All‑trans‑retinoic acid modulates TGF‑β‑induced apoptosis, proliferation, migration and extracellular matrix synthesis of conjunctival fibroblasts by inhibiting PI3K/AKT signaling

  • Authors:
    • Lingling Liang
    • Xiaomei Wang
    • Yajuan Zheng
    • Yang Liu
  • View Affiliations

  • Published online on: July 18, 2019     https://doi.org/10.3892/mmr.2019.10507
  • Pages: 2929-2935
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Conjunctival fiber generation is implicated in a wide spectrum of ocular diseases. Conjunctival wound healing is characterized by inflammation followed by re‑epithelialization, synthesis of new extracellular matrix (ECM), wound contraction and subconjunctival scar formation. The primary cause for the failure of glaucoma filtration surgery results from the excessive scarring of the filtering bleb. All‑trans‑retinoic acid (ATRA), a derivative of vitamin A, is a potent regulator of ECM synthesis, growth and differentiation. Following a previous study, which revealed that ATRA could inhibit transforming growth factor‑β‑induced human conjunctival fibroblast (HConF)‑mediated collagen gel contraction, the present study aimed to investigate the effects of ATRA on HConF migration, apoptosis, proliferation and ECM synthesis. To achieve this, the present study used Transwell migration, wound healing and Cell Counting Kit‑8 assays, flow cytometry and western blot analysis. In addition, the present study aimed to elucidate the mechanism of ATRA in mediating resistance to conjunctival scar formation. ATRA treatment resulted in an increased level of HConF apoptosis, reduced proliferation and migration, decreased collagen I and fibronectin expression, and decreased phosphorylation of PI3K and AKT. Thus, the present study showed a role for ATRA in inhibiting HConF migration, proliferation and ECM synthesis, and in promoting HConF apoptosis through the inhibition of the PI3K/AKT signaling pathway.

Introduction

Glaucoma is the most common cause of irreversible blindness worldwide, accounting for ~15% of all cases and ≥500,000 new cases annually in both developed and developing countries (13). It is characterized by the development of a specific pattern of visual field loss and optic neuropathy. Patients are usually treated using filtration surgery which serves to reduce intraocular pressure in the eye (4). Other treatments include topical medication, laser treatment and surgical modalities. Of the treatments available, surgery is the most effective (5,6). Glaucoma filtration surgery may stimulate conjunctival fibroblast proliferation, migration, differentiation and promote ECM secretion, which are important processes in wound healing and scar formation (79). However, glaucoma filtration surgery is not always effective, and excessive scarring of the filtering bleb after surgery is a major problem following surgery.

Scar formation at the filtering bleb after glaucoma filtration surgery is a multifactorial process; human conjunctival fibroblasts (HConFs) play an essential role in subconjunctival wound healing (10). In addition, the cytokine transforming growth factor (TGF)-β is an important factor in regulating wound healing and scar formation (10). The binding of TGF-β to its heterodimeric receptor activates intracellular signaling cascades, including the canonical SMAD pathway and the mitogen-activated protein kinase (MAPK) pathway, and leads to fibrosis (11). The PI3K/AKT signaling pathway, which regulates survival, was also reported to function in regulating cell migration, proliferation and apoptosis (12,13). The PI3K/AKT pathway can be activated by TGF-β and also plays an important role in modulating ECM synthesis (14).

All-trans-retinoic acid (ATRA), a derivative of vitamin A, can inhibit TGF-β and inhibit fibrosis (15). In a previous study, it was reported that ATRA inhibited TGF-β-induced HConF-mediated collagen gel contraction via the SMAD signaling pathway (16). However, the effects of ATRA on TGF-β-induced HConF migration, proliferation and ECM synthesis remain unclear. In the present study, HConFs were cultured to investigate the effects of ATRA on the proliferation, migration, apoptosis and ECM synthesis in conjunctival fibroblasts. The aim of the present study was to improve understanding of the mechanism of action of ATRA. The mechanistic insights provided by this study may be applicable to preventing scar formation following glaucoma filtration surgery.

Materials and methods

Materials

ATRA and TGF-β (Sigma-Aldrich; Merck KGaA) were prepared as 0.4 and 50 ng/ml stock solutions in DMSO, respectively, and were stored below −20°C in the dark. ATRA and TGF-β were diluted to 10–200 µM and 1 ng/ml, respectively, in DMEM (Sigma-Aldrich; Merck KGaA) containing 10% FBS (Gibco; Thermo Fisher Scientific, Inc.) on the day of the experiment. The highest concentration of DMSO in the test solutions was 0.1%. To exclude the possibility that proliferation may be inhibited by DMSO, all cells were exposed to a final DMSO concentration of 0.1% in DMEM containing 10% FBS.

Cell culture

Primary HConFs (ScienCell Research Laboratories, Inc.) were cultured according to the manufacturer's instructions; three individual lots of HConFs were obtained. The HConFs were maintained and cultured at 37°C in fibroblast medium (ScienCell Research Laboratories, Inc.) containing 2% FBS, fibroblast growth supplement (ScienCell Research Laboratories, Inc.), 100 U/ml penicillin and 100 µg/ml streptomycin in a 5% CO2 humidified incubator. The HConFs were characterized by a spindle-shaped morphology and were positively stained with anti-fibronectin antibodies.

Cell proliferation assay

A HConF proliferation assay was performed as previously described (17). Briefly, HConF suspensions (0.5×104 cells/well) were cultured in fibroblast medium in a 96-well plate (200 µl/well) for 24 h at 37°C. ATRA (0.01–10 µM) was added to the fibroblast medium and the cells were cultured for a further 48 h at 37°C in the absence or presence of TGF-β (1 ng/ml), with 0.1% DMSO as the control. The concentrations of ATRA and TGF-β used were determined according to a previous study (15). Subsequently, each well was incubated with 10 µl Cell Counting Kit-8 solution (BestBio; Sigma) for a further 3 h at 37°C. The absorbance was measured at 450 nm using an automated microplate reader (model 3001–1387; Thermo Fisher Scientific, Inc.).

Apoptosis assay

HConF apoptosis was determined using an Annexin V-FITC apoptosis kit (BestBio) as previously described (17). Briefly, the HConFs (5×105 cells/well) were plated in 6-well plates and cultured for 48 h. After rinsing twice with PBS, the cells were resuspended in 400 µl 1X binding buffer [10 mM HEPES, 140 mM sodium chloride, 2.5 mM calcium chloride (pH 7.4)], into which 5 µl of Annexin V-FITC was added. After 15 min incubation at 2–8°C in the dark, 10 µl of propidium iodide (BestBio) was added to the cells prior to further incubation for 5 min at 2–8°C in the dark. Within 15 min after staining, the cells were analyzed using a flow cytometer (Cytomics FV 500) and CytExpert 2.0 software (both Beckman Coulter, Inc.). The apoptotic rate was calculated as the sum of the percentage of early + late apoptotic cells.

Wound healing assay

HConFs were seeded at a density of 5×105 cells/well in 6-well plates. The culture medium for the HConFs was replaced after 24 h with serum-free medium, and then cells were incubated for a further 2 h at 37°C. Next, a sterile 20-µl pipette tip was used to scratch a line in the confluent cell monolayer, after which the cells were washed three times with PBS. The scratch wound healed after 24 h of incubation in the following conditions: 1 µM ATRA + 1 ng/ml TGF-β, 1 ng/ml TGF-β and fibroblast medium without ATRA and TGF-β. The effects on HConF migration were evaluated by measuring the area of the wound from the images captured at a magnification of ×100 at 0 and 24 h using a light microscope (Olympus Corporation); analysis was performed using ImageJ version 1.5 (National Institutes of Health).

Transwell migration assay

A chemomigration assay was performed using Transwell plates (pore size, 8 µm) as previously described (18). Briefly, the cells in the upper Transwell chamber (1×105) were suspended in 200 µl of DMEM, and DMEM containing 20% FBS was added to the lower chamber. Following a 12-h incubation at 37°C, the medium with non-migrated cells in the upper chamber was removed with a cotton swab. The cells that had migrated to the lower chamber were fixed with 4% paraformaldehyde for 30 min at 37°C and then stained with 0.5% crystal violet for 10 min at 37°C. The cells were counted at ×200 magnification using a light microscope in six different fields of view.

Cell treatment

HConFs were incubated in fibroblast medium in 6-well plates for 24 h. Then, the medium was replaced with the serum-free medium. Serum-starved cells were incubated for 6 h in the presence or absence of ATRA (1 µM), and then for 48 h in the presence or absence of TGF-β (1 ng/ml). Then, cell lysates were analyzed via western blotting.

Western blot analysis

Western blot analysis was performed as previously described (17,19,20). Cells were washed with pre-cooled PBS and lysed with RIPA buffer (Nanjing KeyGen Biotech Co., Ltd.) containing the protease inhibitor phenylmethylsulfonyl fluoride and the phosphatase inhibitor sodium orthovanadate (Nanjing KeyGen Biotech Co., Ltd.) on ice for 30 min. Subsequently, the cells were gently scraped from the plate and centrifuged at 1,500 × g for 12 min at 4°C. Protein concentrations were measured using the bicinchoninic acid method. A total of 20 µg/sample of protein was separated via 6–15% SDS-PAGE and then transferred onto PVDF membranes. Membranes were blocked with 5% non-fat dry milk for 1 h at 37°C before overnight incubation at 4°C with mouse monoclonal anti-collagen I (1:2,500; cat. no. ab88147; Abcam), rabbit polyclonal anti-fibronectin (1:1,500; cat. no. ab137720; Abcam), mouse antibodies against PI3K p85α (1:500; cat. no. sc-1637), phosphorylated (p)-PI3K p85α (1:500; cat. no. sc-12929), AKT (1:500; cat. no. sc-5298) and p-AKT (1:500; cat. no. sc-293125; all from Santa Cruz Biotechnology, Inc.), and mouse monoclonal anti-GAPDH (1:1,000; cat. no. A01020; Abbkine Scientific Co., Ltd.). Each membrane was incubated with anti-rabbit (1:5,000; cat. no. A0208) or anti-mouse (1:5,000; cat. no. A0216) horseradish peroxidase-conjugated secondary antibodies (both from Beyotime Institute of Biotechnology) for 1 h at room temperature. The blots were visualized using ECL reagent (Beyotime Institute of Biotechnology) and the bands were analyzed using ImageJ software.

Inhibition of TGF-β-induced collagen I and fibronectin expression in HConFs using a PI3K inhibitor

HConFs were incubated in fibroblast medium in 6-well plates for 24 h, then the medium was replaced with serum-free medium. Serum-starved cells were incubated with 10 µM LY294002 (Calbiochem; Merck KGaA) for 1 h at 37°C. Subsequently, the cells were treated with 1 ng/ml TGF-β for 48 h at 37°C. The effects of the PI3K inhibitor on the TGF-β-induced expression of collagen I and fibronectin in HConF were investigated via western blot analysis.

Statistical analysis

Data analysis was performed using SPSS version 19.0 (IBM Corp.). The data are presented as the mean ± SD. Each experiment was repeated at least three times. Statistical analysis was performed using Student's t-test or one-way ANOVA; following ANOVA, the least significant difference test was used for pairwise comparisons between groups. P<0.05 was considered to indicate a statistically significant difference.

Results

ATRA inhibits TGF-β-induced HConF proliferation

The effect of ATRA on HConF proliferation was examined. The cells were incubated for 48 h with 0.01–10 µM ATRA in the absence or presence of 1 ng/ml TGF-β with 0.1% DMSO as the control. ATRA was revealed to inhibit HConF proliferation in a concentration-dependent manner (P<0.05; Fig. 1A). HConFs were divided into three treatment groups: The ATRA group (1 µM ATRA + 1 ng/ml TGF-β), the TGF-β group (1 ng/ml TGF-β) and the control group (0.1% DMSO). ATRA inhibited TGF-β-induced HConF proliferation by 77.50±1.88% (P<0.01 vs. TGF-β; Fig. 1B).

ATRA induces HConF apoptosis

The effect of ATRA on HConF apoptosis was examined using flow cytometry. A significantly increased number of apoptotic HConFs were observed in the ATRA group compared with the control group (53.25±1.2 vs. 22.5±1.1%, respectively; P<0.01). The TGF-β group showed a significantly decreased apoptotic rate compared with the control group (14.75±1.4%; P<0.01; Fig. 2).

ATRA inhibits TGF-β-induced cell migration

The migratory ability of the HConFs was evaluated using in vitro wound healing and Transwell assays. Similar results were observed for the two assays. The ATRA group exhibited a ~30% reduction in wound healing compared with the control group (48.9±1.34 vs. 71.30±1.55%, respectively; P<0.05). Wound healing was increased in the TGF-β group compared with the control group (90.50±1.22%; P<0.05; Fig. 3A). Reduced HConF migration was observed for the ATRA group (14.85±1.13 cells; P<0.01), whereas significantly increased migration was noted for the TGF-β group compared with the control group (135.55±1.12 cells vs. 28.65±1.02 cells, respectively; P<0.01; Fig. 3B) in the Transwell migration assays.

ATRA inhibits TGF-β-induced collagen I and fibronectin expression

The effect of ATRA on TGF-β-induced ECM production was investigated. Western blotting analysis and densitometric analysis revealed that exposure of HConFs to 1 ng/ml TGF-β for 48 h induced the production of collagen I and fibronectin, whereas 1 µM ATRA inhibited TGF-β-induced synthesis of collagen I and fibronectin by HConFs (P<0.01; Fig. 4).

ATRA inhibits the PI3K/AKT pathway

The effect of ATRA on TGF-β-induced PI3K and AKT phosphorylation in HConFs was examined (Fig. 5). Western blotting analysis and densitometric analysis revealed that the exposure of HConFs to 1 ng/ml TGF-β for 48 h induced the phosphorylation/activation of PI3K and AKT. Treatment with 1 µM ATRA inhibited TGF-β-induced PI3K and AKT phosphorylation, indicating that ATRA inhibited the TGF-β-induced PI3K/AKT signaling pathway (P<0.05; Fig. 5).

Inhibition of TGF-β-induced collagen I and fibronectin expression in HConFs by a PI3K inhibitor

The effect of the PI3K inhibitor LY294002 on the TGF-β-induced expression of collagen I and fibronectin in HConF was investigated. Cells were incubated with 10 µM LY294002 for 1 h before treatment with 1 ng/ml TGF-β for 48 h. Western blotting analysis and densitometric analysis revealed that LY294002 significantly inhibited TGF-β-induced expression of collagen I and fibronectin in HConFs (P<0.05; Fig. 6). The TGF-β-induced expression of collagen I and fibronectin in HConFs was inhibited by 27 and 47% in the presence of LY294002, respectively. The inhibitor had no effect on collagen I and fibronectin expression in the absence of TGF-β (data not shown).

Discussion

In the present study, it was revealed that ATRA inhibited TGF-β-induced migration, proliferation and ECM synthesis in HConFs. Furthermore, ATRA was found to promote apoptosis and inhibit the TGF-β-induced phosphorylation of PI3K and AKT in HConFs. In addition, the present study showed that the PI3K inhibitor LY294002 attenuated the TGF-β-induced expression of collagen I and fibronectin.

Preventing conjunctival scarring remains a challenge in clinical ophthalmology. TGF-β is a key factor implicated in postoperative scarring as it stimulates the migration, proliferation and differentiation HConFs, and promotes ECM deposition and remodeling (21). ATRA, a derivative of vitamin A, has been shown to regulate ECM expression and serve an important role in fibrotic diseases through the inhibition of TGF-β1 (22). Moreover, ATRA was found to serve a protective role against collagen accumulation, cell injury and proliferation in various types of fibrosis, including liver fibrosis, pulmonary fibrosis and kidney fibrosis (2225). In a previous study, it was demonstrated that ATRA inhibited TGF-β-induced HConF-mediated collagen gel contraction by attenuating the formation of actin stress fibers and focal adhesions, as well as by inhibiting MAPK, c-Jun and SMAD signaling (16). In the present study, it was demonstrated that ATRA inhibited the TGF-β-induced migration and proliferation of HConFs, inhibited ECM synthesis and increased the apoptosis in HConFs. Collectively, these results suggested that ATRA attenuates conjunctival scarring by modulating the function of HConFs and ECM synthesis.

The PI3K/AKT pathway modulates cell proliferation, differentiation, apoptosis, motility, survival and glucose metabolism (26). PI3K generates 3′-phosphorylated phosphoinositides, including phosphatidylinositol-3,4-bisphosphate and phosphatidylinositol-3,4,5-triphosphate, which then recruit target proteins to the plasma membrane (27). AKT is a serine/threonine kinase that acts as an effector of PI3K (27). It has been reported that activation of the PI3K/AKT signaling pathway may induce ECM secretion in several cell types (28,29). LY294002 and AKT small interfering RNA were reported to significantly reduce TGF-β1-induced α-smooth muscle actin expression, a marker of fibroblasts, in conjunctival fibroblasts, indicating that these changes were mediated by the PI3K/AKT pathway (30). In addition, LY294002 could inhibit the proliferation and migration of conjunctival fibroblasts (31,32). In the present study, it was shown that ATRA promoted apoptosis and inhibited proliferation, migration and ECM synthesis. In addition, it was demonstrated that LY294002 could inhibit TGF-β-induced expression of collagen I and fibronectin, similar to ATRA; this is consistent with the results of a previous study (33). Hence, it was deduced that the inhibitory effects of ATRA on HConFs are likely mediated by inhibition of the PI3K/AKT signaling pathway.

TGF-β-mediated signaling can occur via SMAD-dependent or SMAD-independent pathways; the SMAD-independent pathway includes the MAPK and PI3K/AKT pathways (34). TGF-β activates the SMAD pathway via the phosphorylation of Smad2 and Smad3, which then leads to the formation of a Smad2/3 complex; the Smad2/3 complex and Smad4 are subsequently translocated to the nucleus in order to regulate the expression of genes associated with fibroblast proliferation, migration and ECM deposition (35). In our previous study, a role for SMAD signaling was found in the ATRA-mediated inhibition of TGF-β-induced, HConF-mediated collagen gel contraction (16). Therefore, it was hypothesized that the SMAD signaling pathway is also involved in the inhibitory effects of ATRA on proliferation, migration and ECM synthesis in HConFs.

In conclusion, the findings of the present study revealed the mechanism of action of ATRA. ATRA was found to modulate PI3K/AKT signaling and impact on TGF-β-induced apoptosis, proliferation, migration and ECM synthesis. Given the significant global impact of glaucoma and the inadequacy in its treatment methods, conjunctival scarring at the surgical site after glaucoma filtration surgery (36,37) and the adverse side effects of anti-scarring anti-metabolite medication, such as mitomycin-C and 5-fluorouracil, which can potentially lead to blindness (3841), there is a requirement for improved therapeutic approaches. From the present study, it is proposed that ATRA may be a promising compound capable of modulating scar formation after glaucoma filtration surgery. However, as the present study was performed in vitro, future research should be directed towards characterizing the in vivo effects of ATRA.

Acknowledgements

Not applicable.

Funding

The present study was funded by the National Natural Science Foundation of China (grant no. 81770889), the Natural Science Foundation of Guangdong Province (grant no. 2017A030313774) and the Health Programs of Finance Department of Jilin Province (grant no. 3D5177783429).

Availability of data and materials

All the data generated and analyzed in the present study are available from the corresponding author upon reasonable request.

Authors' contributions

YL and YZ conceived and designed the experiments. LL and XW performed the experiments. LL, XW, and YZ analyzed the data. LL and YL wrote the manuscript. All the authors have read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Thylefors B and Négrel AD: The global impact of glaucoma. Bull World Health Org. 72:323–326. 1994.PubMed/NCBI

2 

Foster A and Johnson GJ: Magnitude and causes of blindness in the developing world. Int Ophthalmol. 14:135–140. 1990. View Article : Google Scholar : PubMed/NCBI

3 

Quigley HA: Number of people with glaucoma worldwide. Br J Ophthalmol. 80:389–393. 1996. View Article : Google Scholar : PubMed/NCBI

4 

Sommer A: Doyne lecture. Glaucoma: Facts and figures. Eye (Lond). 10:295–301. 1996. View Article : Google Scholar : PubMed/NCBI

5 

Jay JL: Rational choice of therapy in primary open angle glaucoma. Eye (Lond). 6:243–247. 1992. View Article : Google Scholar : PubMed/NCBI

6 

Migdal C, Gregory W and Hitchings R: Long-term functional outcome after early surgery compared with laser and medicine in open-angle glaucoma. Ophthalmology. 101:1651–1657. 1994. View Article : Google Scholar : PubMed/NCBI

7 

Chang L, Crowston JG, Cordeiro MF, Akbar AN and Khaw PT: The role of the immune system in conjunctival wound healing after glaucoma surgery. Surv Ophthalmol. 45:49–68. 2000. View Article : Google Scholar : PubMed/NCBI

8 

Atreides SP, Skuta GL and Reynolds AC: Wound healing modulation in glaucoma filtering surgery. Int Ophthalmol Clin. 44:61–106. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Desjardins DC, Parrish RK II, Folberg R, Nevarez J, Heuer DK and Gressel MG: Wound healing after filtering surgery in owl monkeys. Arch Ophthalmol. 104:1835–1839. 1986. View Article : Google Scholar : PubMed/NCBI

10 

Saika S, Yamanaka O, Okada Y, Tanaka S, Miyamoto T, Sumioka T, Kitano A, Shirai K and Ikeda K: TGF beta in fibroproliferative diseases in the eye. Front Biosci (Schol Ed). 1:376–390. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Cong M, Iwaisako K, Jiang C and Kisseleva T: Cell Signals Influencing Hepatic Fibrosis. Int J Hepatol. 2012:1585472012. View Article : Google Scholar : PubMed/NCBI

12 

Sang H, Li T, Li H and Liu J: Gab1 regulates proliferation and migration through the PI3K/Akt signaling pathway in intrahepatic cholangiocarcinoma. Tumour Biol. 36:8367–8377. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Zhou H, Li D, Shi C, Xin T, Yang J, Zhou Y, Hu S, Tian F, Wang J and Chen Y: Effects of exendin-4 on bone marrow mesenchymal stem cell proliferation, migration and apoptosis in vitro. Sci Rep. 5:128982015. View Article : Google Scholar : PubMed/NCBI

14 

Zeng R, Xiong Y, Zhu F, Ma Z, Liao W, He Y, He J, Li W, Yang J, Lu Q, et al: Fenofibrate attenuated glucose-induced mesangial cells proliferation and extracellular matrix synthesis via PI3K/AKT and ERK1/2. PLoS One. 8:e768362013. View Article : Google Scholar : PubMed/NCBI

15 

Wen X, Li Y, Hu K, Dai C and Liu Y: Hepatocyte growth factor receptor signaling mediates the anti-fibrotic action of 9-cisretinoic acid in glomerular mesangial cells. Am J Pathol. 167:947–957. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Liu Y, Kimura K, Orita T, Teranishi S, Suzuki K and Sonoda KH: Inhibition by all-trans-retinoic acid of transforming growth factor-β-induced collagen gel contraction mediated by human tenon fibroblasts. Invest Ophthalmol Vis Sci. 55:4199–4205. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Sun L, Dong Y, Zhao J, Yin Y, Tong B, Zheng Y and Xin H: NPPB modulates apoptosis, proliferation, migration and extracellular matrix synthesis of conjunctival fibroblasts by inhibiting PI3K/AKT signaling. Int J Mol Med. 41:1331–1338. 2018.PubMed/NCBI

18 

Marshall J: Transwell(®) invasion assays. Methods Mol Biol. 769:97–110. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Liu Y, Ko JA, Yanai R, Kimura K, Chikama T, Sagara T and Nishida T: Induction by latanoprost of collagen gel contraction mediated by human tenon fibroblasts: Role of intracellular signaling molecules. Invest Ophthalmol Vis Sci. 49:1429–1436. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Liu Y, Yanai R, Lu Y, Kimura K and Nishida T: Promotion by fibronectin of collagen gel contraction mediated by human corneal fibroblasts. Exp Eye Res. 83:1196–1204. 2006. View Article : Google Scholar : PubMed/NCBI

21 

Cordeiro MF: Role of transforming growth factor beta in conjunctival scarring. Clin Sci (Lond). 104:181–187. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Ye Y and Dan Z: All-trans retinoic acid diminishes collagen production in a hepatic stellate cell line via suppression of active protein-1 and c-Jun N-terminal kinase signal. J Huazhong Univ Sci Technolog Med Sci. 30:726–733. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Dong Z, Tai W, Yang Y, Zhang T, Li Y, Chai Y, Zhong H, Zou H and Wang D: The role of all-trans retinoic acid in bleomycin-induced pulmonary fibrosis in mice. Exp Lung Res. 38:82–89. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Morath C, Dechow C, Lehrke I, Haxsen V, Waldherr R, Floege J, Ritz E and Wagner J: Effects of retinoids on the TGF-beta system and extracellular matrix in experimental glomerulonephritis. J Am Soc Nephrol. 12:2300–2309. 2001.PubMed/NCBI

25 

Zhou TB, Drummen GP and Qin YH: The controversial role of retinoic acid in fibrotic diseases: Analysis of involved signaling pathways. Int J Mol Sci. 14:226–243. 2013. View Article : Google Scholar

26 

Chen J: The IL-23/IL-17 axis may be important in obesity-asso-ciated cancer by way of the activation of multiple signal pathways. Int J Obes (Lond). 34:1227–1229. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Xie Y, Shi X, Sheng K, Han G, Li W, Zhao Q, Jiang B, Feng J, Li J and Gu Y: PI3K/Akt signaling transduction pathway, erythropoiesis and glycolysis in hypoxia (Review). Mol Med Rep. 19:783–791. 2019.PubMed/NCBI

28 

Liu Y, Li W, Liu H, Peng Y, Yang Q, Xiao L, Liu Y and Liu F: Inhibition effect of small interfering RNA of connective tissue growth factor on the expression of extracellular matrix molecules in cultured human renal proximal tubular cells. Ren Fail. 36:278–284. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Qin D, Zhang GM, Xu X and Wang LY: The PI3K/Akt signaling pathway mediates the high glucose-induced expression of extracellular matrix molecules in human retinal pigment epithelial cells. J Diabetes Res. 2015:9202802015. View Article : Google Scholar : PubMed/NCBI

30 

Chung EJ, Sohn YH, Kwon SH, Jung SA and Lee JH: Lithium chloride inhibits TGF-β1-induced myofibroblast transdifferentiation via PI3K/Akt pathway in cultured fibroblasts from Tenon's capsule of the human eye. Biotechnol Lett. 36:1217–1224. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Guo X, Yang Y, Liu L, Liu X, Xu J, Wu K and Yu M: Pirfenidone induces G1 arrest in human Tenon's fibroblasts in vitro involving AKT and MAPK signaling pathways. J Ocul Pharmacol Ther. 33:366–374. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Carducci A, Scafetta G, Siciliano C, Carnevale R, Rosa P, Coccia A, Mangino G, Bordin A, Vingolo EM, Pierelli L, et al: GMP-grade platelet lysate enhances proliferation and migration of tenon fibroblasts. Front Biosci (Elite Ed). 8:84–99. 2016.PubMed/NCBI

33 

Li N, Cui J, Duan X, Chen H and Fan F: Suppression of type I collagen expression by miR-29b via PI3K, Akt, and Sp1 pathway in human Tenon's fibroblasts. Invest Ophthalmol Vis Sci. 53:1670–1678. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Zhang YE: Non-Smad pathways in TGF-beta signaling. Cell Res. 19:128–139. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Wen J, Lin X, Gao W, Qu B, Ling Y, Liu R and Yu M: MEK inhibition prevents TGF-β1-induced myofibroblast trans differentiation in human tenon fibroblasts. Mol Med Rep. 19:468–476. 2019.PubMed/NCBI

36 

Addicks EM, Quigley HA, Green WR and Robin AL: Histologic characteristics of filtering blebs in glaucomatous eyes. Arch Ophthalmol. 101:795–798. 1983. View Article : Google Scholar : PubMed/NCBI

37 

Hitchings RA and Grierson I: Clinico pathological correlation in eyes with failed fistulizing surgery. Trans Ophthalmol Soc U K. 103:84–88. 1983.PubMed/NCBI

38 

Mielke C, Dawda VK and Anand N: Intraoperative 5-fluorouracil application during primary trabeculectomy in Nigeria: A comparative study. Eye (Lond). 17:829–834. 2003. View Article : Google Scholar : PubMed/NCBI

39 

Wong TT, Khaw PT, Aung T, Foster PJ, Htoon HM, Oen FT, Gazzard G, Husain R, Devereux JG, Minassian D, et al: The singapore 5-fluorouracil trabeculectomy study: Effects on intraocular pressure control and disease progression at 3 years. Ophthalmology. 116:175–184. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Shin DH, Ren J, Juzych MS, Hughes BA, Kim C, Song MS, Yang KJ and Glover KB: Primary glaucoma triple procedure in patients with primary open-angle glaucoma: The effect of mitomycin-C in patients with and without prognostic factors for filtration failure. Am J Ophthalmol. 125:346–352. 1998. View Article : Google Scholar : PubMed/NCBI

41 

Perkins TW, Gangnon R, Ladd W, Kaufman PL and Heatley GA: Trabeculectomy with mitomycin C: Intermediate-term results. J Glaucoma. 7:230–236. 1998. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2019
Volume 20 Issue 3

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liang L, Wang X, Zheng Y and Liu Y: All‑trans‑retinoic acid modulates TGF‑β‑induced apoptosis, proliferation, migration and extracellular matrix synthesis of conjunctival fibroblasts by inhibiting PI3K/AKT signaling. Mol Med Rep 20: 2929-2935, 2019
APA
Liang, L., Wang, X., Zheng, Y., & Liu, Y. (2019). All‑trans‑retinoic acid modulates TGF‑β‑induced apoptosis, proliferation, migration and extracellular matrix synthesis of conjunctival fibroblasts by inhibiting PI3K/AKT signaling. Molecular Medicine Reports, 20, 2929-2935. https://doi.org/10.3892/mmr.2019.10507
MLA
Liang, L., Wang, X., Zheng, Y., Liu, Y."All‑trans‑retinoic acid modulates TGF‑β‑induced apoptosis, proliferation, migration and extracellular matrix synthesis of conjunctival fibroblasts by inhibiting PI3K/AKT signaling". Molecular Medicine Reports 20.3 (2019): 2929-2935.
Chicago
Liang, L., Wang, X., Zheng, Y., Liu, Y."All‑trans‑retinoic acid modulates TGF‑β‑induced apoptosis, proliferation, migration and extracellular matrix synthesis of conjunctival fibroblasts by inhibiting PI3K/AKT signaling". Molecular Medicine Reports 20, no. 3 (2019): 2929-2935. https://doi.org/10.3892/mmr.2019.10507