, and low ex-
of p53, leading to abnormal cell cycle, apoptosis, and pression of insulin-like growth factor-binding protein 3
DNA repair [19]; therefore, the possibility of restoring leads to uncontrolled cell proliferation.

482 Ophthalmic Res 2022;65:481–492 He/Wu


DOI: 10.1159/000523878
Table 1. The changes of DNA methylation in pterygium ium and control groups [27]. Therefore, the role of the
MMP9 SNP in determining personal susceptibility to pte-
Gene DNA methylation Phenotypes affected Initiator
rygium is still open to debate.
p16 ↑ Proliferation DNMT3b With the development of science and technology,
E-cadherin ↑ EMT DNMT3b weighted gene coexpression network analysis has been
TGM2 ↑ ECM remodeling – used to provide novel insights into the potential regula-
MMP2 ↓ ECM remodeling – tory mechanisms of pterygium. Among the differentially
CD24 ↓ ECM remodeling
MDM2 ↓ Proliferation DNMT3a
expressed genes, 200 upregulated genes in pterygium
LATS1/LATS2 ↑ Apoptosis – were mainly involved in two biological functions: ECM
and cell adhesion or migration. The 139 downregulated
CD24, cluster of differentiation 24; DNMT3a/3b, DNA methyl- genes were enriched in inflammation pathways [28].
transferase 3a/3b; ECM, extracellular matrix; EMT, epithelial-mesen- These differentially expressed genes are mainly involved
chymal transition; LATS1/2, large tumor suppressor kinase 1/2;
MDM2, mouse double minute 2; MMP2, matrix metalloproteinase 2;
in ECM-receptor interactions, the phosphoinositide 3-ki-
TGM2, transglutaminase 2. nase (PI3K)-protein kinase B (Akt) signaling pathway,
and an endoplasmic reticulum-related pathway.

Downloaded from http://karger.com/ore/article-pdf/65/5/481/4021749/000523878.pdf by guest on 05 November 2023


There is a restriction fragment length polymorphism Epigenetic Biomarkers in Pterygium
in intron 16 of angiotensin-converting enzyme (ACE)
based on its presence (insertion I) or absence (deletion D) Epigenetics, including DNA methylation, histone
that leads to three genotypes: DD homozygote, II homo- modification, nucleosome localization, and noncoding
zygote, and ID heterozygote. Detection of the ACE poly- RNA editing, is based on the genetic changes in gene ex-
morphism genotype has been performed in Sardinian pa- pression but without changes in nucleotide sequences.
tients, and the D and DD genotypes are associated with a Abnormal changes have been identified in DNA meth-
higher risk of developing pterygium, while the I allele ylation and microRNA (miRNA) expression in pterygi-
plays a protective role in pterygium occurrence [24]; the um.
DD genotype activates more circulating ACE to promote
vasoconstriction, cell proliferation, and inflammatory re- DNA Methylation Biomarkers of Pterygium
sponses. DNA methylation refers to the covalent binding of a
A missense variant in cysteine-rich transmembrane methyl group at the 5′ C site of the cytosine in genomic
bone morphogenetic protein regulator 1 (CRIM1) CpG dinucleotides due to the action of DNA methyl-
(NM_016441.2: c.1235A>C (H412P)) has been identified transferases. In general, CpG dinucleotides in “useless”
in a family affected by pterygium [25]. CRIM1 is a widely sequences are relatively rare and always in a methylated
expressed gene in the eye and plays a significant role in state, while most coding genes are always unmethylated
cell migration, differentiation, and angiogenesis. Mutat- [29]. Changes in DNA methylation in pterygium report-
ed CRIM1 loses its activation in extracellular single-reg- ed to date are shown in Table 1.
ulated kinase phosphorylation, leading to excessive cell The protein expression of p16 is downregulated in pte-
proliferation and inhibition of apoptosis. Although the rygium, which is due to the high methylation of the p16
CRIM1 mutation was discovered in an Irish family, larger promoter in pterygium caused by DNA methyltransfer-
sample sizes are needed to verify the hypothesis that ase 3b [30]. As a tumor suppressor, p16 participates in the
CRIM1 may serve as a predictive biomarker for evaluat- regulation of the cell cycle. Staining results have shown
ing the pterygium recurrence rate. that p16 is limited to the nuclei of the epithelial layer, in-
The SNP (rs1060043) in the SLC1A5 gene was signifi- dicating that high methylation of the p16 promoter par-
cantly associated with conjunctival UV autofluorescence ticipates in initiating the development of pterygium.
[26]. The SLC1 gene family has been widely detected in In 2011, researchers detected the methylation level of
the human cornea, so the role of SLC1A5 in pterygium 29 genes encoding ECM proteins in pterygium [31].
deserves further study. Transglutaminase 2 was highly methylated in pterygium,
A recent study showed that allelic frequency analysis while matrix metalloproteinase 2 (MMP2) and cluster of
of MMP9 rs3918242 showed no significant difference in differentiation 24 (CD24) had low methylation levels. De-
the distribution of allelic frequencies between the pteryg- creased transglutaminase 2 attenuated cell adhesion and

Review Based on Genetic, Epigenetic, and Ophthalmic Res 2022;65:481–492 483


Proteomic Biomarkers in Pterygium DOI: 10.1159/000523878
facilitated the migration of pterygium tissue, while MMP2 RNAs (circRNA), which do not encode proteins but reg-
and CD24 participated in ECM remodeling and EMT ulate translation. Noncoding RNAs have also been exten-
promotion. Furthermore, CD24 has been verified to play sively studied in pterygium. The abnormal expression of
a role in neovascularization [32]. However, E-cadherin, a noncoding RNAs in pterygium is shown in Table 2.
surface marker of epithelial cells, has a hypermethylated
promoter region, suggesting the presence of EMT in pte- miRNA Biomarkers in Pterygium
rygium [33]. miRNAs are a class of small noncoding RNAs that are
Previous studies have demonstrated that p53 is differ- approximately 22 nt in length and are known to be pow-
entially expressed in pterygium [16, 17], and mouse dou- erful regulators of gene expression. miRNAs bind to the
ble minute 2 (MDM2) acts as an E3 ubiquitin protein li- 3′ untranslated region of target genes and induce gene
gase in the degradation of p53 [34]. Compared to that of silencing or degradation.
control conjunctiva, the methylation rate of the MDM2 Some miRNAs Are Associated with Cell Proliferation or
promoter in pterygium is significantly lower [34, 35]. It is Apoptosis in Pterygium. Compared to control conjuncti-
controversial whether MDM2 suppresses p53 transcrip- val tissues, miR-221 expression is upregulated, and miR-
tional activity despite abundant p53 in pterygium. 215 expression is downregulated in pterygium tissues.

Downloaded from http://karger.com/ore/article-pdf/65/5/481/4021749/000523878.pdf by guest on 05 November 2023


Early studies have demonstrated that large tumor sup- miR-221 suppresses the expression of the cyclin-depen-
pressor kinases 1 and 2 (LATS1 and LATS2) are common dent kinase inhibitor p27 kinase protein 1 to drive the
tumor suppressor genes in the DNA damage response transformation from G1 phase to S phase [39]. Further-
pathway induced by UV radiation [36]. Najafi et al. [36] more, miR-221 levels were significantly higher in the
found that the LATS1 and LATS2 methylation levels are fleshly and intermediate groups than in the atrophic
too high to resist the effects of UV DNA damage. Thus, group, indicating that miR-221 is positively related to
methylation levels of LATS1 and LATS2 are related to the pterygium safety. miR-215 targets minichromosome
risk of recurrence. maintenance protein 10 and cell division cycle 25A, and
both of these genes promote cell proliferation by partici-
Histone Modification Biomarkers of Pterygium pating in G1/S and G2/M phase transitions [40]. For this
Histone modification refers to the process by which reason, it was speculated that miR-215 could be used as a
histones undergo methylation, acetylation, adenylation, therapeutic target in pterygium in the future.
ubiquitination, or ADP ribosylation through related en- miR-21 is reported to activate the Akt signaling path-
zymes [37]. Among the histone modifications, methyla- way in various cancers by inhibiting phosphatase and ten-
tion is the most stable, while acetylation is highly dynam- sin homologue [41, 42]. Li et al. [43] have found that the
ic, and the modification level correlates with the degree miR-21 inhibitor suppresses pterygium fibroblast prolif-
of gene silencing and activation. Thus far, no research has eration and promotes apoptosis via phosphatase and ten-
confirmed the actual role of histone modification in the sin homologue-PI3K/Akt signaling, indicating that an in-
development of pterygium, but some researchers have hibitor of miR-21 may be a novel agent used for treat-
speculated that pterygium may be related to histone mod- ment.
ification. miR-145 has been found to be significantly downregu-
Koga et al. [38] found that butyrate and phenylbutyr- lated in the head part of the pterygium [44]. p53 plays an
ate may be used as histone deacetylase inhibitors to in- agonist role in triggering the transcription of various ap-
hibit the expression of fibrosis-related proteins in pteryg- optosis proteins, while MDM2 binds to p53 to initiate the
ium, such as α-smooth muscle actin, collagen I/III, and ubiquitin-mediated degradation of p53 [45]. Therefore,
matrix metalloproteinase 1. This research suggests that the miR-145-MDM2-p53 axis may serve as a potential
histone acetylation may be involved in the occurrence target pathway for pterygium therapy.
and development of pterygium. However, the specific The expression of miR-122 decreases in pterygium,
biomarkers involved require further study and explora- whereas the expression of the targeted gene, B-cell lym-
tion. phoma-w (Bcl-w), is upregulated [46]. miR-122 functions
as a tumor suppressor by targeting Bcl-w, and overex-
Biomarkers from Noncoding RNA Regulation in pressed miR-122 arrests the cell cycle at the G1 phase and
Pterygium activates the caspase-3/7 signaling pathway [47]. Oleic
Noncoding RNAs are a type of RNA that includes acid-based nanoparticles successfully transfer miR-122
miRNAs, long noncoding RNAs (lncRNA), and circular into hepatocytes in mice to effectively suppress Bcl-w ex-

484 Ophthalmic Res 2022;65:481–492 He/Wu


DOI: 10.1159/000523878
Table 2. The abnormal expression of noncoding RNA in pterygium

Noncoding RNA Expression Target genes Phenotypes affected

miR-215 ↓ Cdc25A, Mcm10 Proliferation


miR-221 ↑ p27Kip1 Proliferation
miR-21 ↑ PTEN Proliferation and apoptosis
miR-145 ↓ MDM2 Cell growth and apoptosis
miR-122 ↓ Bcl-w Apoptosis
miR-3175 ↑ Smad7 EMT
miR-218-5p ↓ EGFR Migration and proliferation
miR-200a ↓ ZEB1/ZEB2 EMT
miR-145 ↓ – Severity degree
miR-200 ↓ – EMT
miR-143-3p, miR-181a-2-3p ↑ – Pathogenesis
miR-377-5p, miR-411a-5p
PISRT1, LOC283761, FOXD2-AS1 ↑ – Proliferation

Downloaded from http://karger.com/ore/article-pdf/65/5/481/4021749/000523878.pdf by guest on 05 November 2023


linc-9432 – Differentiation
circ-LAPTM4B ↑ – Proliferation, apoptosis, and migration

Bcl-w, B-cell lymphoma-w; Cdc25A, cell division cycle 25A; circ-LAPTM4B, circular RNA-lysosomal associated pro-
tein transmembrane 4b; EGFR, epidermal growth factor receptor; EMT, epithelial-mesenchymal transition; FOXD2-
AS1, long noncoding RNA-forkhead box d2 adjacent opposite strand RNA 1; Mcm10, minichromosome maintenance
protein 10; MDM2, mouse double minute 2; p27Kip1, p27 kinase protein 1; PISRT1, long noncoding RNA-polled in-
tersex syndrome regulated transcript 1; PTEN, phosphatase and tensin homologue; Smad7, drosophila mothers
against decapentaplegic protein 7; ZEB1/2, zinc finger E-box binding homeobox 1.

pression [48], which indicates the feasibility of miR-122 ZEB1 and ZEB2 are classified as the main regulators of
in pterygium treatment. EMT by inhibiting p53 [54, 55], and p53 is the down-
Some miRNAs Are Associated with Cell Migration or stream molecule of both miR-200a and miR-145.
EMT of Pterygium. In a recent study, upregulated miR- Additionally, Some miRNAs Are Associated with Pte-
3175 was identified in patients with pterygium and di- rygium Severity. The severity grade of pterygium is based
rectly inhibited the expression of Drosophila mothers on a comprehensive assessment of three aspects of slit-
against decapentaplegic protein 7 to promote prolifera- lamp images: extension, vascularity, and thickness of the
tion, migration, invasion, and EMT in human conjuncti- pathological tissues. In a study by Chien et al. [56], miR-
val epithelial cell lines [49]. miR-3175 promotes patho- 145 expression was negatively correlated with pterygium
logical phenotypes of pterygium in normal conjunctival severity and was significantly higher in primary pterygi-
cells, which might be used in clinical therapy. um tissues than in recurrent tissues, which suggests the
Aberrant epidermal growth factor receptor (EGFR) prognostic value of miR-145.
expression or function has been detected in several eye miRNA and mRNA microarray results revealed the
diseases, including angiogenesis, ECM disorder, and important regulatory capacity of the miR-200 family in
EMT [50, 51]. In pterygium epithelial cells, miR-218-5p pterygium [57]. Members of the miR-200 family are all
suppresses the expression of EGFR and inhibits the down- downregulated in pterygium, while the predicted targets
stream PI3K/Akt/mTOR pathway to promote cell migra- of the miR-200 family, including a large number of ECM-
tion and proliferation [52]. This research shows the sig- related proteins, are upregulated [57], suggesting that the
nificance of EGFR in EMT of pterygium and suggests a miR-200 family initiates EMT in pterygium. Lee et al. [58]
possible therapeutic candidate, as agents against EGFR suggested that, compared to normal fibroblasts, miR-
have been developed in various cancers [53]. 143a-3p, miR-181a-2-3p, miR-377-5p, and miR-411a-5p
miR-200a is significantly lower in pterygium than in are overexpressed in pterygium fibroblasts. However,
conjunctiva controls, and further tests have shown that these studies lacked further exploration, so their clinical
the expression level of miR-200a is negatively associated applications require further research.
with zinc finger E-box binding homeobox (ZEB) 1/2.

Review Based on Genetic, Epigenetic, and Ophthalmic Res 2022;65:481–492 485


Proteomic Biomarkers in Pterygium DOI: 10.1159/000523878
LncRNA Biomarkers in Pterygium Overexpressed peroxiredoxin-2 (PRDX2) has been
LncRNAs are a class of noncoding RNAs with a length found in pterygium using a proteomic approach [62].
greater than 200 nucleotides. Studies have shown that ln- PRDX2 is a cytoplasmic enzyme involved in decreasing
cRNAs play important roles in many physiological ac- intracellular reactive oxygen species levels and protecting
tivities, such as cell cycle and differentiation regulation, cells from oxidative stress induced by UV irradiation. Up-
and have become a research hotspot in epigenetics. regulated PRDX2 reduces peroxide-induced apoptosis
The results of lncRNA microarrays can provide a new and necrosis and leads to aberrant cell proliferation and
focus for pterygium. Researchers have found that ln- EMT. The PRDX2 expression level indicates the cause of
cRNA-polled intersex syndrome regulated transcript 1, the disease in that excessive UV exposure promotes the
LOC283761, and lncRNA-forkhead box d2 adjacent op- occurrence of pterygium.
posite strand RNA 1 are upregulated in pterygium and Among the 230 differentially expressed proteins in
promote cell proliferation, while lncRNA-lipoprotein(a) pterygium, aldehyde dehydrogenase 3 family member
like 2 and lncRNA-small nucleolar RNA host gene 1 are A1, protein disulfide isomerase A3, and PRDX2 were sig-
downregulated in pterygium and suppress cell apoptosis nificantly overexpressed and even increased in recurrent
[59]. These lncRNAs hold great promise for clinical ap- pterygium. These three proteins are protective factors

Downloaded from http://karger.com/ore/article-pdf/65/5/481/4021749/000523878.pdf by guest on 05 November 2023


plication. that protect against UV-induced oxidative stress, such as
A novel lncRNA, linc-9432, is upregulated in pterygi- apoptosis suppression and over-proliferation [63]. These
um and may regulate 30 differentiation-related genes. In three biomarkers demonstrate the predominance of oxi-
pterygium fibroblasts, linc-9432 participates in inhibiting dative stress in pterygium formation and may be used to
differentiation-induced cell death, promoting the expres- evaluate prognosis.
sion of stem cell and EMT markers [60]. Linc-9432 is pos- Based on primary pterygium and conjunctival fibro-
itively related to the process of cell differentiation in pte- blasts, a collection of differentially expressed proteins has
rygium, which may be used as an indicator of progression. identified that a large part of the 433 detected proteins are
ECM and fibroblast-secreted proteins [64]. The collagen
CircRNA Biomarkers in Pterygium family is overexpressed in the pterygium, and the over-
CircRNAs are a special class of noncoding RNA mol- sedimentary collagen produced by pterygium fibroblasts
ecules and are the most recent research focus in the RNA facilitates pterygium growth. The upregulation of fibulin
field. A circRNA molecule has a closed and stably ex- 1 and the downregulation of serpin family E member 1
pressed ring structure that protects it from RNA exonu- assemble elastic matrix fibers and inactivate tissue plas-
cleases. CircRNAs are rich in miRNA binding sites and act minogen activator, leading to ECM changes.
as miRNA sponges to block the effects of the miRNAs; this In a recent study, Linghu et al. [65] identified 156 pro-
is called the competitive endogenous RNA mechanism. teins differentially expressed between pterygium and
CircRNA microarrays have indicated that circular RNA- normal conjunctival tissues; among these genes, matrix
lysosomal associated protein transmembrane 4b (circ- metalloproteinase 10 (MMP10) and cluster of differentia-
LAPTM4B) is upregulated in pterygium. Inhibiting the ex- tion 34 (CD34) were significantly upregulated in pteryg-
pression of circ-LAPTM4B in pterygium fibroblasts and ium. MMP10 is a significant factor in denaturing base-
epithelial cells suppresses cell viability and proliferation but ment membrane components to participate in pterygium
promotes apoptosis [61]. Thus, circ-LAPTM4B may be a invasion, while CD34 is recognized as a surface biomark-
valuable new therapeutic agent that is more stable. er of hematopoietic progenitor cells and stem cells to
evaluate angiogenesis. MMP10 and CD34 may regulate
invasion and neovascularization in pterygium and may
Biomarkers from Proteomic Analysis in Pterygium be used for prognosis evaluation.

Proteomics refers to the large-scale study of protein


characteristics, including protein expression level, post- Discussion
translational modification, and protein-protein interac-
tion, and therefore provides a comprehensive under- In the period of DNA sequencing development, genet-
standing of disease at the protein level. Many studies have ics has become a hotspot in the study of disease biomark-
investigated the protein profile of pterygium using pro- ers. With advancements in chip technology, RNA se-
teomic technology. quencing, mass spectrometry, epigenetics, and pro-

486 Ophthalmic Res 2022;65:481–492 He/Wu


DOI: 10.1159/000523878
Table 3. The disordered source and
phenotypes affected of biomarkers Biomarker Disordered Expression Phenotypes affected
source

HB-EGF Genomics ↑ Proliferation


p53 Progression related Proliferation and apoptosis
K-ras ↑ Proliferation
NGAL ↑ ECM remodeling
MMP9 ↑ Inflammatory
IGFBP3 ↓ Proliferation
ACE ↑ Vasoconstriction
CRIM1 ↓ Differentiation
SLC1A5 ↑ Further study
p16 Epigenetics ↑ Proliferation
E-cadherin ↑ EMT
TGM2 ↑ ECM remodeling
MMP2 ↓ ECM remodeling
CD24 ↓ ECM remodeling

Downloaded from http://karger.com/ore/article-pdf/65/5/481/4021749/000523878.pdf by guest on 05 November 2023


MDM2 ↓ Proliferation
LATS1/LATS2 ↑ Apoptosis
miR-221-p27Kip1 ↑ Proliferation
miR-215-Cdc25A/ ↓ Proliferation
Mcm10
miR-21-PTEN ↑ Proliferation and apoptosis
miR-145-MDM2 Progression related Proliferation and apoptosis
miR-122-Bcl-w ↓ Apoptosis
miR-3175 ↑ EMT
miR-218-5p-EGFR ↓ Migration and proliferation
miR-200a-ZEB1/ZEB2 ↓ EMT
linc-9432 ↑ Differentiation
Circ-LAPTM4B ↑ Proliferation and apoptosis
PRDX2 Proteomics ↑ Necrosis and EMT
ALDH3A1 ↑ Apoptosis
PDLA3 ↑ Apoptosis
FN1 ↑ ECM remodeling
SERPINE1 ↓ ECM remodeling
MMP10 ↑ Invasion
CD34 ↑ Neovascularization

ALDH3A1, aldehyde dehydrogenase 3 family member A1; Bcl-w, B-cell lymphoma-w;


CD24, cluster of differentiation 24; CD34, cluster of differentiation 34; Cdc25A, cell division
cycle 25A; circ-LAPTM4B, circular RNA-lysosomal associated protein transmembrane 4b;
ECM, extracellular matrix; EGFR, epidermal growth factor receptor; EMT, epithelial-mesen-
chymal transition; FN1, fibulin 1; LATS1/2, large tumor suppressor kinase 1/2; Mcm10, mini-
chromosome maintenance protein 10; MDM2, mouse double minute 2; MMP2, matrix me-
talloproteinase 2; MMP10, matrix metalloproteinase 10; p27Kip1, p27 kinase protein 1;
PDLA3, protein disulfide isomerase A3; PISRT1, long noncoding RNA-polled intersex syn-
drome regulated transcript 1; PRDX2, peroxiredoxin-2; PTEN, phosphatase and tensin ho-
mologue; SERPINE1, serpin family 1; TGM2, transglutaminase 2; ZEB1/2, zinc finger E-box
binding homeobox 1; NGAL, neutrophil gelatinase-associated lipocalin; IGFBP3, insulin-like
growth factor-binding protein 3.

teomics have gradually become the core of mechanistic ment for pterygium is surgical excision, and many ad-
research. All the mentioned biomarkers in our review junctive agents may be used, such as doxycycline, mito-
have been listed in Table 3. mycin C, and 5-fluorouracil [66]. However, these agents
Pterygium is a multifactorial disease with a high inci- have toxicity and side effects and are only used postop-
dence and recurrence rate. The most common treat- eratively.

Review Based on Genetic, Epigenetic, and Ophthalmic Res 2022;65:481–492 487


Proteomic Biomarkers in Pterygium DOI: 10.1159/000523878
Epigenetics links genetics to the environment; thus, classified as potential mechanisms and shown in a net-
the importance of epigenetics in environmental respons- work (Fig. 2).
es should be well established in pterygium to better pre- Some core molecules are observed in the complicated
vent the occurrence of disease. Primary pterygium patho- biomarker network. The first central point is p53, whose
genesis is mainly associated with UV light exposure, and expression levels and functions are regulated by various
some biomarkers support this conclusion. LATS1 and factors [16, 17, 19]. First, the mutation of p53 is partly
LATS2 act as effectors against UV-induced DNA damage regulated by UV light [19]. Second, levels of MDM2
and participate in regulating the cell cycle and apoptosis methylation are also involved in p53 expression because
[67]. In addition, aberrant methylation of p16 is specu- MDM2 acts as an E3 ubiquitin protein ligase to degrade
lated to be associated with UV exposure [30]. p53 [72]. Third, miR-145 can regulate p53 via the miR-
As a neoplasm-like disease, cancer includes changes 145-MDM2-p53 axis [44]. Furthermore, p16 is a protec-
similar to those in pterygium. p16 and p53 are both tumor tive factor for p53, whereas abnormally activated K-ras
suppressors that induce growth arrest or apoptosis; p16 inhibits p53 expression. Teng et al. [44] found that miR-
acts as a stabilizer of p53 to interact with the E3 ubiquitin- 145 expression is lower in the head of pterygium than in
protein ligase MDM2 [68]. Both p16 and p53 share a the body, whereas Chien et al. [56] indicated that miR-

Downloaded from http://karger.com/ore/article-pdf/65/5/481/4021749/000523878.pdf by guest on 05 November 2023


common function in controlling the G1 phase of the cell 145 is negatively correlated with pterygium severity. To
cycle. In contrast, K-ras possesses intrinsic GTPase activ- the best of our knowledge, these two conclusions are not
ity, and mutated K-ras induces transcriptional silencing contradictory. The head of pterygium is affected at the
of tumor suppressor genes [69]. Therefore, some antican- beginning of disease development; at this stage, the low
cer therapies targeting cancer genes may also be feasible expression of miR-145 in cells promotes rapid cell pro-
for pterygium treatment. For example, siRNA or shRNAs liferation. An abnormal increase in cell proliferation
can knockdown mutant p53 to restore activity, gentami- leads to increased disease severity and a poor prognosis.
cin can induce readthroughs of premature termination The above research demonstrated that the miR-
codons, and CDB3 can restore sequence-specific DNA 145-MDM2-p53 axis may be a new target for prognosis
binding and transcriptional activity [70]. In addition, or therapy.
MDM2 small-molecule inhibitors have advanced into miRNA microarrays have been used to identify new
human clinical trials [71], which may also be taken into study targets. The latest study performed by Liu et al. [73]
consideration for therapy in the future. mainly focused on the expression patterns of mRNAs, ln-
It is worth noting that p53 is ectopically expressed in cRNAs, and circRNAs. However, much of the informa-
pterygium. miR-145 is predominantly expressed in the tion was listed rather than verified. Based on research us-
basal pterygium epithelium, whereas oncogene MDM2 ing these microarrays, miR-143-3p lies at the intersection
expression is abundant in pterygium epithelium and stro- [40, 46, 57, 58]. miR-143-3p has been reported as a tumor
ma [44]. Ectopic expression of miR-145 in pterygium suppressor that suppresses proliferation, migration, and
cells induces G1 arrest, downregulates MDM2 and ele- invasion, and promotes apoptosis in various tumors [74,
vates p53 expression. 75]. The use of a miR-143-3p mimic has provided a new
In addition to the tumor-related proteins, many ECM strategy for pterygium therapy.
proteins are abnormally expressed in pterygium. The Due to their invertibility, epigenetic biomarkers may
most notable result from the WGCNA was the identifi- be more applicable in clinical settings. Small molecule
cation of fibulin 1, one of the five hub genes that serves drugs for DNA methyltransferases and histone deacety-
as a potential regulator of epithelial cell migration, ECM lases are in the developmental stage and have made some
deposition, and EMT and has also been verified in pro- achievements in tumor treatment. Furthermore, the use
teomic findings [28, 64]. Members of the MMP family, of the miR-122 antagonist in patients with hepatitis C has
including MMP2, MMP9, and MMP10, are overex- entered phase III clinical trials [76]. This application in-
pressed in the epithelial layer of the pterygium to acti- dicates the prospect of miRNA antagonists and activators
vate fibroblasts and promote EMT(22, 32, 65). These being used in the clinic and provides a possibility for non-
profiling findings point to the core role of ECM remod- invasive treatment of pterygium. The first studies using
eling in the disease progression, suggesting an impor- proteomics in pterygium were performed in 2006, and
tant research prospect for ECM remodeling and reversal their study specimens were tears from patients with pte-
of EMT in pterygium treatment. Biomarkers identified rygium [77]. Although many genetic biomarkers and
in genetics, epigenetics, and proteomics studies were proteomic biomarkers lack deep exploration, noninva-

488 Ophthalmic Res 2022;65:481–492 He/Wu


DOI: 10.1159/000523878
miR– miR–
181a– 143a– miR–
2-3p 3p 221 miR–
122
miR– miR– miR–
miR– 377– 145 21
TIMP 411a– 5p miR–
2/3 fibuli 5p 21
n1 miR– FOX
215 D2–
miR– circ– AS1
3175 Pathogenesis LAPT
/Severity M4B LOC
MMP SNH
TGM 2 2837
G1
2 61
miR– Migration Proliferation LPAL
200a/ 2
/EMT /Apoptosis PISR
b
T1
E– MD
cadhe

Downloaded from http://karger.com/ore/article-pdf/65/5/481/4021749/000523878.pdf by guest on 05 November 2023


p16 M2
rin MMP Others CRI
10 PRD M1
CD24 X2
PDL LAS
miR– A3 T1/2
218–
5p linc– K-ras
SERP 9432 ACE
INE1 CD34 ALD p53
H3A1

CTL ZNF2
A4 05

Fig. 2. The mechanism network of biomarkers in pterygium. EMT, epithelial-mesenchymal transition; “others”
include neovascularization, cell differentiation, and fibrosis.

Clinical
applications
Upregulated Downregulated

ACE, MMP2, CD24,


PRDX2, ALDH3A1, miR-145, TGM2,
Severity PDIA3, fibulin 1, E-cadherin, SERPINE1
MMP10, CD34

Prognosis
prediction
HB-EGF, K-ras, NGAL,
CRIM1, IGFBP3,
Recurrence MMP9, Linc-9432,
LATS1/LATS2
VEGFA, VEGFC

MDM2, miR-3175, p53, p16, miR-218-5P,


Therapeutic miR-21, miR-221, miR-145, miR-122,
development circ-LAPTM4B, miR-200a, miR-215,
Semaphorin 7a FGF21

Fig. 3. Clinical applications of biomarkers in pterygium.

Review Based on Genetic, Epigenetic, and Ophthalmic Res 2022;65:481–492 489


Proteomic Biomarkers in Pterygium DOI: 10.1159/000523878
sive detection may be used to evaluate prognosis in the Conclusion
future. The probable clinical applications of these bio-
markers are shown in Figure 3. In summary, this review introduced multiple bio-
In addition to biomarkers from genetics, epigenetics, markers in pterygium identified from genetics, epi-
and proteomics, other biomarkers may also be used clin- genetics, and proteomics studies. It was concluded that
ically. A recent study demonstrated that serum fibroblast these biomarkers might play critical roles in severity de-
growth factor 21, which is a target for neovascularization gree judgement, recurrence risk prediction, and adjuvant
reduction, is lower in patients with pterygium [78]. Fur- therapy in pterygium. Among these biomarkers, it is
thermore, fibroblast growth factor 21 is negatively related noteworthy that p53 is involved in pterygium by abnor-
to cholesterol, triglyceride, and LDL levels; therefore, im- mal changes in various pathways and even has different
paired lipid metabolism should be considered for thera- phenotypes in different development stages. The abnor-
py. Essential biomarkers in tears, including interleukin-6, mal changes of the cell cycle dominated by p53 may be a
interleukin-8, and vascular endothelial growth factor new target for pterygium therapy.
(VEGF), are positively correlated with disease severity,
and may also be used for postoperative monitoring [79].

Downloaded from http://karger.com/ore/article-pdf/65/5/481/4021749/000523878.pdf by guest on 05 November 2023


Han et al. [80] found that semaphorin 7a is upregulated Conflict of Interest Statement
in pterygium tissues, and the level in recurrent pterygium
The authors have no conflicts of interest to declare.
is even higher than that in primary pterygium. Semapho-
rin 7a shows a positive association with some angiogenic
factors, such as VEGFA and VEGFR, and migration-pro-
Funding Sources
moting factors, such as β1-integrin. Based on the malig-
nant nature of semaphorin 7a in pterygium development This manuscript did not receive any funding.
and recurrence, its inhibitor may act as a new target for
prognosis.
Author Contributions

Siying He contributed to the conception and collected the data


for the work; while Zhaoxia Wu drafted the work and revised it.

References
1 Zhong H, Cha X, Wei T, Lin X, Li X, Li J, et al. 7 Liu T, Liu Y, Xie L, He X, Bai J. Progress in the 14 Zada M, Pattamatta U, White A. Modulation
Prevalence of and risk factors for pterygium pathogenesis of pterygium. Curr Eye Res. of fibroblasts in conjunctival wound healing.
in rural adult Chinese populations of the Bai 2013;38(12):1191–7. Ophthalmology. 2018;125(2):179–92.
nationality in Dali: the Yunnan Minority Eye 8 Tsai YY, Chang CC, Chiang CC, Yeh KT, 15 Nolan TM, DiGirolamo N, Sachdev NH,
Study. Invest Ophthalmol Vis Sci. 2012; Chen PL, Chang CH, et al. HPV infection and Hampartzoumian T, Coroneo MT, Wakefield
53(10):6617–21. p53 inactivation in pterygium. Mol Vis. 2009; D. The role of ultraviolet irradiation and hep-
2 Chen YF, Hsiao CH, Ngan KW, Yeung L, Tan 15:1092–7. arin-binding epidermal growth factor-like
HY, Yang KH, et al. Herpes simplex virus and 9 Pinkerton OD, Hokama Y, Shigemura LA. Im- growth factor in the pathogenesis of pterygi-
pterygium in Taiwan. Cornea. 2008; 27(3): munologic basis for the pathogenesis of pteryg- um. Am J Pathol. 2003;162(2):567–74.
311–3. ium. Am J Ophthalmol. 1984;98(2):225–8. 16 Schneider BG, John-Aryankalayil M, Rowsey
3 Chui J, Coroneo MT, Tat LT, Crouch R, Wake- 10 Peiretti E, Dessì S, Putzolu M, Fossarello M. JJ, Dushku N, Reid TW. Accumulation of p53
field D, Di Girolamo N. Ophthalmic pterygi- Hyperexpression of low-density lipoprotein protein in pterygia is not accompanied by
um: a stem cell disorder with premalignant fea- receptors and hydroxy-methylglutaryl-coen- TP53 gene mutation. Exp Eye Res. 2006;82(1):
tures. Am J Pathol. 2011;178(2):817–27. zyme A-reductase in human pinguecula and 91–8.
4 Clearfield E, Muthappan V, Wang X, Kuo IC. primary pterygium. Invest Ophthalmol Vis 17 Tsai YY, Cheng YW, Lee H, Tsai FJ, Tseng SH,
Conjunctival autograft for pterygium. Co- Sci. 2004;45(11):3982–5. Chang KC. P53 gene mutation spectrum and
chrane Database Syst Rev. 2016;2:CD011349. 11 Hacioglu D, Erdol H. Developments and cur- the relationship between gene mutation and
5 Garzon-Chavez DR, Quentin E, Harrison SL, rent approaches in the treatment of pterygi- protein levels in pterygium. Mol Vis. 2005;11:
Parisi AV, Butler HJ, Downs NJ. The geospa- um. Int Ophthalmol. 2017;37(4):1073–81. 50–5.
tial relationship of pterygium and senile cata- 12 Hacıoğlu D, Erdöl H. Developments and cur- 18 Mahesh M, Mittal SK, Kishore S, Singh A,
ract with ambient solar ultraviolet in tropical rent approaches in the treatment of pterygi- Gupta N, Rana R. Expression of p53 and Ki-
Ecuador. Photochem Photobiol Sci. 2018; um. Int Ophthalmol. 2017;37(4):1073–81. 67 proteins in patients with increasing sever-
17(8):1075–83. 13 Wang G, Dong N, Wang Y, Li J, Dong F, Jey- ity and duration of pterygium. Indian J Oph-
6 Pang Y, Rose T. Rapid growth of pterygium alatha V, et al. Epithelial dysplasia in pterygi- thalmol. 2021;69(4):847–50.
after photorefractive keratectomy. Optome- um postoperative granuloma. Exp Eye Res.
try. 2006;77(10):499–502. 2018;175:199–206.

490 Ophthalmic Res 2022;65:481–492 He/Wu


DOI: 10.1159/000523878
19 Maxia C, Perra MT, Demurtas P, Minerba L, 34 Arish M, Kordi-Tamandani DM, Sangterash 49 Zhong X, Tang J, Li H, Shi X, Wu Y, Xia D, et
Murtas D, Piras F, et al. Expression of survivin MH, Poyandeh R. Assessment of promoter al. MiR-3175 promotes epithelial-mesenchy-
protein in pterygium and relationship with hypermethylation and expression profile of mal transition by targeting Smad7 in human
oxidative DNA damage. J Cell Mol Med. 2008; P14ARF and MDM2 genes in patients with conjunctiva and pterygium. FEBS letters.
12(6A):2372–80. pterygium. Eye Contact Lens. 2016;42(1):e4– 2020 Apr;594(7):1207–17.
20 Detorakis ET, Zafiropoulos A, Arvanitis DA, 7. 50 Di Girolamo N, Coroneo M, Wakefield D.
Spandidos DA. Detection of point mutations 35 Cao D, Ng TK, Yip YWY, Young AL, Pang Epidermal growth factor receptor signaling is
at codon 12 of KI-ras in ophthalmic pterygia. CP, Chu WK, et al. p53 inhibition by MDM2 partially responsible for the increased matrix
Eye. 2005;19(2):210–4. in human pterygium. Exp Eye Res. 2018; 175: metalloproteinase-1 expression in ocular epi-
21 Ozturk BT, Yıldırım MS, Zamani A, Bozkurt 142–7. thelial cells after UVB radiation. Am J Pathol.
B. K-ras oncogene mutation in pterygium. 36 Najafi M, Kordi-Tamandani DM, Arish M. 2005;167(2):489–503.
Eye. 2017;31(3):491–8. Evaluation of LATS1 and LATS2 promoter 51 Balasoiu AT, Ciurea RN, Manescu MR, Mo-
22 John-Aryankalayil M, Dushku N, Jaworski methylation with the risk of pterygium for- canu CL, Stepan AE, Balasoiu M, et al. Assess-
CJ, Cox CA, Schultz G, Smith JA, et al. Micro- mation. J Ophthalmol. 2016;2016:5431021. ment of VEGF and EGFR in the study of an-
array and protein analysis of human pterygi- 37 Bannister AJ, Kouzarides T. Regulation of giogenesis of eyelid carcinomas. Rom J Mor-
um. Mol Vis. 2006;12:55–64. chromatin by histone modifications. Cell Res. phol Embryol. 2016;57(4):1229–34.
23 Wong YW, Chew J, Yang H, Tan DT, Beuer- 2011;21(3):381–95. 52 Han S, Chen Y, Gao Y, Sun B, Kong Y. Mi-
man R. Expression of insulin-like growth fac- 38 Koga Y, Maeshige N, Tabuchi H, Uemura M, croRNA-218-5p inhibit the migration and
tor binding protein-3 in pterygium tissue. Br Aoyama-Ishikawa M, Miyoshi M, et al. Sup- proliferation of pterygium epithelial cells by

Downloaded from http://karger.com/ore/article-pdf/65/5/481/4021749/000523878.pdf by guest on 05 November 2023


J Ophthalmol. 2006;90(6):769–72. pression of fibrosis in human pterygium fi- targeting EGFR via PI3K/Akt/mTOR signal-
24 Demurtas P, Orru G, Coni P, Minerba L, Cor- broblasts by butyrate and phenylbutyrate. Int ing pathway. Exp Eye Res. 2018;178:37–45.
rias M, Sirigu P, et al. Association between the J Ophthalmol. 2017;10(9):1337–43. 53 Vecchione L, Jacobs B, Normanno N, Ciardi-
ACE insertion/deletion polymorphism and 39 Wu CW, Cheng YW, Hsu NY, Yeh KT, Tsai ello F, Tejpar S. EGFR-targeted therapy. Exp
pterygium in Sardinian patients: a population YY, Chiang CC, et al. MiRNA-221 negatively Cell Res. 2011;317(19):2765–71.
based case-control study. BMJ Open. 2014; regulated downstream p27Kip1 gene expres- 54 Berx G, Raspé E, Christofori G, Thiery JP, Sl-
4(10):e005627. sion involvement in pterygium pathogenesis. eeman JP. Pre-EMTing metastasis? Recapitu-
25 Kria L, Ohira A, Amemiya T. Immunohisto- Mol Vis. 2014;20:1048–56. lation of morphogenetic processes in cancer.
chemical localization of basic fibroblast 40 Lan W, Chen S, Tong L. MicroRNA-215 regu- Clin Exp Metastasis. 2007;24(8):587–97.
growth factor, platelet derived growth factor, lates fibroblast function: insights from a hu- 55 Kim T, Veronese A, Pichiorri F, Lee TJ, Jeon
transforming growth factor-beta and tumor man fibrotic disease. Cell Cycle. 2015;14(12): YJ, Volinia S, et al. p53 regulates epithelial-
necrosis factor-alpha in the pterygium. Acta 1973–84. mesenchymal transition through microRNAs
Histochem. 1996;98(2):195–201. 41 Cui M, Liu W, Zhang L, Guo F, Liu Y, Chen targeting ZEB1 and ZEB2. J Exp Med. 2011;
26 Yazar S, Cuellar-Partida G, McKnight CM, F, et al. Over-expression of miR-21 and lower 208(5):875–83.
Quach-Thanissorn P, Mountain JA, Coroneo PTEN levels in Wilms’ tumor with aggressive 56 Chien KH, Chen SJ, Liu JH, Woung LC, Chen
MT, et al. Genetic and environmental factors behavior. Tohoku J Exp Med. 2017; 242(1): JT, Liang CM, et al. Correlation of microR-
in conjunctival UV autofluorescence. JAMA 43–52. NA-145 levels and clinical severity of pteryg-
Ophthalmol. 2015;133(4):406–12. 42 Wu Y, Song Y, Xiong Y, Wang X, Xu K, Han ia. Ocul Surf. 2013;11(2):133–8.
27 Tsai CB, Hsia NY, Wang ZH, Yang JS, Hsu B, et al. MicroRNA-21 (MiR-21) promotes 57 Engelsvold DH, Utheim TP, Olstad OK, Gon-
YM, Wang YC, et al. The contribution of cell growth and invasion by repressing tumor zalez P, Eidet JR, Lyberg T, et al. miRNA and
MMP-9 genotypes to pterygium in Taiwan. suppressor PTEN in colorectal cancer. Cell mRNA expression profiling identifies mem-
Anticancer Res. 2020;40(8):4523–7. Physiol Biochem. 2017;43(3):945–58. bers of the miR-200 family as potential regula-
28 Chen Y, Wang H, Jiang Y, Zhang X, Wang Q. 43 Li X, Dai Y, Xu J. MiR-21 promotes pterygium tors of epithelial-mesenchymal transition in
Transcriptional profiling to identify the key cell proliferation through the PTEN/AKT pterygium. Exp Eye Res. 2013;115:189–98.
genes and pathways of pterygium. PeerJ. pathway. Mol Vis. 2018;24:485–94. 58 Lee JH, Jung SA, Kwon YA, Chung JL, Kim
2020;8:e9056. 44 Teng Y, Yam GH, Li N, Wu S, Ghosh A, Wang US. Expression of microRNAs in fibroblast of
29 Robertson KD. DNA methylation and human N, et al. MicroRNA regulation of MDM2-p53 pterygium. Int J Ophthalmol. 2016;9(7):967–
disease. Nat Rev Genet. 2005;6(8):597–610. loop in pterygium. Exp Eye Res. 2018; 169: 72.
30 Chen PL, Cheng YW, Chiang CC, Tseng SH, 149–56. 59 Liu J, Ding X, Yuan L, Zhang X. Identification
Chau PS, Tsai YY. Hypermethylation of the 45 Levine AJ, Oren M. The first 30 years of p53: of pterygium-related long non-coding RNAs
p16 gene promoter in pterygia and its associa- growing ever more complex. Nat Rev Cancer. and expression profiling by microarray analy-
tion with the expression of DNA methyltrans- 2009;9(10):749–58. sis. Int J Mol Med. 2016;38(2):529–36.
ferase 3b. Mol Vis. 2006;12:1411–6. 46 Cui YH, Li HY, Gao ZX, Liang N, Ma SS, 60 Lan W, Hou A, Lakshminarayanan R, Lim
31 Riau AK, Wong TT, Lan W, Finger SN, Meng FJ, et al. Regulation of apoptosis by YP, Tong L. Linc-9432 is a novel pterygium
Chaurasia SS, Hou AH, et al. Aberrant DNA miR-122 in pterygium via targeting Bcl-w. In- lincRNA which regulates differentiation of fi-
methylation of matrix remodeling and cell ad- vest Ophthalmol Vis Sci. 2016;57(8):3723–30. broblasts. FEBS Lett. 2018;592(7):1173–84.
hesion related genes in pterygium. PLoS One. 47 Ma J, Wu Q, Zhang Y, Li J, Yu Y, Pan Q, et al. 61 Li XM, Ge HM, Yao J, Zhou YF, Yao MD, Liu
2011;6(2):e14687. MicroRNA sponge blocks the tumor-sup- C, et al. Genome-wide identification of circu-
32 Lanza M, Benincasa G, Costa D, Napoli C. pressing functions of microRNA-122 in hu- lar RNAs as a novel class of putative biomark-
Clinical role of epigenetics and network anal- man hepatoma and osteosarcoma cells. Oncol ers for an ocular surface disease. Cell Physiol
ysis in eye diseases: a translational science re- Rep. 2014;32(6):2744–52. Biochem. 2018;47(4):1630–42.
view. J Ophthalmol. 2019;2019:2424956. 48 Wang X, Yu B, Ren W, Mo X, Zhou C, He H, 62 Bautista-de Lucio VM, Lopez-Espinosa NL,
33 Young C-H, Chiu Y-T, Shih T-S, Lin W-R, et al. Enhanced hepatic delivery of siRNA and Robles-Contreras A, Perez-Cano HJ, Mejia-
Chiang C-C, Chou Y-E, et al. E-cadherin pro- microRNA using oleic acid based lipid Lopez H, Mendoza G, et al. Overexpression of
moter hypermethylation may contribute to nanoparticle formulations. J Control Release. peroxiredoxin 2 in pterygium. A proteomic
protein inactivation in pterygia. Mol Vis. 2013;172(3):690–8. approach. Exp Eye Res. 2013;110:70–5.
2010;16:1047–53.

Review Based on Genetic, Epigenetic, and Ophthalmic Res 2022;65:481–492 491


Proteomic Biomarkers in Pterygium DOI: 10.1159/000523878
63 Kim SW, Lee J, Lee B, Rhim T. Proteomic 69 Serra RW, Fang M, Park SM, Hutchinson L, 75 He Z, Yi J, Liu X, Chen J, Han S, Jin L, et al.
analysis in pterygium; upregulated protein Green MR. A KRAS-directed transcriptional MiR-143-3p functions as a tumor suppressor
expression of ALDH3A1, PDIA3, and silencing pathway that mediates the CpG is- by regulating cell proliferation, invasion and
PRDX2. Mol Vis. 2014;20:1192–202. land methylator phenotype. Elife. 2014; 3: epithelial-mesenchymal transition by target-
64 Hou A, Law KP, Tin MQ, Lim YP, Tong L. In e02313. ing QKI-5 in esophageal squamous cell carci-
vitro secretomics study of pterygium-derived 70 Kanasty R, Dorkin JR, Vegas A, Anderson D. noma. Mol Cancer. 2016;15(1):51.
fibroblasts by iTRAQ-based quantitative pro- Delivery materials for siRNA therapeutics. 76 Janssen HL, Reesink HW, Lawitz EJ, Zeuzem
teomics strategy. Exp Eye Res. 2016; 153: 14– Nat Mater. 2013;12(11):967–77. S, Rodriguez-Torres M, Patel K, et al. Treat-
22. 71 Wang S, Zhao Y, Aguilar A, Bernard D, Yang ment of HCV infection by targeting microR-
65 Linghu D, Guo L, Zhao Y, Liu Z, Zhao M, CY. Targeting the MDM2-p53 protein-pro- NA. N Engl J Med. 2013;368(18):1685–94.
Huang L, et al. iTRAQ-based quantitative tein interaction for new cancer therapy: prog- 77 Zhou L, Beuerman RW, Foo Y, Liu S, Ang LP,
proteomic analysis and bioinformatics study ress and challenges. Cold Spring Harb Per- Tan DT. Characterisation of human tear pro-
of proteins in pterygia. Proteomics Clin Appl. spect Med. 2017;7(5):a026245. teins using high-resolution mass spectrome-
2017;11(7–8. 72 Manfredi JJ. The Mdm2-p53 relationship try. Ann Acad Med Singap. 2006;35(6):400–7.
66 Young AL, Cao D, Chu WK, Ng TK, Yip evolves: Mdm2 swings both ways as an onco- 78 Yaghoobi G, Shokoohi-Rad S, Jafarzadeh H,
YWY, Jhanji V, et al. The evolving story of gene and a tumor suppressor. Genes Dev. Abdollahi E. Serum fibroblast growth factor
pterygium. Cornea. 2018;37 Suppl 1:S55–7. 2010;24(15):1580–9. 21 in patients with and without pterygia. J
67 Takahashi Y, Miyoshi Y, Takahata C, Irahara 73 Liu X, Zhang J, Nie D, Zeng K, Hu H, Tie J, et Ophthalmic Vis Res. 2020;15(1):38–44.
N, Taguchi T, Tamaki Y, et al. Down-regula- al. Comparative transcriptomic analysis to 79 Van Acker SI, Haagdorens M, Roelant E, Roz-

Downloaded from http://karger.com/ore/article-pdf/65/5/481/4021749/000523878.pdf by guest on 05 November 2023


tion of LATS1 and LATS2 mRNA expression identify the important coding and non-cod- ema J, Possemiers T, Van Gerwen V, et al. Pte-
by promoter hypermethylation and its asso- ing RNAs involved in the pathogenesis of pte- rygium pathology: a prospective case-control
ciation with biologically aggressive pheno- rygium. Front Genet. 2021;12:646550. study on tear film cytokine levels. Mediators
type in human breast cancers. Clin Cancer 74 Chen J, Chen X. MYBL2 is targeted by miR- Inflamm. 2019;2019:9416262.
Res. 2005;11(4):1380–5. 143-3p and regulates breast cancer cell prolif- 80 Han YF, Liu Z, Wang B, Zhu W, Li JZ, Qi YQ,
68 Schafer KA. The cell cycle: a review. Vet eration and apoptosis. Oncol Res. 2018;26(6): et al. Semaphorin 7a participants in pterygi-
Pathol. 1998;35(6):461–78. 913–22. um by regulating vascular endothelial growth
factor. Int J Ophthalmol. 2019;12(6):892–7.

492 Ophthalmic Res 2022;65:481–492 He/Wu


DOI: 10.1159/000523878

You might also like