1. Introduction
Fructus Lycii (FL), also known as the
Lycium barbarum or wolfberry, is a plant native to China’s Ningxia region that can be used as both food and medicine. The first documentation of this herb can be traced back to the
Shennong Herbal Classics, an ancient document of great significance in the field of herbal medicine. The
Compendium of Materia Medica provides a comprehensive overview of the benefits of this substance, highlighting its ability to improve vision by nourishing the liver and kidneys.
Lycium barbarum polysaccharides (LBP) are the primary bioactive components of
Lycium barbarum, and exhibit numerous biological activities, such as inhibition of oxidative stress, anti-inflammatory responses, inhibition of tumor growth, regulation of lipid metabolism, and suppression of fibrosis. By inhibiting cell apoptosis, LBP protects human corneal epithelial cells (HCEC) [
1]. LBP substantially reduces the increased apoptosis rate induced by H
2O
2 and the production of ROS while increasing the level of the antioxidant enzyme SOD [
2]. To date, research on the inhibition of corneal fibrosis by LBP has been limited. In a corneal fibroblast injury model [
3,
4], the ophthalmology research team at the University of Hong Kong Li Ka Shing Faculty of Medicine found that pretreatment with LBP reduced the expression of fibrosis-related proteins, including α-smooth muscle actin (α-SMA), fibronectin (FN), and collagen (Col). LBP pretreatment also decreased the contraction and rigidity of hydrogels embedded with corneal fibroblasts, indicating that LBP can reduce corneal fibrotic scarring by inhibiting the differentiation, proliferation, and Extracellular matrix (ECM) secretion of corneal fibroblasts. Nonetheless, the precise mechanisms of its action remain unclear.
The cornea is a highly specialized, transparent, circular tissue composed of epithelial, anterior elastic, stromal, posterior elastic, and endothelial layers. It serves the dual purpose of protecting the contents of the eye and contributing to the optical system [
5]. Due to its position at the front of the eye, the cornea is susceptible to a variety of external assaults and injuries. Over a million cases of ocular injuries occur annually in the United States [
6]. It is estimated that approximately 20% of the population will experience ocular trauma during their lifetime. Inadequate restoration following corneal injury can result in corneal epithelial defects, stromal fibrotic scarring, vision impairment and, in extreme cases, corneal blindness [
7]. Corneal scarring accounts for 2.4% of the world’s 253 million cases of blindness and visual impairment, making it the fourth leading cause of blindness overall [
8,
9]. There are currently no effective treatments for vision loss caused by corneal injuries. Steroid drugs such as dexamethasone (Dex) are commonly used, but their efficacy is limited and they frequently cause complications. Corneal transplantation is the only treatment option for corneal blindness, but it is accompanied by high surgical costs, a lack of corneal donors, potential infection risks, and rejection reactions, which place a significant emotional and financial burden on patients and significantly diminish their quality of life [
10]. Therefore, the search for new pharmaceuticals and methods for preventing and treating corneal injuries is clinically significant.
Based on the etiology of corneal diseases, corneal injuries can be classified into five main classes: biological, physical, chemical, impact from other organs, and genetic factors. Chemical corneal injury is a prevalent form of corneal damage that can result in corneal epithelial defects, corneal inflammation, corneal opacification, and fibrotic scar formation. It functions as an exhaustive model for corneal injury research. In clinical contexts, corneal injuries are frequently accompanied by inflammation and oxidative stress [
11,
12]. During oxidative stress, excessive production of reactive oxygen species (ROS) can cause cell injury by targeting DNA, proteins, and intracellular pathways [
13]. H
2O
2 is a common reactive oxygen species that can cause oxidative stress and inflammatory responses [
14].
To explore the potential effects and specific mechanisms of LBP on in vivo and in vitro corneal injury models, we first utilized the emerging traditional Chinese medicine network pharmacology approach, based on recent advancements in network theory and systems biology, to predict and analyze key targets and pathways by which LBP may improve corneal injury. Then, we established corneal alkali burn and H2O2-induced HCEC cell injury models in rats and intervened with LBP. Observing the effects of LBP on in vivo corneal injury and ex vivo corneal epithelial cell injury, we investigated the underlying mechanisms. In addition, we validated the network pharmacology-predicted targets in biological processes in an effort to provide reference points for future research and clinical applications of LBP in the treatment of corneal injury.
3. Discussion
Corneal injury repair is a complex process involving oxidative stress, inflammation, cell apoptosis, migration, proliferation, differentiation, and remodeling of the extracellular matrix. LBP are the primary bioactive constituents of goji berries, renowned for their diverse biological activities, such as inhibiting oxidative stress, anti-inflammatory responses, modulating lipid metabolism, and suppressing fibrosis. LBP has been shown to prevent and protect against a variety of ocular diseases. Previous research [
1,
4,
17] indicates that LBP intervention can inhibit HCEC apoptosis, promote proliferation, and reduce the expression of pro-fibrotic proteins and inflammatory cytokines in a model of corneal matrix injury. However, the precise mechanisms remain obscure. In this study, we initially employed network pharmacology techniques to investigate the potential mechanisms by which LBP ameliorates corneal injury. By constructing in vivo and in vitro corneal injury models, we subsequently validated the protective effects of LBP and its associated mechanisms. This provides new insights and directions for LBP corneal injury treatment research.
This study identified ten monosaccharide components of LBP that are efficacious for corneal injury treatment. The most significant effects were exhibited by galacturonic acid, glucuronic acid, rhamnose, and galactose. These four monosaccharide components are regarded as the most vital and essential for the treatment of corneal injury. Galacturonic acid is a functional monosaccharide, and its concentration is a significant factor in LBP activity [
15]. Bao Le and others [
18] discovered in their investigation of soybean residue polysaccharides that this polysaccharide is primarily composed of galacturonic acid, xylose, and arabinose. Experiments on cells in vitro revealed its potent anti-inflammatory properties, as it inhibited the production of nitric oxide, tumor necrosis factor (TNF)-α, interleukin (IL)-1, and IL-6. Sylla Balla and colleagues [
19] discovered that xylose could reduce the intestinal epithelial cell inflammation caused by lipopolysaccharide. Xylose may accomplish this by inhibiting the NF-B pathway and decreasing the NLRP3 inflammasome, as well as other signaling pathways. D-xylosyl is the predominant form of xylose in the human body. After a series of derivations, xylose can produce the small molecule xylose-binding lectin 3 which, when expressed in cells, can regulate cell proliferation, differentiation, and migration. If secreted outside the cell, it can bind directly to lipopolysaccharide, inhibit the production of pro-inflammatory cytokines, and exert an anti-inflammatory effect. In conclusion, the therapeutic effect of LBP on corneal injury may result from its monosaccharide components’ regulation of cell proliferation, apoptosis, antioxidant, and anti-inflammatory effects.
Through KEGG pathway enrichment analysis of the six primary target genes with degrees greater than 20, we discovered that a total of 27 signaling pathways, including IL-17, HIF-1, TNF, and MAPK, were enriched, with the IL-17 signaling pathway being the most significant. Significant enrichment of core target genes, including JUN, CASP3, and MMP9, was observed along this pathway. In distinct cell types, JUN plays different roles in inducing and inhibiting apoptosis [
20]. CASP3, also known as caspase-3, is one of the key mediators of cell apoptosis, and its activation can ultimately result in apoptosis [
21]. MMP9, or matrix metalloproteinase-9, is the predominant matrix metalloproteinase synthesized and secreted by basal corneal epithelial cells. After corneal injury, its expression increases, and it predominantly participates in corneal basement membrane degradation and ECM remodeling. It inhibits corneal epithelial regeneration and promotes pathological ulceration and perforation [
22]. The “component-target” network demonstrates that these three core genes interact with each of LBP’s ten monosaccharide components. The TNF signaling pathway is also enriched for these three essential genes. The IL-17 and TNF signaling pathways are classical inflammatory pathways that mediate diverse reactions, such as inflammation, apoptosis, and extracellular matrix remodeling, which are essential for corneal injury repair. Inducing numerous inflammatory and chemotactic factors, the IL-17 signaling pathway can mediate a series of inflammatory responses and tissue remodeling. The TNF signaling pathway not only mediates inflammatory responses and ECM remodeling, but also promotes cell apoptosis [
23]. Experiments in vitro have demonstrated that IL17 and TNF are highly expressed in corneal epithelial cells under conditions of elevated osmotic stress [
24]. Du [
1] discovered that LBP can inhibit UVB-induced HCEC apoptosis by inhibiting caspase-3 upregulation. Wong [
17] pretreated mouse corneas with 2 mg/mL of LBP for 7 days before causing corneal injury with NaOH. The findings demonstrated that LBP intervention could promote corneal epithelial growth and reduce collagen structural damage following injury. Through in vitro cell experiments, this group [
4] also demonstrated that LBP could reduce pro-fibrotic proteins and pro-inflammatory cytokines in corneal injury and reduce fibrotic scarring. In conclusion, the ten monosaccharide components of LBP may exert their therapeutic effects on corneal injury via the IL17 and TNF signaling pathways mediated by core genes such as JUN, CASP3, and MMP9, influencing inflammation, apoptosis, and extracellular matrix remodeling responses.
Prior to the clinical implementation of a new drug or drug formulation, it is essential to investigate its toxicity. Typically, drug-induced toxicity in the body begins with functional changes in the organism which, if severe, can result in microscopic morphological changes. In a mouse model of dry eye disease (DED), Qin [
25] confirmed the safety and efficacy of LBP eye drops. None of the concentrations of LBP eye drops utilized demonstrated significant eye irritation. In our study, we observed rodents blinking their eyes after receiving LBP eye drops, but this behavior was also observed in the saline group, most likely due to the lower storage temperature of the medication. In addition, this reaction resolved rapidly, and after 14 days of LBP eye drops, there were no statistically significant differences between the two groups’ eye irritation response scores. The results of Hematoxylin and Eosin (HE) staining demonstrated that there were no anomalies in the corneal epithelial cells of either group of rats, and that the stromal fiber arrangement was regular and devoid of inflammatory cell infiltration. These findings demonstrate that the prepared LBP eye drops are safe for short-term, local use in ocular experiments, establishing the groundwork for future research. Numerous studies have demonstrated the protective effect of LBP against corneal injury. LBP intervention can substantially improve the clinical symptoms of dry eye syndrome in mice [
17], promote corneal epithelial growth, and cell experiments corroborate that LBP can reduce pro-inflammatory cytokines [
4,
17]. LBP can also prevent UVB-induced apoptosis in rat corneal epithelial cells [
1]. In this section of the study, we discovered that LBP treatment stimulated corneal epithelial growth after alkaline injury, decreased corneal opacity, and decreased the infiltration of inflammatory cells into the corneal stroma. In the alkali burn model, the restoration of the corneal stromal layer structure following LBP intervention was closer to that of the control group than that of the Dex (dexamethasone) group. However, Dex treatment resulted in a greater increase in cell density in the corneal epithelial layer than LBP treatment. The results of TUNEL immunofluorescence staining demonstrated that both LBP and Dex interventions decreased the number of apoptotic cells. Under typical conditions, the collagen fibers in the corneal stromal layer are arranged and distributed in a uniform manner. Chemical injuries to the cornea can result in the loss of corneal epithelial layers, stromal edema [
26], excessive deposition of the ECM, and decreased degradation, leading to late-stage infection and the formation of fibrotic scars. This can impair a patient’s eyesight. α-SMA, FN, Col I, and Col III are essential proteins in fibrosis. Immunofluorescence staining results indicated that both LBP and Dex could inhibit the expression of important corneal fibrotic proteins following alkali injury. These results corroborate that LBP has a protective effect on corneal injury and can improve the degree of fibrosis during the corneal injury repair process. LBP’s therapeutic effect is not substantially different from that of Dex, and it overcomes Dex’s disadvantage of a short application period, allowing for a longer application period and greater efficacy.
Previous research [
1] has demonstrated that HCEC treated with 0.05–1 mg/mL LBP can prevent the UVB-induced decrease in cell viability. In our study, LBP concentrations between 0.05 mg/mL and 1 mg/mL inhibited H
2O
2-induced cell injury and promoted cell proliferation in HCEC cells. The maximum cell activity was observed in the 0.2 mg/mL LBP group. H
2O
2 inhibited HCEC proliferation and migration, which resulted in a cell cycle arrest in the G2/M phase and a decrease in the G0/G1 phase, which inhibited cell division and decreased cell proliferation. After LBP intervention, however, the G2/M phase cell cycle arrest caused by oxidative stress-induced HCEC damage was reduced, promoting cell division and transition from G2/M to G1/G0 phase, facilitating cell proliferation and migration, and reducing cell damage, thereby restoring normal physiological function. These studies indicate that LBP in the appropriate concentration range has no substantial impact or toxic side effects on the cells themselves, and can enhance epithelial cell proliferation and migration following corneal epithelial injury. It is anticipated to be a safe and efficacious corneal ophthalmic formulation. The detection of reactive oxygen species (ROS) revealed a significant increase in intracellular ROS levels as a result of oxidative stress damage, placing HCEC in a metabolic state of imbalance with ROS accumulation exceeding clearance. ROS is intimately associated with the mitochondrial pathway of apoptosis, where mitochondria are the primary source of ROS production [
27]. The accumulation of ROS can alter mitochondrial membrane permeability, causing apoptotic factors such as Cytochrome C to be released from mitochondria into the cytoplasm, thereby initiating the caspase cascade and activating the final apoptotic executor caspase-3 [
28], resulting in an increase in cell apoptosis [
29,
30], confirming this theory. Flow cytometry apoptosis assay results confirmed that oxidative stress injury increased the number of apoptotic HCEC significantly. LBP intervention ameliorated oxidative stress-induced HCEC injury by decreasing ROS levels in the cells, thereby reducing HCEC cell apoptosis. Oxidative stress is associated with inflammation [
31,
32,
33]; for example, in dry eye syndrome, oxidative stress resulting from an imbalance of reactive oxygen species (ROS) is the primary cause, and inflammatory reactions are a major source of ROS [
34,
35]. Related research has confirmed the relationship between oxidative stress and inflammation [
36,
37], and certain types of inflammation can contribute to the progression of disease through the excessive production of reactive oxygen species (ROS) and other oxidants, and a decrease in antioxidant levels [
38]. In eye diseases, the decrease in lactoferrin levels in the tear film, an antioxidant that bonds with free iron to reduce ROS production, causes an increase in infections and inflammatory responses. Overproduction of reactive oxygen species (ROS) in eye surface cells can activate inflammatory signaling pathways to promote inflammation, leading to an increase in pro-oxidant-sensitive inflammatory cytokines such as TNF-α, IL-6, IL-8, etc. [
39]. In HCEC, oxidative stress substantially increased the levels of reactive oxygen species (ROS) and the expression of inflammatory factors IL6 and TNF-α, according to our research. However, after LBP intervention, both ROS levels and IL6 and TNF-α gene expression were significantly reduced. This suggests that LBP can inhibit the production of reactive oxygen species (ROS) in HCEC, thereby reducing the inflammatory response.
In summary, by examining the gene expression levels of the predicted core targets (JUN, CASP3, and MMP9) in the rat cornea and HCEC after LBP intervention, we found that LBP treatment significantly reduces the mRNA levels of JUN, CASP3, and MMP9 in both in vivo and in vitro corneal injury models. These three factors are all enriched in the TNF and IL17 signaling pathways. Therefore, we hypothesize that LBP may mitigate corneal inflammation, cell apoptosis, and extracellular matrix remodeling after corneal injury by inhibiting the relevant signaling pathways mediated by JUN, CASP3, and MMP9.
4. Materials and Methods
The databases and software used for network pharmacology prediction analysis are detailed in
Supplementary Table S1.
4.1. Screening Targets for LBP-Mediated Corneal Injury Repair and Constructing Protein–Protein Interaction (PPI) Network
The monosaccharide composition information of LBP was obtained from cutting-edge research. Through the TCMSP platform, we searched for the chemical structures, oral bioavailability (OB), and drug-likeness (DL) of individual monosaccharide components of LBP. Furthermore, we retrieved the linear notations of each monosaccharide component from the PubChem database, and predicted the relevant targets of LBP based on these linear notations in the SwissTarget database. Using five databases (GeneCards, OMIM, DrugBank, PharmGKB, and DisGeNET), we conducted keyword searches with terms such as “corneal fibrosis, corneal injury, corneal scar” to obtain genes related to corneal injury diseases. The intersection of disease-related targets and LBP active ingredient targets was obtained using Venn diagram online software (
http://jvenn.toulouse.inra.fr/app/example.html, accessed on 29 August 2023) to identify potential targets for LBP in the treatment of corneal injury diseases. The PPI network diagram was created using the STRING 11.5 online database. The topological parameters of the network were calculated using Cytoscape (Version 3.7.2) software, and targets with high degree values were selected for subsequent analysis.
4.2. Construction and Analysis of “Drug Components—Key Targets” Network
LBP monosaccharide components, intersecting targets, and other data were imported into Cytoscape software (Version 3.7.2) for visual analysis and network creation. Network topology analysis tools were used to calculate the degree of each node, which represents the number of nodes directly connected to it. A higher degree value indicates a more critical role of that node in the network.
4.3. KEGG Pathway and GO Function Enrichment Analysis
Enrichment analysis of the obtained key targets was conducted using the Metascape database. GO analysis was performed in three modules: biological processes (BP), molecular functions (MF), and cellular components (CC), to understand the biological processes, molecular functions, and cellular components involved with the target genes in vivo. KEGG pathway enrichment analysis was carried out to identify the primary signaling pathways associated with LBP’s treatment of corneal injury diseases, with a significance level set at p < 0.05. The results of both enrichment analyses were visualized as bar charts and bubble charts using a bioinformatics platform.
4.4. Animal Experiment Grouping and Topical Administration
LBP solution was prepared by accurately weighing LBP powder (SP9311, Solarbio, Beijing, China) in a clean bench and dissolving it in physiological saline to achieve a final concentration of 2 mg/mL. The solution was then sterilized by filtering through a 0.22 μm sterile micropore membrane and stored in a 4 °C refrigerator.
Healthy male SD rats provided by the Ningxia Medical University Animal Center were examined with a slit lamp to exclude any anterior segment lesions before being randomly assigned to experimental groups. SD rats were placed in 12 h light/12 h dark conditions, free access to food and water in a room controlled by temperature and humidity; rat feed (23103223, Keao Xieli Feed, Beijing, China) raw material composition: corn, soybean meal, fish meal, flour, yeast powder, vegetable oil, salt, vitamins, and mineral elements. A total of 48 rats were used in the experiment, and were divided into 6 groups with 8 rats in each group: (1) Control group (two groups in total): topical administration of saline. (2) LBP group: topical administration of 2 mg/mL LBP solution.(3) NaOH group: topical administration of saline after alkali burn model creation.(4) NaOH + LBP group: topical administration of 2 mg/mL LBP solution after alkali burn model creation. (5) NaOH + Dex group: topical administration of Dex (H20020497, qilu-pharma, Jinan, China) after alkali burn model creation.
The right eye of each rat was used for the experiment, and each group received the corresponding solution in their eyes, four times a day, for a total of 14 days, with each eye closure lasting for more than 10 s after eye drop administration. Schematic representation of the experimental design of the effectiveness of LBP treatment (
Figure 4A).
4.5. Construction of Rat Corneal Alkali Burn Model
Rats were anesthetized by intraperitoneal injection of a mixture of 90 mg/kg ketamine and 10 mg/kg xylazine (Alfamedic, Singapore). Local anesthesia was administered by instilling lidocaine hydrochloride on the corneal surface, and excess fluid in the conjunctival sac was wiped away with a cotton swab. A unilateral corneal alkali burn model [
40,
41,
42] was established by using a corneal trephine to create a circular filter paper with a diameter of 3 mm, which was then immersed in a 1M NaOH (A620617, Sangon Biotech, Shanghai, China) solution for 5 s. The filter paper was removed, excess alkali was blotted with filter paper, and it was gently applied to the central cornea of the right eye of the rat. After a 30 s burn, the burned area and the conjunctival sac were rinsed with physiological saline for 1 min. A circular gray-white burn spot, approximately 3 mm in diameter, was visible in the central cornea. The success criteria for the model were corneal stromal edema and obvious opacity, with the iris faintly visible.
4.6. Slit Lamp Examination
Before and after administration of the corresponding solution, slit lamp examination was conducted to observe and record the cornea, conjunctiva, and iris of the rats. Evaluation was performed based on the modified Draize eye irritation test scoring system, with the scores added up and multiplied by weighting factors to obtain weighted scores for cornea, iris, and conjunctiva. The corneal inflammation index for each group of rats was calculated by summing the scores and dividing by 9, as per the internationally recognized scoring standard [
43]. Corneal opacity in rats was scored according to the criteria described in relevant literature [
44].
4.7. Hematoxylin and Eosin (H&E) Staining
Whole rat eyeballs were fixed in 4% paraformaldehyde overnight at 4 °C. The tissues were dehydrated using an automated dehydrator and then rapidly embedded in a 60 °C paraffin oven. Paraffin blocks were sectioned into 4 μm thick paraffin sections using a rotary microtome, followed by HE staining. Neutral resin was used for mounting the slides. The corneal thickness and corneal cell density were counted and statistically analyzed using ImageJ software (Version 1.46r).
4.8. TUNEL Apoptosis Staining and Immunofluorescence Staining
Fixed rat corneal sections were incubated in 4% paraformaldehyde and permeabilized with 0.5% Triton X-100 at room temperature. After blocking with 5% BSA for 60 min, TUNEL apoptosis staining was performed by incubating the sections in TUNEL detection solution (C1090, Beyotime, Shanghai, China) in a light-protected environment for 1 h. Immunofluorescence staining involved incubating the sections with the respective primary antibody at 4 °C overnight, followed by incubation with fluorescent secondary antibody for 1 h. DAPI-containing anti-fluorescence quenching mounting medium (H-1200-10, Vector Laboratories, Newark, CA, USA) was used for nuclear staining. Observations and photography were carried out under a fluorescence microscope. The average fluorescence intensity (MFI) of apoptosis and fibrosis-related proteins was statistically analyzed using ImageJ software.
This section involves antibodies and reagents, including anti-α-SMA (AF1032-100, Affinity, San Francisco, CA, USA), anti-fibronectin (66042-1-Ig-100, Proteintech, Wuhan, China), anti-collagen I (66761-1-Ig-100, Proteintech, China), and anti-collagen III (22734-1-AP-100, Proteintech, CHN) antibodies, fluorescent secondary antibody (111-545-144, Jackson ImmunoResearch Inc., West Grove, PA, USA), and DAPI-containing anti-fluorescence quenching mounting medium(H-1200-10, Vector Laboratories, Newark, CA, USA).
4.9. Cell Culture and Reagents
HCEC (342430, BNCC, CHN) was cultured in EMEM medium supplemented with 10% fetal bovine serum (FBS, Gibco, Grand Island, NY, USA) and antibiotics (100 mg/mL streptomycin and 100 U/mL penicillin) at 37 °C in a 5% CO2 incubator. LBP powder was dissolved in pre-configured EMEM complete medium to a final concentration of 10 mg/mL. The solution was filtered and sterilized through a 0.22 μm sterile micropore membrane and stored at −20 °C. The mother solution was diluted with EMEM medium to the required concentration before use.
4.10. Cell Counting Kit-8 (CCK-8) Assay
Cells were seeded as a suspension (100 μL/well) in a 96-well plate. After 24 h in a cell culture incubator to allow cells to adhere to the well bottom, the old medium was removed, and cells were treated as per the experimental design. After a specified incubation period, 10 μL of CCK-8 reagent (K04, DOJINDO, Kumamoto, Japan) was added to each well. After incubating for 1.5 h, the absorbance (OD) at 450 nm was measured using a microplate reader to calculate cell viability.
4.11. Flow Cytometry
HCEC cells were seeded in 6-well plates at a density of 2 × 105 cells/well. Once cell confluence reached 70–80%, the cells were divided into control group, H2O2 group, and LBP group. The control group was cultured normally without any treatment, and the H2O2 group was treated with 603 μmol/L H2O2 (30%, Cas: 7722-84-1, Yantai Shuangshuang, Yantai, China) for 24 h to create an oxidative stress-damaged cell model. The LBP group received 0.2 mg/mL LBP and 603 μmol/L H2O2 co-treatment for 24 h. Cell ROS levels, cell cycle, and apoptosis rate were determined using the ROS detection kit (KGT010-1, KeyGEN, Nanjing, China), Annexin V-FITC/PI apoptosis detection kit (KGA107, KeyGEN, China), and cell cycle detection kit (KGA512, KeyGEN, China) following the manufacturer’s instructions. BD AccuriTM C6(Becton, Dickinson and Company, Franklin Lakes, NJ, USA) was used for the determination of cell ROS levels, cell cycle, and apoptosis rate.
4.12. Scratch Assay
Healthy HCEC cells were plated at a density of 2 × 105 cells/well in a 6-well plate. When cell confluence reached above 80%, a 10 μL pipette tip was used to create a “—” pattern scratch on the cell monolayer. The old culture medium was removed, and cells were treated according to the respective group. After 24 h, the changes in cell scratch were observed under an inverted microscope.
4.13. Real-Time Quantitative Polymerase Chain Reaction (qRT-PCR)
Total RNA was extracted from rat corneal and cell samples from each group using Trizol reagent (ThermoFisher, Waltham, MA, USA). The RNA was reverse-transcribed into cDNA using the PrimeScript™ RT reagent Kit with gDNA Eraser (Perfect Real Time) (RR047A, Takara, Shiga, Japan). TB Green
® Premix Ex Taq™ II (Tli RNaseH Plus) (RR820A, Takara, Japan) was used for subsequent experiments. PCR was carried out for 40 cycles using a fluorescence quantitative PCR instrument (StepOne Software V2.3, ThermoFisher, USA), with GAPDH as the internal reference gene. The relative gene expression of various genes was analyzed using the 2
−ΔΔCT method. Specific primer sequences are available in
Supplementary Table S2.
4.14. Data Analysis
Data from this study are presented as mean ± standard deviation ( ± s). Statistical analysis and graph plotting were performed using GraphPad Prism 9.0 software. The differences between groups were analyzed using t-tests or one-way analysis of variance (ANOVA) for homogenous variance, with p < 0.05 indicating statistically significant differences.