Combined treatment with vitamin E and gefitinib has synergistic effects to inhibit TGF-β1-induced renal fibroblast proliferation

  • Authors:
    • Giou‑Teng Yiang
    • Jen‑Ni Chen
    • Pei‑Shiuan Lin
    • Hsiao‑Chun Liu
    • Shu‑Ying Chen
    • Chyou‑Wei Wei
  • View Affiliations

  • Published online on: April 21, 2016     https://doi.org/10.3892/mmr.2016.5155
  • Pages: 5372-5378
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Renal fibroblast proliferation is key in renal fibrosis and chronic kidney disease. Transforming growth factor-β1 (TGF-β1) has been demonstrated to be an important factor that induces cell proliferation in renal fibroblasts. Epidermal growth factor receptor (EGFR) is also recognized as a factor promoting renal fibroblast proliferation. In addition, mitogen‑activated protein kinase signaling pathways are associated with TGF‑β1‑ and EGFR‑induced cell proliferation. Gefitinib, an EGFR tyrosine kinase inhibitor, is predominantly used as an anti‑tumor therapeutic agent in clinical therapeutic strategies. However, gefitinib has been suggested to exert anti‑proliferative effects on renal fibroblasts, however, high‑dose gefitinib may result in serious side effects. The present study aims to determine whether low‑dose gefitinib reduces gefitinib‑induced side effects and maintains the anti‑proliferative effects on renal fibroblasts. TGF‑β1 promotes cell proliferation in renal fibroblasts, and the current study demonstrates that low‑dose gefitinib treatment exhibits anti‑proliferative effects similar to those of high‑dose gefitinib treatment. Thus, although high‑dose gefitinib is a conventional anti‑tumor drug, low‑dose gefitinib may be of use in renal fibrosis treatment. Furthermore, the present study demonstrates that a combined treatment with low-dose gefitinib and vitamin E has synergistic effects that reduce TGF‑β1‑induced fibroblast proliferation, cell-cycle arrest and the ERK phosphorylation pathway.

Introduction

Renal fibroblast proliferation induces tubulointerstitial fibrosis resulting in renal filtration dysfunction (1) and chronic kidney disease (CKD) (2,3), thus, the inhibition of fibroblast proliferation to prevent CKD is an important area. Transforming growth factor-β1 (TGF-β1) is important in the induction of proliferation in human renal fibroblasts (4,5). Previous studies have suggested that induction of renal fibrosis by TGF-β1 is associated with p53 (6,7), reactive oxygen species (8,9), the Smad signaling pathway (10,11), mitogen activated protein kinase (MAPK) signaling pathways (12,13), and RhoA/Rho kinase (9,14). These studies indicated that TGF-β1 is a critical factor in activating numerous signal transduction pathways that result in proliferation in renal fibroblasts. Thus, TGF-β1 was used in the present study as a cell model for investigating anti-proliferative effects on renal fibroblasts by gefitinib and vitamin E treatment alone and in combination. Results from the current study demonstrated that 0.2 nM TGF-β1 promoted renal fibroblast proliferation.

The epidermal growth factor receptor (EGFR) signaling pathway induces cell proliferation in various cells (1518). Previous studies have demonstrated that the EGFR signaling pathway mediates renal fibroblast proliferation and renal fibrogenesis (19,20). Gefitinib, an EGFR tyrosine kinase inhibitor, inhibits EGFR signaling activation resulting in cell growth arrest (21,22). Thus, gefitinib has generally been used for clinical tumor treatment (2325). As EGFR mediates renal fibroblast proliferation and EGFR is blocked by gefitinib, a previous study has successfully used gefitinib to inhibit renal fibroblast proliferation (26). The present study demonstrated that gefitinib attenuates fibroblast proliferation by blocking the EGFR signaling pathway and by inhibiting the TGF-β1-mediated pathway. In addition, previous studies have suggested that the EGFR signaling pathway is associated with the TGF-β1-mediated pathway (8,27). Similar to these studies, experimental data from the present study also demonstrated that gefitinib inhibits TGF-β1-induced fibroblast proliferation. Although gefitinib effectively inhibits fibroblast proliferation to prevent renal fibrosis, the side effects as a result of gefitinib are also clinically important (2831).

Vitamin E exerts an anti-oxidative and protective effect against various oxidative stress-associated diseases, including hypertension, cardiovascular disease, hemorrhagic liposis, and obesity-associated diseases (3235). However, vitamin E exerts an anti-fibrotic effect on the renal cell-mediated TGF-β1 signaling pathway (3638). A previous study indicated that vitamin E reduces progression of fibrosis in obstructed kidneys (36). Other previous studies have demonstrated that vitamin E in combination with pentoxifylline or Fuzheng Huayu recipe, a traditional Chinese medicine, inhibits TGF-β1-induced fibrosis (37,38). Results from the present study also demonstrated that vitamin E inhibits cell proliferation in TGF-β1-treated renal fibroblasts. Furthermore, the present study indicates that a combination treatment of low-dose vitamin E and low-dose gefitinib has a more marked anti-proliferative effect on TGF-β1-treated renal fibroblasts than high-dose vitamin E treatment or high-dose gefitinib treatment alone. This suggests that combination treatment with low-dose vitamin E and low-dose gefitinib is a potential therapeutic strategy to inhibit fibroblast proliferation and prevent high-dose gefitinib treatment-induced side effects.

Three major MAPK signaling pathways contain extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase, and p38 mitogen-activated protein kinase (39,40). Previous studies have suggested that renal fibroblast proliferation is mediated by MAPK signaling pathways (41,42). The present study demonstrates that ERK phosphorylation is increased in TGF-β1-treated renal fibroblasts, suggesting TGF-β1-induced proliferation is mediated by the ERK signaling pathway. In addition, the present study demonstrated that combination treatment of low-dose vitamin E and low-dose gefitinib reduces TGF-β1-induced increases in ERK phosphorylation levels. The present study indicates that combination treatment with low-dose gefitinib and low-dose vitamin E has synergistic effects to inhibit TGF-β1-induced renal fibroblast proliferation mediated by the ERK phosphorylation signaling pathway.

Materials and methods

Materials

TGF-β1 was obtained from R&D Systems, Inc. (Minneapolis, MN, USA). Anti-ERK (1:400; cat. no. BS3627), anti-p-ERK (1:400; cat. no. BS5016), anti-p38 (1:400; cat. no. BS3567) and anti-p-p38 (1:400; cat. no. BS4766) primary rabbit polyclonal antibodies were purchased from Bioworld (Louis Park, MN, USA). Horseradish peroxidae-conjugated goat anti-rabbit IgG, secondary antibody (1:2,000, cat. no. 7074) was purchased from Cell Signaling Technology (Danvers, MA, USA). 5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium (MTT) assay kit was bought from Bio Basic Canada, Inc. (Markham, ON, Canada). Fetal bovine serum (FBS), Dulbecco's modified Eagle's medium (DMEM), non-essential amino acids, L-glutamine, and penicillin/streptomycin were purchased from Hyclone (GE Healthcare Life Sciences, Logan, UT, USA). Vitamin E and dimethyl sulfoxide (DMSO) was obtained from Sigma-Aldrich (St. Louis, MO, USA). Gefitinib was purchased from AstraZeneca UK Limited (London, UK).

Cell line and cell culture

The NRK-49F rat renal fibroblast cell line was obtained from Bioresource Collection and Research Center (Hsinchu, Taiwan). The cell line was cultured in DMEM supplemented with 10% FBS, 2 mM L-glutamine, 100 IU/ml penicillin/streptomycin, and 0.1 mM non-essential amino acids, and maintained in a humidified atmosphere with 5% CO2 at 37°C.

Cell survival rate assay

Survival rates of NRK-49F cells were measured with the MTT assay method as described in previous studies (43,44). Briefly, cells were cultured in 96-well plates. On the second day, cells were divided into control group and experimental groups and MTT assays (with DMSO treatment). were determined at 24 and 72 h according to the manufacturer's protocols. Absorbance was determined under a multi-well ELISA reader (SpectraMax Paradigm Multi-Mode Microplate Reader; Molecular Devices, Sunnyvale, CA, USA) at a wavelength of 570 nm. Survival rates were indicated using the following formula: A570 experimental group / A570 control group.

Cell cycle analysis

Cell cycle analysis was conducted using fluorescence-activated cell sorting as described previously (45,46). Briefly, NRK-49F cells from the control and experimental groups were collected and washed with phosphate-buffered saline (PBS; containing 140 mM NaCl, 2.5 mM KCl, 15 mM Na2HPO4 and 1.6 mM KH2PO4), then fixed with 70% ethanol (Echo Chemical Co., Ltd., Miaoli, Taiwan) at 4°C for 1 h. The fixed cells were washed with PBS and then treated with 1 ml propidium iodide (PI) solution (50 μg/ml PI, 100 μg/ml RNase A, and 0.1% Triton X-100) for 30 min at 37°C. Following this, cells were washed with PBS and analyzed by flow cytometry (Partec CyFlow® SL; Sysmex Partec GmbH, Görlitz, Germany). The resulting data was analyzed with WinMDI version 2.8 software (http://winmdi.software.informer.com/2.8/).

Sodium dodecyl sulfate (SDS) electrophoresis and western blotting

Gel electrophoresis and western blotting were performed as previously described (47,48). Briefly, cells were treated with lysis buffer (containing 50 mM Tris-HCl, 120 mM NaCl, 1 mM EDTA and 1% NP-40) and centrifuged at 16,000 × g for 10 min at 4°C. The supernatant layer containing proteins was collected and the protein level was determined using a Bicinchoninic Acid Protein Assay Reagent kit (Pierce Biotechnology, Rockford, IL, USA) with a DU 530 spectrophotometer (OD 562 nm; Beckman Coulter, Inc., Brea, CA, USA). Equal quantities of protein (60 μg) were loaded and run on an SDS-PAGE for 45 min and transferred to a PVDF membrane. The membranes were blocked with 5% milk for 2 h and washed three times with PBS. The membranes were incubated with primary antibodies in 5% milk for 2 h. The membranes were then washed with PBS three times and incubated with secondary antibodies for 1 h. Protein levels were analyzed with Western Lightning® Chemiluminescence Plus reagent (PerkinElmer, Inc., Waltham, MA, USA) and were observed with a Luminescence Image Analysis system (LAS-4000, FUJIFILM Electronic Materials Taiwan Co., Ltd., Tainan, Taiwan).

Statistical analysis

Data were measured from four independent experiments and are presented as the mean ± standard deviation. The data was analyzed using a Student's t-test with Excel 2010 (http://microsoft-excel-2010.updatestar.com/zh-tw). P<0.05 was considered to indicate a statistically significant difference between two groups.

Results

TGF-β1 induces renal fibroblast proliferation in a time-dependent manner

Consistent with data from previous studies (4,5), the data from the present study demonstrates that TGF-β1 induces proliferation in renal fibroblasts. Compared with growth in the control cells (without TGF-β1 treatment), the survival rate is ~150% in TGF-β1-treated cells at 24 h. However, the survival rate is significantly increased by >360% in TGF-β1-treated cells at 72 h (P<0.01; Fig. 1). The present study suggested that TGF-β1 induces cell proliferation in renal fibroblasts in a time-dependent manner. The present study then used TGF-β1-induced cell proliferation as an experimental model to investigate the antiproliferative effects of gefitinib treatment, vitamin E treatment, and combination treatment of gefitinib and vitamin E on renal fibroblasts.

Gefitinib exerts antiproliferative effects on TGF-β1-treated renal fibroblasts

The present study aimed to investigate whether gefitinib inhibits TGF-β1-induced cell proliferation. The anti-proliferative effects of gefitinib (high dose, 100 μM; low dose for clinical tumor treatment, 13 μM; and low dose, 1 μM) were examined in TGF-β1-treated renal fibroblasts. Compared with the control group, the 24-h survival rates in the present study were ~150% in the TGF-β1-treated group and ~100% in the TGF-β1 + gefitinib-treated groups (Fig. 2A). In addition, the 72-h survival rates were >360% in the TGF-β1-treated group and <250% in the TGF-β1 + gefitinib-treated group (P<0.01; Fig. 2B). Results from the present study demonstrated that gefitinib reduces TGF-β1-induced cell proliferation. Furthermore, as shown in Fig. 2, there is no marked difference in survival rates among the TGF-β1 + gefitinib (100, 13 and 1 μM)-treated groups at 24 and 72 h. The data from the present study suggested that high- and low-dose gefitinib (100, 13 and 1 μM) are equally effective at inhibiting TGF-β1-induced cell proliferation (as shown in Fig. 2B).

Vitamin E reduces cell proliferation in TGF-β1-treated renal fibroblasts in a dose-dependent manner

A previous study indicated that vitamin inhibits the progression of fibrosis in obstructed kidneys (36). Numerous other studies have demonstrated that epithelial-mesenchymal transition (EMT) and fibroblast proliferation induce renal fibrosis (4952). The present study further determined whether vitamin E inhibits fibroblast proliferation directly to prevent progression of fibrosis. As presented in Fig. 3, the 72-h survival rate is >360% in the TGF-β1-treated group, ~330% in the TGF-β1 + 5 μM vitamin E-treated group, and <230% in the TGF-β1 + gefitinib-treated group and the TGF-β1 + 50 μM vitamin E-treated group. The present study demonstrated that high-dose vitamin E (50 μM) treatment reduces TGF-β1-induced cell proliferation (P<0.01 vs. the TGF β1 group, Fig. 3) similarly to gefitinib treatment. However, low-dose vitamin E (5 μM) treatment did not markedly reduce TGF-β1-induced cell proliferation. Thus, the results from the present study suggest that vitamin E reduces TGF-β1-induced renal cell proliferation in a dose-dependent manner.

Gefitinib enhances the antiproliferative effects of vitamin E on TGF-β1-treated renal fibroblasts

The present study aimed to investigate whether gefitinib promotes the antiproliferative effects of vitamin E on TGF-β1-treated cells. The 72-h survival rate was >360% in the TGF-β1-treated group, ~330% in the TGF-β1 + 5 μM vitamin E-treated group, and ~160% in TGF-β1 + 5 μM vitamin E with various concentrations of gefitinib-treated groups (Fig. 4A). In addition, the 72-h survival rate was >360% in the TGF-β1-treated group, ~220% in the TGF-β1 + 50 μM vitamin E-treated group, and ~150% in the TGF-β1 + 50 μM vitamin E with various concentrations of gefitinib-treated groups (Fig. 4B). These data suggest that gefitinib enhances the antiproliferative effects of vitamin E on TGF-β1-treated cells. However, the antiproliferative effects were not markedly different among those treated with vitamin E + various concentrations (1, 13 and 100 μM) of gefitinib. Furthermore, the data demonstrated that although low-dose vitamin E does not have notable antiproliferative effects, combination treatment of low-dose vitamin E and gefitinib effectively reduces TGF-β1-induced cell proliferation (P<0.01 vs. the TGF-β1 group, Fig. 4A).

Combination treatment of low-dose gefitinib and low-dose vitamin E has synergistic effects to reduce TGF-β1-induced renal fibroblast proliferation

As presented in Figs. 2 and 3, gefitinib and vitamin E have been demonstrated to exert anti-proliferative effects on TGF-β1-treated cells. The current study further analyzed the anti-proliferative effects on TGF-β1-induced cell proliferation in the gefitinib-treated group, the vitamin E-treated group, and the gefitinib + vitamin E-treated group. As presented in Fig. 5, the 72-h survival rate was >360% in TGF-β1-treated cells, ~340% in TGF-β1 with low-dose vitamin E-treated group, and ~250% in TGF-β1 with high-dose vitamin E-treated or gefitinib groups. These data indicate that low-dose vitamin E does not have marked anti-proliferative effects on TGF-β1-induced cell proliferation; however, high-dose vitamin E and low-dose gefitinib have similar anti-proliferative effects on TGF-β1-induced cell proliferation. Furthermore, the present study demonstrated that the 72-h survival rates are ~160% in TGF-β1-induced cells with low-dose gefitinib + high-dose or low-dose vitamin E-treated groups. These data indicate that the anti-proliferative effects in combination treatment with low-dose gefitinib and low-dose vitamin E is similar to combination treatment with low-dose gefitinib and high-dose vitamin E. Furthermore, the combination treatment with gefitinib and vitamin E has stronger anti-proliferative effects than gefitinib treatment alone or vitamin E treatment alone. Thus, the results of the present study suggest that combination treatment of low-dose gefitinib and low-dose vitamin E reduces TGF-β1-induced cell proliferation (P<0.01 vs. the TGF-β1 group, Fig. 5).

Combination treatment with gefitinib and vitamin E reduces TGF-β1-induced cell proliferation associated with the cell cycle and ERK signaling pathway

The cell cycle was analyzed in the control group, the TGF-β1-treated group, and the TGF-β1 with gefitinib + vitamin E-treated group. As presented in Fig. 6A, the G1 phase was ~69.6% and the S-phase is ~2.45% in the control group. As presented in Fig. 6B, the G1 phase was ~41.9% and the S-phase was ~10.87% in TGF-β1-treated group. As presented in Fig. 6C, the G1 phase was ~61.16% and the S-phase was ~4.77% in TGF-β1 with gefitinib + vitamin E-treated group. All data obtained from flow cytometry were analyzed using Student's t-test. The S-phase percentage was significantly increased in the TGF-β1-treated group compared with the control (P<0.05, as determined from four independent flow cytometry experiments, data not shown), this indicates that TGF-β1 accelerate entry to S-phase, resulting in cell proliferation. Furthermore, S-phase percentage is significantly increased in the TGF-β1-treated group compared with the TGF-β1 with gefitinib + vitamin E-treated group. The result suggested that combination treatment with gefitinib and vitamin E may ameliorate the increase in cells entering the S-phase in TGF-β1-treated cells. Furthermore, previous studies have demonstrated MAPK signaling pathways, including ERK and p38 phosphorylation, are associated with renal fibroblast proliferation (41,42). Thus, ERK and p38 phosphorylation, p-ERK and p-p38, were analyzed in the control group, the TGF-β1-treated group, and the TGF-β1 with gefitinib + vitamin E-treated group (Fig. 7). Results from the present study demonstrated that p-ERK was not observed in the control group (Fig. 7, lane 1), but is evident in the TGF-β1-treated group (Fig. 7, lane 2). This suggests TGF-β1 may induce renal cell proliferation may be via ERK phosphorylation. However, p-ERK was also not observed in the TGF-β1 with gefitinib + vitamin E-treated group (Fig. 7, lane 3). The data indicated gefitinib + vitamin E treatment inhibits ERK phosphorylation. However, the p-p38 levels were not significantly different among the three groups. The results from the current study suggest that combination treatment with gefitinib + vitamin E reduces TGF-β1-induced cell proliferation associated with ERK phosphorylation.

Discussion

Gefitinib, an EGFR tyrosine kinase inhibitor, inhibits cell growth (21,22) and has been used for various tumor treatments, including, lung, esophageal and breast cancer (25,29,53). Numerous studies have demonstrated that therapeutic doses of gefitinib for clinical tumor treatment result in side effects, including severe hepatotoxicity (29), acneiform eruption, severe xerosis of skin, paronychia (30), and empyema (31). However, in the present study, the results indicated that there are similar anti-proliferative effects on TGF-β1-treated renal fibroblasts among high-dose, therapeutic dose, and low-dose gefitinib treatments (Fig. 2). The results of the present study indicate that gefitinib, a conventional therapeutic agent for tumor treatment, may be useful for the treatment of renal fibrosis at a low-dose.

Multiple studies have demonstrated that renal fibrosis is induced via the EMT process and renal fibroblast proliferation (4952). In addition, it has been reported that EMT and fibroblast proliferation are induced by activation of the TGF-β1 signaling pathway (5457). Similar to these studies, data from the present study also showed that TGF-β1 induces renal fibroblast proliferation. Previous research has indicated that vitamin E in combination with other therapeutic agents reduces progression of TGF-β1-induced fibrosis (37,38). The current study further demonstrated that vitamin E alone inhibits TGF-β1-induced fibroblast proliferation (Fig. 3). In addition, high-dose vitamin E, like gefitinib, has a more marked anti-proliferative effect than low-dose vitamin E. Although the anti-fibrotic effects exerted by vitamin E remain to be elucidated, the present study demonstrated that vitamin E reduces proliferation in TGF-β1-treated fibroblasts.

Results from the present study demonstrate that combination treatment with gefitinib and vitamin E has an increased anti-proliferative effect on TGF-β1-treated cells compared with gefitinib or vitamin E treatment alone. Furthermore, these results also demonstrated that combination treatment with low-dose gefitinib and low-dose vitamin E has anti-proliferative effects similar to combination treatment with high-dose gefitinib and high-dose vitamin E. The results of the current study demonstrate that low-dose gefitinib and low-dose vitamin E treatment may be a potential therapeutic strategy for renal fibrosis and an effective alternative to avoid high-dose gefitinib-induced side effects.

Previous studies have suggested that TGF-β1 induces cell cycle-associated protein expression (4,58) and activates MAPK signaling pathways (59,60). Similar to these studies, the results from the present study have also demonstrated that TGF-β1 promotes cells to enter the S-phase and activate ERK phosphorylation. In addition, the present study also demonstrated that combination treatment with low-dose gefitinib and vitamin E induces G1 arrest and reduces ERK phosphorylation levels to inhibit TGF-β1-induced proliferation.

In conclusion, the present study demonstrated that combination treatment with low-dose gefitinib and vitamin E has anti-proliferative effects on TGF-β1-treated fibroblasts via cell cycle arrest and inactivation of the ERK signaling pathway.

Acknowledgments

The present study was supported by the Taipei Tzu Chi Hospital (grant. nos. TCRD-TPE-103-48, TCRD-TPE-104-34 and TCRD-105-20).

References

1 

Hundae A and McCullough PA: Cardiac and renal fibrosis in chronic cardiorenal syndromes. Nephron Clin Pract. 127:106–112. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Chen YX, Zhang W, Wang WM, Yu XL, Wang YM, Zhang MJ and Chen N: Role of moesin in renal fibrosis. PloS One. 9:e1129362014. View Article : Google Scholar : PubMed/NCBI

3 

Eddy AA: Overview of the cellular and molecular basis of kidney fibrosis. Kidney Int Suppl (2011). 4:2–8. 2014. View Article : Google Scholar

4 

Strutz F, Zeisberg M, Renziehausen A, Raschke B, Becker V, van Kooten C and Müller G: TGF-beta 1 induces proliferation in human renal fibroblasts via induction of basic fibroblast growth factor (FGF-2). Kidney Int. 59:579–592. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Oujo B, Muñoz-Félix JM, Arévalo M, Núñez-Gómez E, Pérez-Roque L, Pericacho M, González-Núñez M, Langa C, Martínez-Salgado C, Perez-Barriocanal F, et al: L-endoglin overexpression increases renal fibrosis after unilateral ureteral obstruction. PloS One. 9:e1103652014. View Article : Google Scholar : PubMed/NCBI

6 

Deshpande SD, Putta S, Wang M, Lai JY, Bitzer M, Nelson RG, Lanting LL, Kato M and Natarajan R: Transforming growth factor-β-induced cross talk between p53 and a microRNA in the pathogenesis of diabetic nephropathy. Diabetes. 62:3151–3162. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Overstreet JM, Samarakoon R, Meldrum KK and Higgins PJ: Redox control of p53 in the transcriptional regulation of TGF-β1 target genes through SMAD cooperativity. Cell Signal. 26:1427–1436. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Samarakoon R, Dobberfuhl AD, Cooley C, Overstreet JM, Patel S, Goldschmeding R, Meldrum KK and Higgins PJ: Induction of renal fibrotic genes by TGF-β1 requires EGFR activation, p53 and reactive oxygen species. Cell Signal. 25:2198–2209. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Manickam N, Patel M, Griendling KK, Gorin Y and Barnes JL: RhoA/Rho kinase mediates TGF-β1-induced kidney myofibroblast activation through Poldip2/Nox4-derived reactive oxygen species. Am J Physiol Renal Physiol. 307:F159–F171. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Kim D, Lee AS, Jung YJ, Yang KH, Lee S, Park SK, Kim W and Kang KP: Tamoxifen ameliorates renal tubulointerstitial fibrosis by modulation of estrogen receptor α-mediated transforming growth factor-β1/Smad signaling pathway. Nephrol Dial Transplant. 29:2043–2053. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Wang L, Chi YF, Yuan ZT, Zhou WC, Yin PH, Zhang XM, Peng W and Cai H: Astragaloside IV inhibits renal tubulointerstitial fibrosis by blocking TGF-β/Smad signaling pathway in vivo and in vitro. Exp Biol Med (Maywood). 239:1310–1324. 2014. View Article : Google Scholar

12 

Park SH, Cho HJ, Jeong YJ, Shin JM, Kang JH, Park KK, Choe JY, Park YY, Bae YS, Han SM, et al: Melittin inhibits TGF-β-induced pro-fibrotic gene expression through the suppression of the TGFβRII-Smad, ERK1/2 and JNK-mediated signaling pathway. Am J Chin Med. 42:1139–1152. 2014. View Article : Google Scholar

13 

Zhang L, Zhang J, Liu X, Liu S and Tian J: Tribbles 3 regulates the fibrosis cytokine TGF-β 1 through ERK1/2-MAPK signaling pathway in diabetic nephropathy. J Immunol Res. 2014:2403962014. View Article : Google Scholar

14 

Chen G, Chen X, Sukumar A, Gao B, Curley J, Schnaper HW, Ingram AJ and Krepinsky JC: TGFβ receptor I transactivation mediates stretch-induced Pak1 activation and CTGF upregulation in mesangial cells. J Cell Sci. 126:3697–3712. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Lanaya H, Natarajan A, Komposch K, Li L, Amberg N, Chen L, Wculek SK, Hammer M, Zenz R, Peck-Radosavljevic M, et al: EGFR has a tumour-promoting role in liver macrophages during hepatocellular carcinoma formation. Nat Cell Biol. 16:972–981. 2014. View Article : Google Scholar :

16 

Salazar N, Muñoz D, Kallifatidis G, Singh RK, Jordà M and Lokeshwar BL: The chemokine receptor CXCR7 interacts with EGFR to promote breast cancer cell proliferation. Mol Cancer. 13:1982014. View Article : Google Scholar : PubMed/NCBI

17 

Yoo YH, Kim YR, Kim MS, Lee KJ, Park KH and Hahn JH: YAC tripeptide of epidermal growth factor promotes the proliferation of HaCaT keratinocytes through activation of EGFR. BMB Rep. 47:581–586. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Kundu S, Sengupta S and Bhattacharyya A: NF-κB acts downstream of EGFR in regulating low dose cadmium induced primary lung cell proliferation. Biometals. 26:897–911. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Liu N, Guo JK, Pang M, Tolbert E, Ponnusamy M, Gong R, Bayliss G, Dworkin LD, Yan H and Zhuang S: Genetic or pharmacologic blockade of EGFR inhibits renal fibrosis. J Am Soc Nephrol. 23:854–867. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Ponnusamy M, Zhou X, Yan Y, Tang J, Tolbert E, Zhao TC, Gong R and Zhuang S: Blocking sirtuin 1 and 2 inhibits renal interstitial fibroblast activation and attenuates renal interstitial fibrosis in obstructive nephropathy. J Pharmacol Exp Ther. 350:243–256. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Han SY, Ding HR, Zhao W, Teng F and Li PP: Enhancement of gefitinib-induced growth inhibition by Marsdenia tenacissima extract in non-small cell lung cancer cells expressing wild or mutant EGFR. BMC Complement Altern Med. 14:1652014. View Article : Google Scholar : PubMed/NCBI

22 

Sakurai MA, Ozaki Y, Okuzaki D, Naito Y, Sasakura T, Okamoto A, Tabara H, Inoue T, Hagiyama M, Ito A, et al: Gefitinib and luteolin cause growth arrest of human prostate cancer PC-3 cells via inhibition of cyclin G-associated kinase and induction of miR-630. PloS One. 9:e1001242014. View Article : Google Scholar : PubMed/NCBI

23 

Bersanelli M, Tiseo M, Artioli F, Lucchi L and Ardizzoni A: Gefitinib and afatinib treatment in an advanced non-small cell lung cancer (NSCLC) patient undergoing hemodialysis. Anticancer Res. 34:3185–3188. 2014.PubMed/NCBI

24 

Deangelo DJ, Neuberg D, Amrein PC, Berchuck J, Wadleigh M, Sirulnik LA, Galinsky I, Golub T, Stegmaier K and Stone RM: A phase II study of the EGFR inhibitor gefitinib in patients with acute myeloid leukemia. Leuk Res. 38:430–434. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Kalykaki A, Agelaki S, Kallergi G, Xyrafas A, Mavroudis D and Georgoulias V: Elimination of EGFR-expressing circulating tumor cells in patients with metastatic breast cancer treated with gefitinib. Cancer Chemother Pharmacol. 73:685–693. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Chen SC, Guh JY, Lin TD, Chiou SJ, Hwang CC, Ko YM and Chuang LY: Gefitinib attenuates transforming growth factor-beta1-activated mitogen-activated protein kinases and mitogenesis in NRK-49F cells. Transl Res. 158:214–224. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Midgley AC, Rogers M, Hallett MB, Clayton A, Bowen T, Phillips AO and Steadman R: Transforming growth factor-β1 (TGF-β1)-stimulated fibroblast to myofibroblast differentiation is mediated by hyaluronan (HA)-facilitated epidermal growth factor receptor (EGFR) and CD44 co-localization in lipid rafts. J Biol Chem. 288:14824–14838. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Zhao C, Chen J, Yu B, Wu X, Dai C, Zhou C and Chen X: Effect of modified taohongsiwu decoction on patients with chemotherapy-induced hand-foot syndrome. J Tradit Chin Med. 34:10–14. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Yonesaka K, Suzumura T, Tsukuda H, Hasegawa Y, Ozaki T, Sugiura T and Fukuoka M: Erlotinib is a well-tolerated alternate treatment for non-small cell lung cancer in cases of gefitinib-induced hepatotoxicity. Anticancer Res. 34:5211–5215. 2014.PubMed/NCBI

30 

Madke B, Gole P, Kumar P and Khopkar U: Dermatological side effects of epidermal growth factor receptor inhibitors: 'PRIDE' complex. Indian J Dermatol. 59:271–274. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Funakoshi Y, Takeuchi Y and Maeda H: Pneumonectomy after response to gefitinib treatment for lung adenocarcinoma. Asian Cardiovasc Thorac Ann. 21:482–484. 2013. View Article : Google Scholar

32 

Kuwabara A, Nakade M, Tamai H, Tsuboyama-Kasaoka N and Tanaka K: The association between vitamin E intake and hypertension: Results from the re-analysis of the national health and nutrition survey. J Nutr Sci Vitaminol (Tokyo). 60:239–245. 2014. View Article : Google Scholar

33 

Ahmadi A, Mazooji N, Roozbeh J, Mazloom Z and Hasanzade J: Effect of alpha-lipoic acid and vitamin E supplementation on oxidative stress, inflammation and malnutrition in hemodialysis patients. Iran J Kidney Dis. 7:461–467. 2013.PubMed/NCBI

34 

Hsieh CL, Chen KC, Lin PX, Peng CC and Peng RY: Resveratrol and vitamin E rescue valproic acid-induced teratogenicity: The mechanism of action. Clin Exp Pharmacol Physiol. 41:210–219. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Shen X, Tang Q, Wu J, Feng Y, Huang J and Cai W: Effect of vitamin E supplementation on oxidative stress in a rat model of diet-induced obesity. Int J Vitam Nutr Res. 79:255–263. 2009. View Article : Google Scholar

36 

Tasanarong A, Kongkham S, Thitiarchakul S and Eiam-Ong S: Vitamin E ameliorates renal fibrosis in ureteral obstruction: Role of maintaining BMP-7 during epithelial-to-mesenchymal transition. J Med Assoc Thai. 94(Suppl 7): S10–S18. 2011.

37 

Hamama S, Gilbert-Sirieix M, Vozenin MC and Delanian S: Radiation-induced enteropathy: Molecular basis of pentoxifylline-vitamin E anti-fibrotic effect involved TGF-β1 cascade inhibition. Radiother Oncol. 105:305–312. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Wang QL, Yuan JL, Tao YY, Zhang Y, Liu P and Liu CH: Fuzheng Huayu recipe and vitamin E reverse renal interstitial fibrosis through counteracting TGF-beta1-induced epithelial-to-mesenchymal transition. J Ethnopharmacol. 127:631–640. 2010. View Article : Google Scholar

39 

Knebel B, Lehr S, Hartwig S, Haas J, Kaber G, Dicken HD, Susanto F, Bohne L, Jacob S, Nitzgen U, et al: Phosphorylation of sterol regulatory element-binding protein (SREBP)-1c by p38 kinases, ERK and JNK influences lipid metabolism and the secretome of human liver cell line HepG2. Arch Physiol Biochem. 120:216–227. 2014. View Article : Google Scholar : PubMed/NCBI

40 

He W, Wang Z, Luo Z, Yu Q, Jiang Y, Zhang Y, Zhou Z, Smith AJ and Cooper PR: LPS promote the odontoblastic differentiation of human dental pulp stem cells via MAPK signaling pathway. J Cell Physiol. 230:554–561. 2015. View Article : Google Scholar

41 

Wang D, Warner GM, Yin P, Knudsen BE, Cheng J, Butters KA, Lien KR, Gray CE, Garovic VD, Lerman LO, et al: Inhibition of p38 MAPK attenuates renal atrophy and fibrosis in a murine renal artery stenosis model. Am J Physiol Renal Physiol. 304:F938–F947. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Xiao HB, Liu RH, Ling GH, Xiao L, Xia YC, Liu FY, Li J, Liu YH, Chen QK, Lv JL, et al: HSP47 regulates ECM accumulation in renal proximal tubular cells induced by TGF-beta1 through ERK1/2 and JNK MAPK pathways. Am J Physiol Renal Physiol. 303:F757–F765. 2012. View Article : Google Scholar : PubMed/NCBI

43 

Yiang GT, Chou PL, Hung YT, Chen JN, Chang WJ, Yu YL and Wei CW: Vitamin C enhances anticancer activity in methotrexatetreated Hep3B hepatocellular carcinoma cells. Oncol Rep. 32:1057–1063. 2014.PubMed/NCBI

44 

Yu YL, Yiang GT, Chou PL, Tseng HH, Wu TK, Hung YT, Lin PS, Lin SY, Liu HC, Chang WJ and Wei CW: Dual role of acetaminophen in promoting hepatoma cell apoptosis and kidney fibroblast proliferation. Mol Med Rep. 9:2077–2084. 2014.PubMed/NCBI

45 

Nho KJ, Chun JM and Kim HK: Agrimonia pilosa ethanol extract induces apoptotic cell death in HepG2 cells. J Ethnopharmacol. 138:358–363. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Yu YL, Yu SL, Su KJ, Wei CW, Jian MH, Lin PC, Tseng IH, Lin CC, Su CC, Chan DC, et al: Extended O6-methylguanine methyltransferase promoter hypermethylation following n-butylidenephthalide combined with 1,3-bis (2-chloroethyl)-1-nitrosourea (BCNU) on inhibition of human hepatocellular carcinoma cell growth. J Agric Food Chem. 58:1630–1638. 2010. View Article : Google Scholar : PubMed/NCBI

47 

Wei CW, Lin CC, Yu YL, Lin CY, Lin PC, Wu MT, Chen CJ, Chang W, Lin SZ, Chen YL and Harn HJ: n-Butylidenephthalide induced apoptosis in the A549 human lung adenocarcinoma cell line by coupled down-regulation of AP-2alpha and telomerase activity. Acta Pharmacol Sin. 30:1297–1306. 2009. View Article : Google Scholar : PubMed/NCBI

48 

Yu YL, Su KJ, Chen CJ, Wei CW, Lin CJ, Yiang GT, Lin SZ, Harn HJ and Chen YL: Synergistic anti-tumor activity of isochaihulactone and paclitaxel on human lung cancer cells. J Cell Physiol. 227:213–222. 2012. View Article : Google Scholar

49 

Guo Y, Li Z, Ding R, Li H, Zhang L, Yuan W and Wang Y: Parathyroid hormone induces epithelial-to-mesenchymal transition via the Wnt/β-catenin signaling pathway in human renal proximal tubular cells. Int J Clin Exp Pathol. 7:5978–5987. 2014.

50 

Wei J, Li Z and Yuan F: Evodiamine might inhibit TGF-beta1-induced epithelial-mesenchymal transition in NRK52E cells via Smad and PPAR-gamma pathway. Cell Biol Int. 38:875–880. 2014. View Article : Google Scholar : PubMed/NCBI

51 

Sun YB, Qu X, Li X, Nikolic-Paterson DJ and Li J: Endothelial dysfunction exacerbates renal interstitial fibrosis through enhancing fibroblast Smad3 linker phosphorylation in the mouse obstructed kidney. PloS One. 8:e840632013. View Article : Google Scholar

52 

Qu X, Zhang X, Yao J, Song J, Nikolic-Paterson DJ and Li J: Resolvins E1 and D1 inhibit interstitial fibrosis in the obstructed kidney via inhibition of local fibroblast proliferation. J Pathol. 228:506–519. 2012. View Article : Google Scholar : PubMed/NCBI

53 

Dutton SJ, Ferry DR, Blazeby JM, Abbas H, Dahle-Smith A, Mansoor W, Thompson J, Harrison M, Chatterjee A, Falk S, et al: Gefitinib for oesophageal cancer progressing after chemotherapy (COG): A phase 3, multicentre, double-blind, placebo-controlled randomised trial. Lancet Oncol. 15:894–904. 2014. View Article : Google Scholar : PubMed/NCBI

54 

Liao XH, Zhang L, Chen GT, Yan RY, Sun H, Guo H and Liu Q: Augmenter of liver regeneration inhibits TGF-β1-induced renal tubular epithelial-to-mesenchymal transition via suppressing TβR II expression in vitro. Exp Cell Res. 327:287–296. 2014. View Article : Google Scholar : PubMed/NCBI

55 

Lan A, Qi Y and Du J: Akt2 mediates TGF-β1-induced epithelial to mesenchymal transition by deactivating GSK3β/snail signaling pathway in renal tubular epithelial cells. Cell Physiol Biochem. 34:368–382. 2014. View Article : Google Scholar

56 

Qi R, Li W and Yu S: FK506 inhibits the mice glomerular mesangial cells proliferation by affecting the transforming growth factor-β and Smads signal pathways. Renal Fail. 36:589–592. 2014. View Article : Google Scholar

57 

Guo W, Xu H, Chen J, Yang Y, Jin JW, Fu R, Liu HM, Zha XL, Zhang ZG and Huang WY: Prohibitin suppresses renal interstitial fibroblasts proliferation and phenotypic change induced by transforming growth factor-beta1. Mol Cell Biochem. 295:167–177. 2007. View Article : Google Scholar

58 

Zhu B, Jin Y, Han L, Chen H, Zhong F, Wang W and Chen N: Proteasome inhibitor inhibits proliferation and induces apoptosis in renal interstitial fibroblasts. Pharmacol Rep. 65:1357–1365. 2013. View Article : Google Scholar

59 

Rodríguez-Barbero A, Dorado F, Velasco S, Pandiella A, Banas B and Lopez-Novoa JM: TGF-beta1 induces COX-2 expression and PGE2 synthesis through MAPK and PI3K pathways in human mesangial cells. Kidney Int. 70:901–909. 2006. View Article : Google Scholar : PubMed/NCBI

60 

Loeffler I, Hopfer U, Koczan D and Wolf G: Type VIII collagen modulates TGF-β1-induced proliferation of mesangial cells. J Am Soc Nephrol. 22:649–663. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2016
Volume 13 Issue 6

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
Yiang GT, Chen JN, Lin PS, Liu HC, Chen SY and Wei CW: Combined treatment with vitamin E and gefitinib has synergistic effects to inhibit TGF-β1-induced renal fibroblast proliferation. Mol Med Rep 13: 5372-5378, 2016
APA
Yiang, G., Chen, J., Lin, P., Liu, H., Chen, S., & Wei, C. (2016). Combined treatment with vitamin E and gefitinib has synergistic effects to inhibit TGF-β1-induced renal fibroblast proliferation. Molecular Medicine Reports, 13, 5372-5378. https://doi.org/10.3892/mmr.2016.5155
MLA
Yiang, G., Chen, J., Lin, P., Liu, H., Chen, S., Wei, C."Combined treatment with vitamin E and gefitinib has synergistic effects to inhibit TGF-β1-induced renal fibroblast proliferation". Molecular Medicine Reports 13.6 (2016): 5372-5378.
Chicago
Yiang, G., Chen, J., Lin, P., Liu, H., Chen, S., Wei, C."Combined treatment with vitamin E and gefitinib has synergistic effects to inhibit TGF-β1-induced renal fibroblast proliferation". Molecular Medicine Reports 13, no. 6 (2016): 5372-5378. https://doi.org/10.3892/mmr.2016.5155