TLR3 Gene Regulates Cataract-Related Mechanisms Through The Jagged-1/Notch Signaling Pathway

DOI: https://doi.org/10.21203/rs.3.rs-1321658/v1

Abstract

Objective: Epithelial-mesenchymal transition (EMT) is the main cause of organ fibrosis and a common pathogenesis of most cataracts. Therefore, TLR3 may play a vital role in the progress of PCO EMT. This study aims to explore the molecular mechanism of TLR3 in the occurrence and development of post-cataract EMT, and to provide new ideas for the prevention and treatment of posterior capsule opacification (PCO).

Materials and Methods: The expression of TLR3 and EMT-related factors in samples from normal and PCO patients was measured by qPCR. In the presence or absence of TLR3, human lens epithelial cell line SRA01/04 are treated with TGF-β2. The CCK-8 method measures cell proliferation, and the Transwell method detects migration and invasion. The expression of EMT-related molecules and Jagged-1/Notch signaling pathway was analyzed by Western blot and qPC experiments.

Result: In terms of mechanism, the functional gain and loss experiments of human LEC show that TLR3 regulates the proliferation, migration, invasion of LEC and EMT induced by TGF-β2 by targeting the Jagged-1/Notch signaling pathway to affect the development of fibrotic cataract.

Conclusion: These findings indicate that TLR3 prevents the progression of lens fibrosis by targeting the Jagged-1/Notch signaling pathway to regulate LEC proliferation, migration, invasion and TGF-β2-induced EMT. "TLR3-Jagged-1/Notch" signal axis may be a therapeutic target for the treatment of fibrotic cataract.

Introduction

Fibrosis is a common cause of organ dysfunction and organ failure. The survival rate of some fibrotic diseases is even lower than that of cancer[1, 2]. With the development of fibrotic disease research, fibrosis models are constantly improving. Due to its unique biological characteristics, the lens has become an ideal biological tool for the study of fibrosis. The adult lens is separated from other tissues and has no nerve and blood vessel distribution, but draws nutrients from aqueous humor and vitreous humor. Lens epithelial cells are distributed under the anterior capsule of the lens and under the capsule of the equator of the lens. The lens epithelial cells proliferate, differentiate and migrate inward to form lens fibers. Under physiological conditions, the lens fibers are arranged neatly, but some pathological conditions lead to the destruction of the lens structure and lens fibrosis[3]. Although fibrotic diseases can affect many organs, such as liver fibrosis, kidney fibrosis, lung fibrosis and lens fibrosis, there are still some potential common pathogenesis mechanisms for fibrosis in different organs. In recent years, more and more evidence has shown that epithelial-mesenchymal transition (EMT) is the main cause of the progression of organ fibrosis[4], and lens EMT is also a common mechanism of most cataracts[5].

Cataract is a common eye disease that causes visual impairment and is the main cause of blindness. It is characterized by the gradual increase in lens opacity, obstructed vision, and gradual loss of vision[6, 7]. According to reports, 96% of people over 60 years old have varying degrees of lens opacities[8, 9]. Cataracts can be divided into anterior subcapsular cataract (ACS) and posterior capsular opacification (PCO) according to the location where fibrosis occurs. At present, surgery can effectively restore the vision of patients with cataracts, and may be the most commonly used and most effective treatment for cataracts. However, despite the continuous improvement of cataract surgery methods, postoperative complications such as posterior capsule opacity still severely affect the vision of patients[1013].

Excessive proliferation and migration of lens epithelial cells and transformation into mesenchymal cells (ie EMT) are common causes of cataracts. In the process of epithelial-mesenchymal transition, epithelial cells secrete excessive extracellular matrix, including collagen type Ⅰ (Col Ⅰ) and fibronectin (Fibronectin, FN), which downregulate E-cadherin (E-Cadherin, E-Cad) deprives cell polarity[14] and weakens intercellular adhesion, which ultimately leads to changes in the phenotype of epithelial cells, which synthesize α-smooth muscle actin (α-SMA), Vimentin, etc. to obtain the interstitial cell phenotype[15, 16].

TLR3 is an important member of the TLRs family. It is a type I transmembrane protein and can recognize double-stranded RNA viruses and Poly I: C. Existing studies have shown that TLR3 has a dual role in tumors. On the one hand, most studies reported that the activation of TLR3 has an inhibitory effect on tumors and can promote tumor cell apoptosis[17], which may be mainly related to the production of type I interferon and natural killer cells, and the activation of immune cells such as natural killer cells, dendritic cells, and macrophages[18, 19], so TLR3 agonists can be used as immune adjuvants to treat tumors. On the other hand, some studies have shown that TLR3 can promote tumor progression. The prognosis of breast cancer patients with high TLR3 expression is relatively poor[20], which may be because the activation of TLR3 triggers the β-catenin and NF-κB signaling pathways, which promotes the transformation of breast cancer cells into cancer stem cells[21]. However, the mechanism of TLR3 in cataract fibrosis is rarely reported.

The Notch signaling pathway is evolutionary and conservative. It controls many cellular processes through the interaction between neighboring cells and the interaction with other signals, including cell differentiation, proliferation, tumor angiogenesis, maintenance of stemness, and apoptosis[22]. Among them, the Notch receptor family is a transmembrane protein, a heterodimer composed of an extracellular domain and an intracellular domain[22]. The ligand Jagged1 is a single-pass transmembrane glycoprotein with a small intracellular domain and a larger extracellular domain. It contains four parts necessary for the normal function of the protein: N-signal peptide, DSL domain, and 16 EGF-like Repeat region and 1 cysteine-rich region. The Notch signaling pathway, especially the ligand Jagged1, is closely related to the occurrence and biological behavior of a variety of tumors, such as tumor angiogenesis, maintenance of stem cell characteristics, and metastasis[23]. At present, the specific role and mechanism of Jagged-1/Notch signal in cataract are not yet clear.

In this study, we identified TLR3 as a key regulator to prevent lens fibrosis. We have shown that TLR3 is significantly elevated in PCO cataracts, and TGF-β2 significantly up-regulates TLR3 expression in fibrotic human lens epithelial cell line SRA01/04. Importantly, by showing the effect of TLR3 upregulation on LEC proliferation, migration, invasion and EMT, we revealed a new mechanism of TGF-β2 induced lens fibrosis by acting on the Jagged-1/Notch signaling pathway.

Methods And Materials

Cell culture

In the study, the fresh postmortem human lenses were used which were collected from the donors dead within 8 hours (20 males and 8 females). The fibrotic lens part came from the PCO tissues of 4 donors who had undergone cataract surgery before death. The fibrotic part of the posterior capsule was separated with capsulotomy scissors. For normal transparent lens, use capsulotomy scissors to cut the anterior capsule with epithelium along the equatorial region of the lens, and peel it from the lens fibers with capsulorhexis to collect the lens epithelial explant. The human lens epithelial cell line SRA 01/04 was purchased from ATCC and cultured in Dulbecco's modified Eagle medium (DMEM, Thermo Fisher Scientific) containing 8% FBS. The lens fibrosis model induced by TGF-β2 is made by treating the LEC cell line SRA01/04 with 5 ng/mL TGF-β2 (302-B2; R&D) for 48-72 hours.

Transfection

The si-NC and si-TLR3 purchased from GenePharma (Shanghai, China) were transfected into LEC cell line SRA01/04 by Lipofectamine 2000 (Invitrogen, CA, USA). The pcDNA 3.1-TLR3 or pcDNA 3.1-NC adenovirus vector was transfected from HanBio Technology Co. Ltd. (Shanghai, China) into human LECs for overexpression of TLR3 or as a control for the following experiments.

CCK-8 assay

The cells were digested into cell suspension with a density of 2 × 104 cells/mL. 100mL of cell suspension was added into a 96-well plate, then placed in a 37°C, 5% CO2 incubator to culture adherence. After the cells are processed, 10µL of CCK-8 reagent was added into each well and continue to be incubated for 2 hours. Microplate reader was used to detect the OD value at 450 nm for statistical analysis of the data.

Migration and invasion experiment

Migration experiment: adjust the processed cells to 2 × 105 cells/ml, take the pre-prepared Matrigel-free Transwell chamber and place it in a 24-well plate, LEC cells were added in the upper chamber, and 500 µl of 10% serum-containing was added in the lower chamber DMEM medium for 24 h. The chamber was taken out and the upper chamber liquid was discarded. After washed with PBS, the cells were fixed with formaldehyde. The upper cells of the chamber were wiped with a cotton swab. Fix with formaldehyde for 10-15 minutes and stain with crystal violet for 20 minutes. The migrated cells are counted under a microscope.

Invasion experiment: Take 50 µl of Matrigel (Corning) and spread it in an 8 µm Transwell chamber, and place it in a 37 ℃ incubator for 1 h. The logarithmic growth phase LEC cells were seeded into the upper chamber of 8 µm Transwell small wells, 200 µl per well. Add 500 µl of DMEM medium containing 10% serum to the lower chamber and culture for 24 h.

qRT-PCR

The treated LEC cells were collected, the total mRNA was extracted by the Trizol method, and operate according to the instructions of the reverse transcription kit to obtain cDNA. qRT-PCR is as follows: SYBR Premix Ex Taq (Bimake) 5 µl, upstream and downstream primers 1 µl each, cDNA 2 µl, ddH2O 2 µl. Reaction conditions: 95 ℃ 30 s pre-denaturation; 95 ℃ 5 s, 55 ℃ 30 s amplification 45 cycles. Statistical data uses 2-ΔΔCt to calculate the relative expression of the target gene.

Western blot

LEC cells were lysed with radioimmunoprecipitation assay buffer supplemented with protease inhibitors (P0013C; Beyotime). The liquid was aspirated and centrifuged to collect the protein for quantification. A sample of 30 µg of protein was taken and transferred to membrane after SDS-PAGE electrophoresis. Antibody ZO-1 (13663; CST), E-cad (3195; CST), N-cad (13116; CST), Vim (5741; CST), Jagged-1 (70109; CST), Notch-1 (3608; CST), Notch-2 (5732; CST), GAPDH (5174; CST) were diluted 1:1000 and incubated overnight at 4°C. HRP-labeled secondary antibody was added and incubated for 2 h at 37 ℃ in the dark, rinsed with PBS-T repeatedly, and finally added ECL reagent, the chemiluminescence instrument was used to detect the expression of the target protein.

Statistical analysis

The Student’s t-test or the one-way ANOVA was used to evaluate the difference between groups. A value of P < .05 was considered statistically significant.

Results

FN, α-SMA, Col I, Acan and TLR3 are up-regulated in fibrotic lens tissue

The transparent lens capsule and fibrotic lens capsule from healthy donors and fibrotic cataract patients were collected. Compared with normal lens epithelium, the expression levels of fibrotic lens epithelial mesenchymal markers α-SMA, ECM markers FN, Col I, and Acan increased significantly (Figure 1A), indicating that lens fibrosis involves hyperproliferation and EMT. At the same time, the gene expression level of TLR3 in the fibrotic lens epithelium was significantly increased (Figure 1B), which may be related to the development of lens fibrosis.

TLR3 is upregulated in LEC cell line SRA01/04

In order to further explore the role of TLR3 in lens fibrosis. TGF-β2 was used to stimulate human lens in vitro implants. The increase of α-SMA, FN, Col I and Acan in LEC cell line SRA01/04 treated with TGF-β2 indicated that the cells have morphological and molecular changes similar to fibrotic cataracts (Figure 2A). The expression of occlusion zone tissue (ZO1) and E-cadherin (E-cad) decreased, and the expression of N-cadherin and vimentin (Vim) increased, which transferred from the cell membrane to the cytoplasm, and transformed into mesenchymal-like, lost Epithelial features (Figure 2B). At the same time, the gene expression level of TLR3 was significantly increased in the LEC cell line SRA01/04 treated with TGF-β2 (Figure 2C).

TLR3 promotes the proliferation, migration, invasion and EMT of human lens epithelial cells

In order to study the function of TLR3 in lens fibrosis, we transfected human lens epithelial cells with si-TLR3 or overexpressed TLR3 (Figure 3A). In human lens epithelial cells treated with TGF-β2, knockdown of TLR3 prevented the expression of mesenchymal transition markers FN, α-SMA, Col I and Acan (Figure 3B), and reduced the cell proliferation rate (Figure 3B). 3C). After transfection with si-TLR3, the migration and invasion of human lens epithelial cells were significantly reduced (Figure 3D-E). At the same time, the results of the WB experiment showed that the epithelial markers ZO-1 and E-cad were retained, and the expression of N-cad and Vim protein was significantly reduced (Figure 3F). The above experimental results indicate that TLR3 has a functional effect on the proliferation, migration, invasion and EMT of human lens epithelial cells.

TLR3 acts directly on the Jagged-1/Notch signaling pathway

In order to confirm the potential role of the Jagged-1/Notch signaling pathway in the process of lens fibrosis, we used Western blot to detect Jagged-1/Notch signaling pathway related proteins in human lens epithelial cells treated with TGF-β2 and transfected with si-TLR3 and overexpressing TLR3. The results showed that compared with the control group, Jagged-1, Notch-1, and Notch-2 proteins in human lens epithelial cells treated with TGF-β2 were significantly increased (Figure 4A). After transfection of si-TLR3 and overexpression of TLR3, Jagged-1, Notch-1, and Notch-2 proteins in the overexpression TLR3 group remained elevated, and the protein expression of si-TLR3 group decreased significantly (Figure 4B). The experimental results show that TLR3 has a direct effect on the Jagged-1/Notch signaling pathway.

TLR3 promotes the proliferation, migration, invasion and EMT of human lens epithelial cells by directly acting on the Jagged-1/Notch signaling pathway

In order to study whether the Jagged-1/Notch signaling pathway mediates the biological function of TLR3 in the process of human lens fibrosis, we used both transfection si-TLR3 and TLR3 activator valproic acid (VA). The results of the qRT-PCR experiment showed that compared with the TGF-β2 group, the FN, α-SMA, Col I and E-cad genes in the si-TLR3 group were significantly reduced, while compared with the si-TLR3 group, the FN and α-SMA in the VA group, Col I and E-cad genes were relatively elevated (Figure 5A). The CCK8 experiment results showed that compared with the TGF-β2 group, the proliferation rate of the si-TLR3 group was significantly decreased, while the VA group was significantly increased compared with the si-TLR3 group (Figure 5B). Compared with the TGF-β2 group, the cell migration and invasion ability of the si-TLR3 group was significantly decreased, and the VA group was significantly improved compared with the si-TLR3 group (Figure 5C-D). The WB experiment results showed that compared with the TGF-β2 group, the ZO-1 and E-cad protein expressions in the si-TLR3 group increased, and the N-cad and Vim proteins decreased. Compared with the si-TLR3 group, the results were opposite in the VA group, compared with the TGF-β2 group, Jagged-1/Notch signaling pathway related proteins decreased significantly in the si-TLR3 group, and improved in the VA group compared with the si-TLR3 group (Figure 5E). The above results indicate that in the process of human lens epithelial cell fibrosis, TLR3 promotes the proliferation, migration, invasion and EMT of human lens epithelial cells by directly acting on the Jagged-1/Notch signaling pathway.

Discussion

PCO is a common fibrotic disease and the most common complication after cataract surgery. EMT is the common pathological basis of many fibrotic diseases such as PCO[4]. Understanding the pathological mechanism of EMT will not only help the prevention and treatment of PCO, but may also provide a new understanding of the occurrence and development of fibrotic diseases.

When trauma or surgical damage leads to the destruction of the lens structure, the remaining lens epithelial cells proliferate excessively and migrate to the posterior capsule, resulting in EMT, which changes the color of the lens or reduces the transparency, and ultimately leads to the occurrence of cataracts[3]. The typical characteristics of EMT include: E-cadherin, a key molecule that maintains cell polarity and intercellular adhesion, is degraded or lost function, and the abundance of tight junction proteins such as occludin and zonules occlusion 1 (ZO1) is decreased. The expression of keratin decreases. At the same time, the characteristic proteins of mesenchymal cells such as Fibronectin, α-SMA, Vimentin, N-Cadherin (N-Cadherin), and Col Ⅰ increase[24, 25]. The connections between cells are dissolved, the cytoskeleton is reconstructed, and the cell morphology is transformed into a mesenchymal-like multidendrite spindle shape, and cell migration and invasion capabilities are enhanced[26]. TGF-β2 is currently known as a potent inducer of EMT. TGF-β2 is not only involved in EMT during growth and development[27], but is also closely related to EMT during wound healing, fibrosis and cancer[28, 29]. In this study, the real-time fluorescent quantitative PCR test results showed that the transparent lens capsule and the fibrotic lens capsule were compared between healthy donors and fibrotic cataract patients, the expression levels of fibrotic lens epithelial mesenchymal marker α-SMA, ECM markers FN, Col I, and Acan were significantly increased. TGF-β2 was used to stimulate human lens in vitro implants. In the LEC cell line SRA01/04 treated with TGF-β2, α-SMA, FN, Col I and Acan increased, and the closed zone tissue (ZO1) and E-cadherin (The expression of E-cad) decreased, and the expression of N-cadherin and vimentin (Vim) increased, confirming that lens fibrosis involves hyperproliferation and EMT, and TGF-β2 can significantly promote the occurrence of EMT in lens epithelial cells.

The human TLR3 gene is located on chromosome 4, and its expression product TLR3 receptor protein can recognize double-stranded RNA associated with viral infection. Studies have shown that TLR3, as an innate immune receptor, plays a role in activating anti-inflammatory signaling pathways during injury and infection. Chronic liver alcohol accumulation can inhibit TLR3-dependent signaling pathways in NK cells during the late stage of liver fibrosis and alcoholic liver disease. The lack of TLR3-mediated killing of NK cells played an important role in accelerating the progression of the disease[30]. However, the expression and function of TLR3 in the lens have not been reported yet. In this study, we detected the expression of TLR3 in human lens epithelial cells. More importantly, we found that TLR3 was significantly increased in the mouse lens anterior capsule injury model, and in the EMT model of lens epithelial cells induced by TGF-β, 2 TLR3 also increased significantly, suggesting that TLR3 may be involved in the lens EMT process, but its role in the lens epithelial cell EMT process is still unclear.

In order to further study the role of TLR3 in EMT of lens epithelial cells, we constructed siRNA and TLR3 overexpression lentiviral vectors that specifically interfere with TLR3, aiming to observe differences in TLR3 expression on the proliferation, migration, invasion and EMT of lens epithelial cells cultured in vitro. To explore the regulatory effect of TLR3 on the EMT of lens epithelial cells. We found that under the stimulation of TGF-β2, the expression levels of α-SMA, Vimentin, and Col Ⅰ of cells transfected with si-TLR3 increased significantly, while the expression level of E-Cadherin decreased significantly, indicating that both groups of cells had EMT, but the expression of FN, α-SMA, Col I and Acan in the si-TLR3 group decreased, the proliferation rate decreased, and migration and invasion were significantly decreased. The results of WB experiments showed that the epithelial markers ZO-1 and E-cad were retained, and the expression of N-cad and Vim protein was significantly reduced, indicating that TLR3 has a functional effect on the proliferation, migration, invasion and EMT of human lens epithelial cells. The Notch signaling pathway is highly conserved in evolution and is involved in physiological processes such as cell proliferation, differentiation, apoptosis, and maintenance of cell stemness[31]. Notch ligands in mammals are also called DSL proteins. There are five types, namely Jagged1, Jagged2, DLL1, DLL3, and DLL4, all of which are type I transmembrane proteins. TGF-β2 can induce the expression of Notch ligands. In patients with diabetic nephropathy, the expression of Jagged1 and Hey1 induced by TGF-β2 plays a particularly important role, which may be related to the pathological process of the disease[32]. It has been confirmed that Jagged is the target gene of TGF-β2 in a variety of mammalian cells. TGF-β2 relies on Smad3 to regulate the expression of Jagged1 and Hey1 in the process of inducing EMT in cells[33]. In this study, Western blot results showed that Jagged-1, Notch-1, and Notch-2 proteins were significantly increased in human lens epithelial cells treated with TGF-β2. After transfection of si-TLR3 and overexpression of TLR3, Jagged-1, Notch-1, and Notch-2 proteins in the overexpression TLR3 group remained elevated, and the expression of si-TLR3 histone decreased significantly, indicating that TGF-β2 effectively activated Notch signal path. In order to study whether the Jagged-1/Notch signaling pathway mediates the biological function of TLR3 in the process of human lens fibrosis, transfection of si-TLR3 and the use of TLR3 activator valproic acid (VA) were used at the same time. The results showed that compared with the TGF-β2 group, the FN, α-SMA, Col I and E-cad genes in the si-TLR3 group were significantly decreased, the proliferation rate was significantly decreased, and the migration and invasion capabilities were significantly decreased; Compared with the si-TLR3 group, the FN, α-SMA, Col I, and E-cad genes were relatively higher in the VA group, and the proliferation rate increased significantly, and the migration and invasion capabilities increased significantly. The results of WB experiments showed that compared with the TGF-β2 group, the ZO-1 and E-cad protein expressions in the si-TLR3 group increased, and the N-cad and Vim proteins decreased. The VA group and the si-TLR3 group had the opposite results. Compared with the TGF-cβ2 group, Jagged-1/Notch signaling pathway related proteins decreased significantly in the si-TLR3 group, and improved in the VA group compared with the si-TLR3 group. It shows that in the process of human lens epithelial cell fibrosis, TLR3 promotes the proliferation, migration, invasion and EMT of human lens epithelial cells by directly acting on the Jagged-1/Notch signaling pathway.

Our results provide the first evidence that TLR3 and Jagged-1/Notch pathway play an important role in lens fibrosis. Our data indicate that the blockade of TLR3 and Jagged-1/Notch pathways may be a promising strategy for the prevention and treatment of organ fibrosis. These understandings of the regulatory relationship between TLR3 and Jagged-1/Notch signaling pathway are helpful to understand the pathogenesis of fibrotic diseases.

Declarations

Authors’ contributions 

Yu Chen and Rui Yang designed the research study. Yu Chen performed the research. Yu Chen wrote the manuscript. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript.

Data Availability 

The analyzed datasets generated during the study are available from the corresponding author on reasonable request.

Acknowledgments

The authors would like to acknowledge all of the participants for their technical and scholastic.

Declaration of competing interest

The authors claim that there is no conflict of interests.

Ethics statement

This study was conducted after being approved by the Ethics Committee of the Affiliated Hospital of North Sichuan Medical College.

References

  1. Rockey DC, Bell PD, Hill JA (2015) Fibrosis--a common pathway to organ injury and failure, N. Engl J Med 372(12):1138–1149. .https://www.ncbi.nlm.nih.gov/pubmed/25785971
  2. McAnulty RJ (2007) Fibroblasts and myofibroblasts: their source, function and role in disease. Int J Biochem Cell Biol 39(4):666–671. https://www.ncbi.nlm.nih.gov/pubmed/17196874
  3. Eldred JA, Dawes LJ, Wormstone IM (2011) The lens as a model for fibrotic disease. Philos Trans R Soc Lond B Biol Sci 366(1568):1301–1319. https://www.ncbi.nlm.nih.gov/pubmed/21402588
  4. Thiery JP, Acloque H, Huang RY, Nieto MA (2009) Epithelial-mesenchymal transitions in development and disease, Cell. 139:871–8905https://www.ncbi.nlm.nih.gov/pubmed/19945376
  5. Chen X, Xiao W, Chen W et al (2017) MicroRNA-26a and -26b inhibit lens fibrosis and cataract by negatively regulating Jagged-1/Notch signaling pathway. Cell Death Differ 24(8):1431–1442. https://www.ncbi.nlm.nih.gov/pubmed/28622289
  6. Bamdad S, Bolkheir A, Sedaghat MR, Motamed M (2018) Changes in corneal thickness and corneal endothelial cell density after phacoemulsification cataract surgery: a double-blind randomized trial. Electron Physician 10(4):6616–6623. https://www.ncbi.nlm.nih.gov/pubmed/29881523
  7. Gonzalez-Salinas R, Guarnieri A, Guirao Navarro MC, Saenz-de-Viteri M (2016) Patient considerations in cataract surgery - the role of combined therapy using phenylephrine and ketorolac. Patient Prefer Adherence 10:1795–1801. .https://www.ncbi.nlm.nih.gov/pubmed/27695298
  8. Wang W, Yan W, Fotis K et al (2016) Cataract Surgical Rate and Socioeconomics: A Global Study. Invest Ophthalmol Vis Sci 57(14):5872–5881. https://www.ncbi.nlm.nih.gov/pubmed/27802517
  9. Yan W, Wang W, van Wijngaarden P, Mueller A, He M (2019) Longitudinal changes in global cataract surgery rate inequality and associations with socioeconomic indices. Clin Exp Ophthalmol 47(4):453–460. .https://www.ncbi.nlm.nih.gov/pubmed/30362287
  10. Zukin LM, Pedler MG, Groman-Lupa S et al (2018) Aldose Reductase Inhibition Prevents Development of Posterior Capsular Opacification in an In Vivo Model of Cataract Surgery. Invest Ophthalmol Vis Sci 59(8):3591–3598. https://www.ncbi.nlm.nih.gov/pubmed/30025084
  11. Wei Z, Caty J, Whitson J et al (2017) Reduced Glutathione Level Promotes Epithelial-Mesenchymal Transition in Lens Epithelial Cells via a Wnt/beta-Catenin-Mediated Pathway: Relevance for Cataract Therapy. Am J Pathol 187(11):2399–2412. https://www.ncbi.nlm.nih.gov/pubmed/28827139
  12. Ngan K, Fraser E, Buller S, Buller A (2018) A cost minimisation analysis comparing iStent accompanying cataract surgery and selective laser trabeculoplasty versus topical glaucoma medications in a public healthcare setting in New Zealand. Graefes Arch Clin Exp Ophthalmol 256(11):2181–2189. https://www.ncbi.nlm.nih.gov/pubmed/30132278
  13. Jarstad JS, Jarstad AR, Chung GW, Tester RA, Day LE (2017) Immediate Postoperative Intraocular Pressure Adjustment Reduces Risk of Cystoid Macular Edema after Uncomplicated Micro Incision Coaxial Phacoemulsification Cataract Surgery. Korean J Ophthalmol 31(1):39–43. https://www.ncbi.nlm.nih.gov/pubmed/28243022
  14. Sun W, Liu J, Li J et al (2018) Human lens epithelial cell apoptosis and epithelial to mesenchymal transition in femtosecond laser-assisted cataract surgery. Int J Ophthalmol 11(3):401–407. https://www.ncbi.nlm.nih.gov/pubmed/29600173
  15. Marcantonio JM, Syam PP, Liu CS, Duncan G (2003) Epithelial transdifferentiation and cataract in the human lens. Exp Eye Res 77(3):339–346. .https://www.ncbi.nlm.nih.gov/pubmed/12907166
  16. Raghavan CT, Smuda M, Smith AJ et al (2016) AGEs in human lens capsule promote the TGFbeta2-mediated EMT of lens epithelial cells: implications for age-associated fibrosis. Aging Cell 15(3):465–476. https://www.ncbi.nlm.nih.gov/pubmed/26853893
  17. Salaun B, Coste I, Rissoan MC, Lebecque SJ, Renno T (2006) TLR3 can directly trigger apoptosis in human cancer cells. J Immunol 176(8):4894–4901. https://www.ncbi.nlm.nih.gov/pubmed/16585585
  18. Ebihara T, Azuma M, Oshiumi H et al (2015) Identification of a polyI:C-inducible membrane protein that participates in dendritic cell-mediated natural killer cell activation. J Exp Med 212(8):1337. https://www.ncbi.nlm.nih.gov/pubmed/26169938
  19. Shime H, Matsumoto M, Oshiumi H et al (2012) Toll-like receptor 3 signaling converts tumor-supporting myeloid cells to tumoricidal effectors. Proc Natl Acad Sci U S A 109(6):2066–2071. .https://www.ncbi.nlm.nih.gov/pubmed/22308357
  20. Gonzalez-Reyes S, Marin L, Gonzalez L et al (2010) Study of TLR3, TLR4 and TLR9 in breast carcinomas and their association with metastasis,BMC Cancer10(665.https://www.ncbi.nlm.nih.gov/pubmed/21129170
  21. Jia D, Yang W, Li L et al (2015) beta-Catenin and NF-kappaB co-activation triggered by TLR3 stimulation facilitates stem cell-like phenotypes in breast cancer. Cell Death Differ 22(2):298–310. .https://www.ncbi.nlm.nih.gov/pubmed/25257174
  22. Yao Y, Ni Y, Zhang J, Wang H, Shao S (2017) The role of Notch signaling in gastric carcinoma: molecular pathogenesis and novel therapeutic targets. Oncotarget 8(32):53839–53853. https://www.ncbi.nlm.nih.gov/pubmed/28881855
  23. Li D, Masiero M, Banham AH, Harris AL (2014) The notch ligand JAGGED1 as a target for anti-tumor therapy,Front Oncol4(254.https://www.ncbi.nlm.nih.gov/pubmed/25309874
  24. Singh S, Sadacharan S, Su S et al (2003) Overexpression of vimentin: role in the invasive phenotype in an androgen-independent model of prostate cancer. Cancer Res 63(9):2306–2311. https://www.ncbi.nlm.nih.gov/pubmed/12727854
  25. Das V, Bhattacharya S, Chikkaputtaiah C, Hazra S, Pal M (2019) The basics of epithelial-mesenchymal transition (EMT): A study from a structure, dynamics, and functional perspective,J Cell Physiolhttps://www.ncbi.nlm.nih.gov/pubmed/30723913
  26. Lamouille S, Xu J, Derynck R (2014) Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol 15(3):178–196. https://www.ncbi.nlm.nih.gov/pubmed/24556840
  27. Mercado-Pimentel ME, Runyan RB (2007) Multiple transforming growth factor-beta isoforms and receptors function during epithelial-mesenchymal cell transformation in the embryonic heart. Cells Tissues Organs 185(1–3):146–156. .https://www.ncbi.nlm.nih.gov/pubmed/17587820
  28. Schnaper HW, Hayashida T, Hubchak SC, Poncelet AC (2003) TGF-beta signal transduction and mesangial cell fibrogenesis. Am J Physiol Renal Physiol 284(2):F243–F252. .https://www.ncbi.nlm.nih.gov/pubmed/12529270
  29. Willis BC, Borok Z (2007) TGF-beta-induced EMT: mechanisms and implications for fibrotic lung disease. Am J Physiol Lung Cell Mol Physiol 293(3):L525–L534. https://www.ncbi.nlm.nih.gov/pubmed/17631612
  30. Jeong WI, Park O, Gao B (2008) Abrogation of the antifibrotic effects of natural killer cells/interferon-gamma contributes to alcohol acceleration of liver fibrosis. Gastroenterology 134(1):248–258. https://www.ncbi.nlm.nih.gov/pubmed/18166357
  31. Brzozowa M, Mielanczyk L, Michalski M et al (2013) Role of Notch signaling pathway in gastric cancer pathogenesis. Contemp Oncol (Pozn) 17(1):1–5. https://www.ncbi.nlm.nih.gov/pubmed/23788953
  32. Walsh DW, Roxburgh SA, McGettigan P et al (2008) Co-regulation of Gremlin and Notch signalling in diabetic nephropathy. Biochim Biophys Acta 1782(1):10–21. .https://www.ncbi.nlm.nih.gov/pubmed/17980714
  33. Zavadil J, Cermak L, Soto-Nieves N, Bottinger EP (2004) Integration of TGF-beta/Smad and Jagged1/Notch signalling in epithelial-to-mesenchymal transition. EMBO J 23(5):1155–1165. https://www.ncbi.nlm.nih.gov/pubmed/14976548