Open Access

Gremlin mediates the TGF‑β‑induced induction of profibrogenic genes in human retinal pigment epithelial cells

  • Authors:
    • Dong Qin
    • Xuemin Jin
    • Yanrong Jiang
  • View Affiliations

  • Published online on: January 21, 2020     https://doi.org/10.3892/etm.2020.8463
  • Pages: 2353-2359
  • Copyright: © Qin et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Proliferative vitreoretinopathy (PVR) is characterised by the contraction and growth of fibrotic membranes on the retina and within the vitreous body. Retinal pigment epithelial (RPE) cells, a major cellular component of the fibrotic membrane, is one of the cell types that have been previously reported to associate with PVR pathogenesis. During PVR, RPE cells undergo increased cell proliferation, migration and the secretion of extracellular matrix molecules, such as fibronectin and type I collagen. A variety of cytokines and growth factors are involved in the formation of the fibrotic membrane. Although gremlin has been reported to serve an important role in the regulation of epithelial‑to‑mesenchymal transition in PVR, the relationship between gremlin and the expression of profibrogenic factors in human RPE cells remains unclear. In the present study, gremlin promoted RPE cell proliferation and the expression of type I collagen and fibronectin. In addition, knocking down gremlin expression by siRNA significantly suppressed the transforming growth factor (TGF)‑β1‑ and TGF‑β2‑induced expression of type I collagen and fibronectin in RPE cells. These findings suggest that gremlin may serve an important role in the development of PVR.

Introduction

Proliferative vitreoretinopathy (PVR) is a serious complication that is caused by rhegmatogenous retinal detachment and is the leading cause of retinal detachment surgery failure (1,2). It is physiologically characterised by increased cell proliferation, migration and secretion of extracellular matrix (ECM) proteins, which results in the formation of fibrotic membranes in response to retinal detachment. Retinal pigment epithelial (RPE) cells are one of the major cellular components of the fibrotic membrane. Of the number of cytokines and growth factors that have been previously reported to contribute to PVR pathogenesis, transforming growth factor (TGF)-β is of particular importance (3).

Gremlin is a highly conserved 184-amino-acid protein that contains a cysteine-rich region (47). Structurally, it is a member of the cysteine knot superfamily, which can be present in both soluble and cell-associated forms (811). Gremlin belongs to a family of bone morphogenetic protein antagonists that participate in a number of physiological processes, including cell survival, differentiation, growth and development (4,812). Gremlin is predominantly localised to the outer retina, and high levels of its expression have been demonstrated in bovine retinal pericytes in response to elevated glucose levels compared with control treated pericytes (13). The dysfunction of gremlin has also been observed to be associated with a number of diseases, such as diabetic fibrotic disease (4,812,14).

Although it has been previously shown that, during PVR, gremlin induces epithelial-to-mesenchymal transition (EMT) in RPE cells (15), information on the potential link between gremlin and the expression of pro-fibrogenic factors in human RPE cells remain limited. The present study demonstrated that gremlin increased the proliferation and expression of fibronectin and type I collagen in human RPE cells, whereas gremlin knockdown by small interfering (si)RNA expression significantly reduced TGF-β1- and TGF-β2-induced expression of fibronectin and type I collagen in human RPE cells.

Materials and methods

Reagents

Gremlin-1 (cat. no. SRP3285) and DAPI (cat. no. D9542) were purchased from Sigma-Aldrich; Merck KGaA. Recombinant human TGF-β1 and TGF-β2 were obtained from Cell Signaling Technology, Inc. Anti-fibronectin (cat. no. ab2413) and anti-type I collagen (cat. no. ab34710) antibodies, type I collagen (human pro-collagen Iα, cat. no. ab229389) and fibronectin (cat. no. ab219046) ELISA kits were purchased from Abcam. TRITC conjugated goat anti-rabbit IgG secondary antibody (cat. no. ZF-0316) was obtained from Zhongshan Golden Bridge Biotechnology Co., Ltd. Human gremlin siRNA and siRNA control (cat. no. siP06969473939) were purchased from Guangzhou RiboBio Co., Ltd. The Gremlin siRNA sequence is CCACCTACCAAGAAGAAGA.

Cell culture

Human RPE cells (ARPE-19; CRL-2302) were purchased from the American Type Culture Collection. The cells were cultured in DMEM (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% FBS (Gibco; Thermo Fisher Scientific, Inc.), 100 U/ml penicillin and 100 ng/ml streptomycin at 37°C and 5% CO2 in a humidified atmosphere. ARPE-19 cells were transfected as described below. Transfected ARPE-19 cells were incubated with recombinant human TGF-β1 or TGF-β2.

Gremlin siRNA transfection

The cells were transfected with gremlin siRNA or control siRNA (50 nM each). Transfections were performed using the riboFECT™ CP regeant according to the manufacturer' protocol. Assays were performed 48 h after transfection, including assessment of mRNA and protein levels and immunofluorescent staining.

Cell viability and proliferation assay

Cell viability and proliferation was measured using MTT and 5′bromo-2-deoxyuridine (BrdU) assays. For MTT assay, RPE cells were cultured in 96-well plates at 1.0×104 cells/well for 24 h and were partially starved in DMEM supplemented with 1% FBS for 12 h, following which they were treated with various concentrations (25, 50 and 100 ng/ml) of gremlin at various time points (12, 24 and 48 h). MTT (5 mg/ml; 20 µl) was then added to the culture medium, and the cells were incubated for an additional 4 h at 37°C. DMSO was then added to each well to dissolve the formazan crystals formed. Absorbance in each well was measured at 490 nm using a microplate reader. The BrdU assay was performed according to the manufacturer's protocol. A total of 5.0×103 cells were seeded in a 96-well plate for 24 h and were partially starved in DMEM supplemented with 1% FBS for 12 h, and then treated with or without tested substances for 48 h at 37°C. A 10 µM concentration of BrdU was added to the medium. Measurement of cell proliferation was quantified by BrDU ELISA kit (Roche Diagnostics). Absorbance in each well was measured at 450 nm with a microplate reader (Bio-Rad Laboratories, Inc.)

Reverse transcription-quantitative PCR (RT-qPCR)

TRIzol® (Invitrogen; Thermo Fisher Scientific, Inc.) was used to extract total RNA from the RPE cells. cDNA synthesis was performed with 2 µg total RNA using RevertAid First Strand cDNA Synthesis kit (Fermentas; Thermo Fisher Scientific, Inc.) according to manufacturer's protocols. The reaction mixtures contained the following: 1 µl oligo(dT) primer, 4 µl 5× reaction buffer, 1 µl Ribolock™ RNase Inhibitor, 2 µl 10 mM dNTP mix and 1 µl RevertAid™ M-MuLV Reverse Transcriptase in a total volume of 20 µl. Incubation was for 60 min at 42°C before the reaction was terminated by heating at 70°C for 5 min. The ABI Sequence Detector System 7500 (Thermo Fisher Scientific, Inc.) was used to perform RT-qPCR. The PCR solution consisted of specific primers (0.3 µM each), 2.5 µl cDNA, and 12.5 µl Maxima SYBR-Green qPCR Master Mix (Fermentas; Thermo Fisher Scientific, Inc.) to a final reaction volume of 25 µl. The sequences of the primers used were as follows: Human fibronectin forward, 5′-GATAAATCAACAGTGGGAGC-3′ and reverse, 5′-CCCAGATCATGGAGTCTTTA-3′; human type I collagen forward, 5′-TGGTGGTTATGACTTTGGTTACGAT-3 and reverse, 5′-TGTGCGAGCTGGGTTCTTTCTA-3′; human GAPDH forward, 5′-TGTTCGACAGTCAGCCGCAT-3′ and reverse, 5′-ACTCCGACCTTCACCTTCCC-3′; and human gremlin forward, 5′-AAGCGAGACTGGTGCAAAAC-3′ and reverse, 5′-CTTGCAGAAGGAGCAGGACT-3′. The thermocycling conditions were as follows: Initial denaturation at 95°C for 10 min, followed by 39 cycles of 95°C for 15 sec, 61°C for 30 sec and 72°C for 30 sec. The relative mRNA expression levels were calculated as the difference between target and GAPDH expression levels using the 2−ΔΔCq method (16).

Immunofluorescence staining

Human RPE cells were fixed with 4% paraformaldehyde for 20 min at 23°C, washed three times with PBS and permeabilised using 0.5% Triton X-100 for 15 min at 23°C. The cells were subsequently blocked with 10% goat serum (Zhongshan Golden Bridge Biotechnology) for 1 h at 23°C before incubation with rabbit anti-fibronectin (dilution, 1:100) or rabbit anti-collagen I (dilution, 1:100) primary antibodies, which were diluted in PBS supplemented with 10% goat serum, overnight at 4°C. The next day, the cells were incubated with TRITC-conjugated goat anti-rabbit IgG (dilution, 1:200) secondary antibodies at room temperature for 1 h. The samples were counterstained with DAPI (1:1,000; cat. no. D9542, Sigma-Aldrich; Merck KGaA) at room temperature for 10 min. The samples were examined using a fluorescence microscope (DS-Ril-U2; Nikon Corporation) and photographed at ×200 magnification in five random fields (DS-U2; Nikon Corporation).

ELISA

The measurement of fibronectin protein levels of using ELISA have been previously reported (1719). Cells were seeded at 4×103 cells per well in 12-well plates. After reaching a confluency of approximately 85–90%, the medium was changed to a serum-free medium for 12 h. Cells were transfected with control siRNA or gremlin siRNA using the riboFECT™ CP reagent. After transfection, cells were incubated with recombinant human TGF-β1 or TGF-β2 for 48 h at 37°C. The samples were collected after the cells were treated. Fibronectin and type I collagen levels in the culture supernatants of RPE cells were measured using fibronectin and type I collagen ELISA kits according to manufacturer's protocols.

Statistical analysis

All statistical differences were evaluated using one-way analysis of variance followed by Tukey's test. All data are presented as the mean ± SD and were analysed using SPSS 17.0 software (SPSS, Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

Gremlin induces human RPE cell proliferation and increases fibronectin and type I collagen expression

To examine the role of gremlin in the proliferation and expression of fibronectin and type I collagen in human RPE cells, cells were treated with ascending concentrations of gremlin (0, 25, 50 and 100 ng/ml) for 48 h. At a concentration of 50 ng/ml or 100 ng/ml, gremlin significantly increased RPE cell proliferation compared with the control group (Fig. 1A and C). In addition, RPE cell proliferation was significantly enhanced following incubation with 100 ng/ml gremlin for 24 and 48 h compared with the control group (Fig. 1B and D). Subsequently, the mRNA and protein levels of fibronectin and type I collagen were measured using RT-qPCR, ELISA and immunofluorescence staining. At a concentration of 25, 50 or 100 ng/ml, recombinant gremlin significantly increased the expression of fibronectin (Fig. 2A and B) and type I collagen (Fig. 2C and D) compared with the control group. In addition, strong positive staining of fibronectin (Fig. 2E) and type I collagen (Fig. 2F) was observed following treatment of RPE cells with gremlin (100 ng/ml, 48 h).

TGF-β1 and TGF-β2 increase the expression of gremlin, fibronectin and type I collagen in human RPE cells

The protein and mRNA levels of gremlin, fibronectin and type I collagen were measured using ELISA and RT-qPCR following treatment of RPE cells with TGF-β. TGF-β1 and TGF-β2 treatment increased the expression of gremlin in human RPE cells, which was significantly reversed by gremlin siRNA (Fig. 3A and B). TGF-β1 (10 mg/ml) increased fibronectin (Fig. 3C and D) and type I collagen (Fig. 3E and F) expression in human RPE cells and their levels in the cell supernatant. Additionally, TGF-β2 (5 ng/ml) significantly upregulated the levels of fibronectin (Fig. 3G and H) and type I collagen (Fig. 3I and J) in human RPE cells and RPE cell supernatant.

Gremlin mediated TGF-β induces the expression of fibronectin and type I collagen in human RPE cells

Gremlin knockdown following TGF-β-treatment decreased the expression of fibronectin and type I collagen in human RPE cells. RT-qPCR and ELISA were used to measure the expression or secretion of fibronectin and type I collagen. Gremlin knockdown significantly reversed the TGF-β1-induced expression and secretion of fibronectin (Fig. 4A and B) and type I collagen (Fig. 4C and D) compared with the TGF-β1-treated group. Compared with the TGF-β2-treated group, gremlin knockdown also significantly reversed TGF-β2 induced expression and secretion of fibronectin (Fig. 4E and F) and type I collagen (Fig. 4G and H).

Discussion

Fibrotic membrane contractions can result in severe visual impairment or blindness (20). RPE cell proliferation, migration and ECM deposition are some of the main physiological changes that occur during PVR, which is under the influence of a variety of cytokines and growth factors (2123). In PVR, RPE cells undergo EMT and begin losing epithelial characteristics whilst adopting a more mesenchymal, fibroblast-like phenotype, resulting in increased proliferation and migration, resistance to apoptosis and production of ECM proteins (24,25). However, the relationship between gremlin and the expression of proteins associated with the ECM in human RPE cells currently remains unclear. In the present study, the role of gremlin in the expression of fibronectin and type I collagen, and in RPE cell proliferation, was investigated. It was found that gremlin significantly increased the proliferation and expression of fibronectin and type I collagen in RPE cells, suggesting that gremlin may serve a role in the development of PVR.

TGF-β is a major cytokine involved in the regulation of cell proliferation, migration, cell death, differentiation, protein synthesis, and the pathological and physiological processes associated with tissue repair and development (26). Elevated TGF-β levels have been reported in the subretinal fluids, vitreous and epiretinal membranes of patients with PVR (2730). TGF-β serves a vital role in PVR formation since it regulates cell proliferation, enhances ECM protein synthesis and induces ECM deposition (23,31). It has been previously demonstrated that the inhibition of TGF-β expression may prevent PVR progression (32). In addition, silencing of gremlin expression has been shown to significantly reduce TGF-β1-induced ECM expression in human tubular epithelial cells and TGF-β2-induced ECM synthesis in human lens epithelial cells (33,34). However, the relationship between gremlin and TGF-β1 and TGF-β2 in human RPE cells remains currently unclear. To clarify this, the results of the present study showed that TGF-β1 and TGF-β2 increased gremlin expression in human RPE cells, whilst gremlin knockdown significantly reversed TGF-β1- and TGF-β2-induced ECM expression in human RPE cells. This observation suggested that TGF-β1 and TGF-β2 can both induce the expression of ECM proteins by increasing gremlin expression, further implicating gremlin to be a mediator in PVR pathogenesis associated with TGF-β.

The present study does have a number of limitations. Apart from fibronectin and collagen type I, epithelial markers such as E-cadherin and zonula occludens-1, and mesenchymal markers including N-cadherin and α-smooth muscle actin, are also involved in the development of PVR (35,36). In the present study, the migration of RPE cells was not studied. In addition, the levels of fibronectin and collagen I were measured using ELISA and not by western blotting.

In conclusion, the data from the present study suggested that gremlin can increase RPE cell proliferation and induce fibronectin and collagen I expression in human RPE cells. Silencing of gremlin expression significantly reduced TGF-β1- and TGF-β2-induced fibronectin and collagen I expression in human RPE cells. Thus, TGF-β induced fibronectin and collagen I expression via regulation of gremlin. Therefore, gremlin may serve a role in the development of PVR, which potentially extends our knowledge regarding the role of gremlin in PVR pathogenesis.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Department of Science and Technology of Henan Province (grant no. 162300410112).

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

DQ performed the experiments and wrote the paper. YJ analysed the data and revised the paper. XJ designed the experiments. All authors read and approved the final 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 

Sadaka A, Sisk RA, Osher JM, Toygar O, Duncan MK and Riemann CD: Intravitreal methotrexate infusion for proliferative vitreoretinopathy. Clin Ophthalmol. 10:1811–1817. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Claes C and Lafetá AP: Proliferative vitreoretinopathy. Dev Ophthalmol. 54:188–195. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Pena RA, Jerdan JA and Glaser BM: Effects of TGF-beta and TGF-beta neutralizing antibodies on fibroblast-induced collagen gel contraction: Implications for proliferative vitreoretinopathy. Invest Ophthalmol Vis Sci. 35:2804–2808. 1994.PubMed/NCBI

4 

Lappin DW, McMahon R, Murphy M and Brady HR: Gremlin: An example of the re-emergence of developmental programmes in diabetic nephropathy. Nephrol Dial Transplant. 17 (Suppl 9):S65–S67. 2002. View Article : Google Scholar

5 

Grillo E, Ravelli C, Corsini M, Ballmer-Hofer K, Zammataro L, Oreste P, Zoppetti G, Tobia C, Ronca R, Presta M and Mitola S: Monomeric gremlin is a novel vascular endothelial growth factor receptor-2 antagonist. Oncotarget. 7:35353–35368. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Kišonaitė M, Wang X and Hyvönen M: Structure of Gremlin-1 and analysis of its interaction with BMP-2. Biochem J. 473:1593–1604. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Yin Y, Yang Y, Yang L, Yang Y, Li C, Liu X and Qu Y: Overexpression of Gremlin promotes non-small cell lung cancer progression. Tumour Biol. 37:2597–2602. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Wordinger RJ, Zode G and Clark AF: Focus on molecules: Gremlin. Exp Eye Res. 87:78–79. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Müller I, Schönberger T, Schneider M, Borst O, Ziegler M, Seizer P, Leder C, Müller K, Lang M, Appenzeller F, et al: Gremlin-1 is an inhibitor of macrophage migration inhibitory factor and attenuates atherosclerotic plaque growth in ApoE−/− Mice. J Biol Chem. 288:31635–31645. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Sethi A, Wordinger RJ and Clark AF: Gremlin utilizes canonical and non-canonical TGFb signaling to induce lysyl oxidase (LOX) genes in human trabecular meshwork cells. Exp Eye Res. 113:117–127. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Cahill E, Costello CM, Rowan SC, Harkin S, Howell K, Leonard MO, Southwood M, Cummins EP, Fitzpatrick SF, Taylor CT, et al: Gremlin plays a key role in the pathogenesis of pulmonary hypertension. Circulation. 125:920–930. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Mitola S, Ravelli C, Moroni E, Salvi V, Leali D, BallmerHofer K, Zammataro L and Presta M: Gremlin is a novel agonist of the major proangiogenic receptor VEGFR2. Blood. 116:3677–3680. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Kane R, Stevenson L, Godson C, Stitt AW and O'Brien CO: Gremlin gene expression in bovine retinal pericytes exposed to elevated glucose. Br J Ophthalmol. 89:1638–1642. 2005. View Article : Google Scholar : PubMed/NCBI

14 

McMahon R, Murphy M, Clarkson M, Taal M, Mackenzie HS, Godson C, Martin F and Brady HR: IHG-2, a mesangial cell gene induced by high glucose, is human gremlin. Regulation by extracellular glucose concentration, cyclic mechanical strain, and transforming growth factor-beta 1. J Biol Chem. 275:9901–9904. 2000. View Article : Google Scholar : PubMed/NCBI

15 

Lee H, O'Meara SJ, O'Brien C and Kane R: The role of Gremlin, a BMP antagonist, and epithelial-to-mesenchymal transition in proliferative vitreoretinopathy. Invest Ophthalmol Vis Sci. 48:4291–4299. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Chen J, Ma Y, Wang Z, Wang H, Wang L, Xiao F, Wang H, Tan J and Guo Z: Thrombin promotes fibronectin secretion by bone marrow mesenchymal stem cells via the protease-activated receptor mediated signalling pathways. Stem Cell Res Ther. 5:362014. View Article : Google Scholar : PubMed/NCBI

18 

Zhang S, Shiels IA, Ambler JS and Taylor SM: Pirfenidone reduces fibronectin synthesis by cultured human retinal pigment epithelial cells. Aust N Z J Ophthalmol. 26 (Suppl 1):S74–S76. 1998. View Article : Google Scholar : PubMed/NCBI

19 

Osusky R, Soriano D, Ye J and Ryan SJ: Cytokine effect on fibronectin release by retinal pigment epithelial cells. Curr Eye Res. 13:569–574. 1994. View Article : Google Scholar : PubMed/NCBI

20 

Kwon OW, Song JH and Roh MI: Retinal detachment and proliferative vitreoretinopathy. Dev Ophthalmol. 55:154–162. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Por ED, Greene WA, Burke TA and Wang HC: Trichostatin a inhibits retinal pigmented epithelium activation in an in vitro model of proliferative vitreoretinopathy. J Ocul Pharmacol Ther. 32:415–524. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Pennock S, Haddock LJ, Mukai S and Kazlauskas A: Vascular endothelial growth factor acts primarily via platelet-derived growth factor recptor α to promote proliferative vitreoretinopathy. Am J Pathol. 184:3052–3068. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Xiao W, Chen X, Liu X, Luo L, Ye S and Liu Y: Trichostatin A, a histone deacetylase inhibotor, suppresses proliferation and epithelial-mesenchymal transition in retinal pigment epithelial cells. J Cell Mol Med. 18:646–655. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Rouberol F and Chiquet C: Proliferative vitreoretinopathy: Pathophysiology and clinical diagnosis. J Fr Ophtalmol. 37:557–565. 2014.(In French). View Article : Google Scholar : PubMed/NCBI

25 

Winkler J and Hoerauf H: TGF-β and RPE-derived cells in taut subretinal strands from patients with proliferative vitreoretinopathy. Eur J Ophthalmol. 21:422–426. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Saika S: TGF beta pathobiology in the eye. Lab Invest. 86:106–115. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Chen YS, Hackett SF, Schoenfeld CL, Vinores MA, Vinores SA and Campochiaro PA: Localisation of vascular endothelial growth factor and its receptors to cells of vascular and avascular epiretinal membranes. Br J Ophthalmol. 81:919–926. 1997. View Article : Google Scholar : PubMed/NCBI

28 

Bochaton-Piallat ML, Kapetanios AD, Donati G, Redard M, Gabbiani G and Pournaras CJ: TGF-beta1, TGF-beta receptor II and ED-A fibronectin expression in myofibroblast of vitreoretinopathy. Invest Ophthalmol Vis Sci. 41:2336–2342. 2000.PubMed/NCBI

29 

Gaudric A, Glacet-Bernard A, Clement G, Falquerho L, Barritault D and Coscas G: Transforming growth factor beta in the vitreous of patients with epiretinal proliferation. Ophthalmologe. 4:51–52. 1990.(In French).

30 

Hirase K, Sugiyama T, Ikeda T, Sotozono C, Yasuhara T, Koizumi K and Kinoshita S: Transforming growth factor beta (2) increases in subretinal fluid in rhegmatogenous retinal detachment with subretinal strands. Ophthalmologica. 219:222–225. 2005. View Article : Google Scholar : PubMed/NCBI

31 

Cai W, Wei Q, Liu Q, Ren C, Liu J, Zhang R, He M, Wang Q, Du Y and Yu J: Effect of bradykinin on TGF-β1-induced retinal pigment epithelial cell proliferation and extracellular matrix secretion. BMC Ophthalmol. 16:1992016. View Article : Google Scholar : PubMed/NCBI

32 

Nassar K and Grisanti S, Tura A, Lüke J, Lüke M, Soliman M and Grisanti S: A TGF-b receptor 1 inhibitor for prevention of proliferative vitreoretinopathy. Exp Eye Res. 123:72–86. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Rodrigues-Diez R, Lavoz C, Carvajal G, Rayego-Mateos S, Rodrigues Diez RR, Ortiz A, Egido J, Mezzano S and Ruiz-Ortega M: Gremlin is a downstream profibrotic mediator of transforming growth factor-beta in cultured renal cells. Nephron Exp Nephrol. 122:62–74. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Ma B, Kang Q, Qin L, Cui L and Pei C: TGF-b2 induces transdifferentiation and fibrosis in human lens epithelial cells via regulating gremlin and CTGF. Biochem Biophys Res Commun. 447:689–695. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Feng H, Zhao X, Guo Q, Feng Y, Ma M, Guo W, Dong X, Deng C, Li C, Song X, et al: Autophagy resists EMT progress to maintain retinal pigment epithelium homeostasis. Int J Biol Sci. 15:507–521. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Shanmuganathan S, Sumantran VN and Angayarkanni N: Epigallocatechin gallate and curcumin prevent transforming growth factor beta 1-induced epithelial to mesenchymal transition in ARPE-19 cells. Indian J Med Res. 146 (Suppl 2):S85–S96. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2020
Volume 19 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Qin D, Jin X and Jiang Y: Gremlin mediates the TGF‑β‑induced induction of profibrogenic genes in human retinal pigment epithelial cells. Exp Ther Med 19: 2353-2359, 2020
APA
Qin, D., Jin, X., & Jiang, Y. (2020). Gremlin mediates the TGF‑β‑induced induction of profibrogenic genes in human retinal pigment epithelial cells. Experimental and Therapeutic Medicine, 19, 2353-2359. https://doi.org/10.3892/etm.2020.8463
MLA
Qin, D., Jin, X., Jiang, Y."Gremlin mediates the TGF‑β‑induced induction of profibrogenic genes in human retinal pigment epithelial cells". Experimental and Therapeutic Medicine 19.3 (2020): 2353-2359.
Chicago
Qin, D., Jin, X., Jiang, Y."Gremlin mediates the TGF‑β‑induced induction of profibrogenic genes in human retinal pigment epithelial cells". Experimental and Therapeutic Medicine 19, no. 3 (2020): 2353-2359. https://doi.org/10.3892/etm.2020.8463