ERK1/2 pathway mediates epithelial-mesenchymal transition by cross-interacting with TGFβ/Smad and Jagged/Notch signaling pathways in lens epithelial cells

  • Authors:
    • Xiaoyun Chen
    • Shaobi Ye
    • Wei Xiao
    • Wencong Wang
    • Lixia Luo
    • Yizhi Liu
  • View Affiliations

  • Published online on: April 4, 2014     https://doi.org/10.3892/ijmm.2014.1723
  • Pages: 1664-1670
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Epithelial-mesenchymal transition (EMT) of lens epithelial cells (LECs) is the major pathological mechanism in anterior subcapsular cataract (ASC) and posterior capsule opacification (PCO), which are important causes of visual impairment. Extracellular signal-regulated kinase (ERK)1/2 pathway has been reported to play a major role in carcinogenesis, cancer metastasis and various fibrotic diseases. We hypothesized that ERK1/2 signaling can cross-interact with canonical transforming growth factor β (TGFβ)/Smad signaling and the Notch pathway, which subsequently contributes to LECs EMT. In this study, we demonstrated that ERK1/2 signaling was activated in TGFβ2‑induced EMT in human LECs, whereas the blockade of TGFβ2/Smad2/3 signaling with SB431542 did not inhibit the activation of ERK1/2 induced by TGFβ2. In addition, inactivation of ERK1/2 signaling with a specific MEK/ERK1/2 inhibitor, U0126, completely prevented the TGFβ2-induced upregulation of α-SMA, collagen type I, collagen type IV and fibronectin. We also demonstrated that inactivation of ERK1/2 signaling inhibited canonical TGFβ/Smad signaling, as well as the Jagged/Notch pathway. By contrast, blockade of the Notch pathway by DAPT inhibited the TGFβ2‑induced activation of ERK1/2 pathway in LECs. Thus, results of this study provide evidence for the complex interplay between ERK1/2, TGFβ/Smad, and Jagged/Notch signaling pathways in the regulation of EMT in LECs. Inhibition of the ERK1/2 pathway may therefore have therapeutic value in the prevention and treatment of ASC and PCO.

Introduction

Cataract is the most common cause of visual impairment in the elderly worldwide, particularly in developing countries (1). Anterior subcapsular cataract (ASC) and posterior capsule opacification (PCO) are different types of cataract that share similar cellular and molecular features (2,3). PCO, also known as a secondary cataract, is the most common long-term complication of modern cataract surgery. In the past few decades, although advances in surgical techniques, intraocular lens materials and designs have reduced the PCO rate, the incidence of PCO is still ~20–40% in adults and 100% in children (4,5). At present, cataract surgery and Nd:YAG laser capsulotomy are the only effective treatments for ASC and PCO, however, they are likely to induce many other complications and risks. Therefore, a better understanding of the pathogenesis of these diseases is critical for the development of new pharmacologic treatments.

Accumulating evidence has shown that the epithelial-mesenchymal transition (EMT) of lens epithelial cells (LECs) is a key pathological mechanism involved in the development of ASC (6,7) and PCO (8,9). PCO is caused by a wound healing response of residual LECs following cataract surgery. After surgery, the levels of various cytokines and growth factors increase in the aqueous humor and stimulate the residual LECs to proliferate and undergo EMT (10). Transforming growth factor β (TGFβ), especially TGFβ2, the major isoform in the aqueous humor of the eye, plays a central role in the cell biology of PCO (11). During the process of EMT, LECs undergo cytoskeletal rearrangement and loss of epithelial phenotype, then migrate away from the original location onto the posterior capsule, with the addition of a large amount of extracellular matrix proteins (collagen and fibronectin) deposition, and finally contribute to the development of PCO (7,10). Unlike PCO, ASC is a primary cataract that is mainly caused by ocular trauma, inflammation or irritation (12). The proliferation and EMT of LECs in situ lead to the formation of subcapsular plaques just beneath the lens anterior capsule, similar to the transdifferentiated cells in PCO (2). Thus, inhibition of the proliferation of LECs and EMT may be a promising strategy to prevent ASC and PCO.

Several signaling pathways are involved in the process of LECs EMT in ASC and PCO development. Among these, canonical TGFβ/Smad signaling has been identified to occupy a crucial position in the signaling networks that control EMT of LECs. TGFβ/Smad signaling transmits signals by binding to the related transmembrane type I and II receptors, which subsequently phosphorylate receptor-regulated Smad2 and Smad3 (13). The phosphorylated Smad2/3 bind to the common mediator Smad4 to form a stable hetero-oligomeric complex, and then the complex translocates to the nucleus where the target gene expression is regulated (13). Recent studies have demonstrated that the blockade of TGFβ2/Smad2/3 efficiently prevents the effect of TGFβ2 on LECs migration, extracellular matrix production and EMT (14,15). In addition to the canonical Smad signaling, extracellular signal-regulated kinase (ERK) signaling is involved in TGFβ-induced EMT in different types of cells (1619). The activation of ERK1/2 signaling enhances TGFβ-induced EMT, accompanied by morphological changes, the upregulation of EMT markers and extracellular matrix components. Blocking the function of ERK1/2 using a special inhibitor results in the inhibition of TGFβ-induced EMT (17,20). In LECs EMT, it has been previously reported that ERK1/2 is rapidly activated by TGFβ, and the specific inhibitor of ERK1/2 blocks the morphologic change of LECs and the upregulation of Slug induced by TGFβ (19).

Although the role of ERK1/2 signaling in EMT during cancer progression and some fibrotic disorders has been studied, the interaction of ERK1/2 with the canonical TGFβ/Smad signaling pathway and other signaling pathways in fibrotic diseases is poorly understood. In this study, we demonstrated that the TGFβ2-induced activation of ERK1/2 is independent of TGFβ/Smad signaling in human LECs, while the blockade of ERK1/2 signaling with the inhibitor U0126 completely prevents TGFβ2-induced EMT. Moreover, blockade of ERK1/2 signaling inhibits the canonical Smad signaling pathway, as well as the Jagged/Notch pathway. We also found that non-canonical TGFβ/ERK1/2 signaling can also be mediated by the Notch pathway. Taken together, these results suggested that ERK1/2 signaling cross-interacts with the TGFβ/Smad and the Jagged/Notch signaling pathways, thus mediating EMT in LECs.

Materials and methods

Reagents and antibodies

U0126 (a selective inhibitor of MEK1 and MEK2) and recombinant human TGFβ2 were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA). SB431542 (a specific inhibitor for TGFβ receptor type I/ALK5 kinase that phosphorylates Smad2/3) and DAPT (an inhibitor of Notch receptor cleavage) were purchased from Sigma-Aldrich (St. Louis, MO, USA). Antibodies against ERK1/2, p-ERK1/2, Jagged-1, Notch-1, Notch-2, p-Smad2, p-Smad3, goat anti-rabbit and horse anti-mouse horseradish peroxidase (HRP)-conjugated secondary antibodies were purchased from Cell Signaling Technology Inc. Antibodies against β-actin, α-SMA, collagen type I (Col I), collagen type IV (Col IV), and fibronectin (FN) were purchased from Abcam (Cambridge, UK).

Cell culture and treatment

The SRA01/04 human LEC line was kindly provided by Professor Fu Shang at the Laboratory for Nutrition and Vision Research (Boston, MA, USA), and cultured in Dulbecco’s modified Eagle’s medium (DMEM) containing 10% fetal bovine serum (FBS). The cells were grown at 37°C in a humidified atmosphere containing 5% CO2 and dissociated with 0.25% trypsin-0.02% ethylenediaminetetraacetic acid (EDTA) solution.

For TGFβ2 and U0126 treatments, the cells were seeded in 6-well plates and treated with 10 ng/ml recombinant human TGFβ2 and different concentrations of U0126 for different time-points.

Quantitative PCR analysis for gene expression

Total RNA was isolated from LECs using TRIzol reagent (Invitrogen, Carlsbad, CA, USA), and the RNA was then treated with DNase I (Sigma-Aldrich) to remove genomic DNA contamination. The concentration of total RNA was quantified by spectrophotometry and cDNA was synthesized with a reverse transcription kit (Takara Bio Inc., Otsu, Japan). For quantitative analysis of mRNA expression, the SYBR PrimeScript RT-PCR kit (Takara Bio Inc.) was used to amplify the target genes, and the reactions were performed with the ABI Prism 7000 sequence detection system (Applied Biosystems, Foster City, CA, USA) according to the manufacturer’s protocol. Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was used as an internal control.

Western blot analysis for protein expression

The cells were washed twice with PBS, and then lysed in 100 μl of RIPA buffer with protease inhibitor cocktail for total protein extraction. Protein was collected after centrifugation and mixed with 5X SDS sample buffer. The samples were separated by 10% SDS-PAGE, and then transferred to PVDF membranes. The membranes were blocked with 5% non-fat milk for 1 h and the membranes were subsequently incubated with different primary antibodies at 4°C overnight. The membranes were washed with 1X PBS containing 0.1% Tween-20 (PBST) three times, and incubated with HRP-conjugated secondary antibodies for 1 h at room temperature. The protein bands were detected with chemiluminescence detection reagents. β-actin was used as the loading control. Densitometric analysis was conducted by ImageJ software 1.41 (National Institutes of Health, Bethesda, MD, USA).

Statistical analysis

Experiments presented in the figures are representative of three or more different repetitions. Data were presented as mean ± standard error of the mean (SEM) and analyzed with SPSS 15.0 software (SPSS, Inc., Chicago, IL, USA). A standard Student’s t-test was used for statistical analysis. P<0.05 was considered to indicate statistical significance.

Results

Blockade of ERK1/2 signaling by U0126 prevents TGFβ2- induced EMT in LECs

To examine whether the blockade of ERK1/2 signaling prevented TGFβ2-induced EMT in LECs, U0126 (a selective inhibitor of MEK1 and MEK2) was used. EMT markers such as α-SMA, Col I, Col IV and FN were investigated at mRNA and protein levels by quantitative PCR and western blot analysis, respectively. As shown in Fig. 1, quantitative PCR results showed that the mRNA expression of α-SMA, Co1 I, Col IV and FN were upregulated ~5.5- 4.3- 7.2- and 17.7-fold in TGFβ2-induced LECs for 24 h. In addition, western blot analysis results showed that TGFβ2 significantly increased the protein expression of α-SMA, Col I, Col IV and FN in LECs (Fig. 2). Co-treatment with U0126 markedly abrogated the upregulation of α-SMA, Col I, Col IV and FN induced by TGFβ2 at the mRNA and protein levels (Figs. 1 and 2: P<0.05 vs. TGFβ2 treated with DMSO group). Maximum effect of U0126 was observed at a concentration of 20.0 μM, however, there was no obvious difference between 10.0 and 20.0 μM at mRNA level. These data suggested that the blockade of ERK1/2 pathway by U0126 effectively attenuated TGFβ2-induced EMT in LECs.

TGFβ2-induced ERK1/2 activation is independent of the canonical TGFβ/Smad pathway

To determine whether the canonical TGFβ/Smad signaling is required for the activation of ERK1/2 pathway by TGFβ2, SB431542 (a specific inhibitor for TGFβ receptor type I/ALK5 kinase that phosphorylates Smad2/3) was used. As shown in Fig. 3A and B, when LECs were stimulated by TGFβ2 for 30 min, ERK1/2 was activated via phosphorylation but with an unchanged total protein level, while U0126 treatment completely inhibited the TGFβ2-induced activation of ERK1/2. However, SB431542 treatment had no effect on the phosphorylation of ERK1/2 (Fig. 3A and B: P<0.05 vs. TGFβ2 treated with DMSO group). These results indicated that TGFβ2-induced ERK1/2 activation is independent of the canonical TGFβ/Smad pathway in LECs.

U0126 mediates canonical TGFβ/Smad signaling by inhibiting the phosphorylation of Smad2

To examine whether there is a crosstalk between the ERK1/2 signaling and the canonical TGFβ/Smad pathway, the effect of U0126 on the activation of receptor-regulated Smad proteins Smad2 and Smad3 was examined. As shown in Fig. 3C and D, TGFβ2 alone clearly induced apparent phosphorylation of Smad2 and Smad3 following 60-min treatment, whereas co-treatment with U0126 inhibited the phosphorylation of Smad2, but had no effect on the phosphorylation of Smad3 in LECs (Fig. 3C and D: P<0.05 vs. TGFβ2 treated with DMSO group). Collectively, these data suggested that U0126 inhibits the canonical TGFβ2/Smad signaling transduction by inhibiting the phosphorylation of Smad2. Thus, there is a crosstalk between ERK1/2 signaling and the canonical TGFβ2/Smad signaling pathway in LECs.

U0126 prevents TGFβ2-induced EMT partly by inhibiting the Jagged/Notch pathway

Accumulating evidence suggests that the Notch signaling pathway is a vital regulator in the induction of EMT during embryonic development, cancer metastasis and various fibrotic diseases (21). Results of a previous study also found that the Jagged/Notch pathway is activated through canonical TGFβ2/Smad signaling during EMT in human LECs, while blockade of the Notch pathway with the specific inhibitor DAPT strongly inhibited TGFβ2-induced EMT (unpublished data). Therefore, we investigated whether inactivation of ERK1/2 signaling with U0126 inhibited Notch signaling activated by TGFβ2, and subsequently inhibited LECs EMT. As shown in Figs. 4 and 5, TGFβ2 treatment alone significantly increased the expression of Jagged-1, Notch-1 and Notch-2 at mRNA and protein levels, while U0126 completely attenuated the TGFβ2-induced upregulation of Jagged-1, Notch-1 and Notch-2 (Figs. 4 and 5: P<0.05 vs. TGFβ2 treated with DMSO group). In addition, U0126 treatment attenuated TGFβ2-induced Notch target genes Hes-1 and Hey-1 expression (Fig. 4: P<0.05 vs. TGFβ2 treated with DMSO group). These results suggested that U0126 prevents TGFβ2-induced EMT partly by downregulating the Jagged/Notch pathway. Thus, non-canonical ERK1/2 signaling also contributes to the TGFβ2-induced activation of the Notch pathway in LECs.

Non-canonical TGFβ/ERK1/2 signaling can be mediated by the Notch pathway

It is unclear whether blockade of Notch signaling is able to modulate ERK1/2 signaling pathway activated by TGFβ2. As expected, blockade of the Notch pathway by DAPT clearly inhibited the TGFβ2-induced activation of ERK1/2 pathway in LECs (Fig. 6: P<0.05 vs. TGFβ2 treated with DMSO group). These results suggested that the non-canonical TGFβ/ERK1/2 signaling can be mediated by the Notch pathway conversely in LECs. This finding also indicated that there is a crosstalk between the ERK1/2 signaling and Notch pathways.

Discussion

A growing number of studies have proven that the development of ASC and PCO largely attributes to the EMT of LECs in response to a variety of cytokines, typically TGFβ2. Activation of ERK1/2 pathway plays a critical role in carcinogenesis, cancer metastasis, and various fibrotic diseases, including PCO (19,2224). In this study, we investigated the role of ERK1/2 signaling in TGFβ2-induced EMT in human LECs, with a focus on the interaction of ERK1/2 signaling with the canonical TGFβ2/Smad and the Jagged/Notch pathways. We found that the activation of ERK1/2 signaling by TGFβ2 is independent of canonical TGFβ2/Smad signaling in LECs, while the blockade of ERK1/2 signaling with U0126 markedly prevented TGFβ2-induced EMT. Furthermore, the blockade of ERK1/2 signaling inhibits the canonical Smad signaling pathway, as well as the Jagged/Notch pathway. By contrast, we demonstrated that non-canonical TGFβ/ERK1/2 signaling can also be mediated by the Notch pathway. Therefore, our data suggest that ERK1/2 signaling cross-interacts with the canonical TGFβ/Smad and Jagged/Notch signaling pathways, thus regulating EMT in LECs.

TGFβ signaling occupies a key position in the signaling networks that regulates EMT. It includes canonical Smad signaling and non-canonical Smad independent signaling pathways. Previous studies have reported that ERK1/2 signaling is involved in TGFβ-induced EMT in LECs and other types of cells (1619). The activation of ERK1/2 signaling promotes TGFβ-induced EMT and ECM components deposition, whereas the inactivation of ERK1/2 inhibits TGFβ-induced EMT effectively (19,20). In the present study, we found that ERK1/2 is rapidly activated by TGFβ2 stimulation, and that MEK1/2 inhibitor U0126 blocks this response completely. Nevertheless, SB431542, a specific inhibitor for the canonical TGFβ/Smad2/3 signaling transduction, has no effect on the activation of ERK1/2 induced by TGFβ2. These data indicate that TGFβ2-induced ERK1/2 activation is independent of the TGFβ/Smad pathway in LECs. In addition, inactivation of ERK1/2 signaling strongly prevents the upregulation of EMT markers induced by TGFβ2. These results suggest that ERK1/2 signaling pathway is a critical mediator for TGFβ induction of EMT in LECs, and ERK1/2 inhibitor can be useful for abrogating EMT phenotype.

It has been reported that non-canonical Smad signaling, such as the p38MAPK and PI3K/AKT pathways, can crosstalk and integrate with the canonical TGFβ/Smad signaling, thereby contributing to EMT (25). To examine whether there is a crosstalk between the non-canonical TGFβ/ERK1/2 signaling and the canonical TGFβ/Smad signaling, the effect of U0126 on the activation of receptor-regulated Smad2 and Smad3 induced by TGFβ2 was investigated. We found that U0126 inhibits the phosphorylation of Smad2 induced by TGFβ2, but cannot inhibit the phosphorylation of Smad3 in LECs. These results suggest that U0126 mediates the canonical TGFβ/Smad signaling by inhibiting the phosphorylation of Smad2. Therefore, there is a crosstalk between the non-canonical TGFβ/ERK1/2 and the canonical TGFβ/Smad signaling in LECs EMT.

Evidence suggests that the Notch signaling pathway is a vital regulator in the induction of EMT during embryonic development, cancer metastasis and various fibrotic diseases (21). Activated Jagged/Notch signaling has been confirmed in a large range of fibrotic diseases developed in the kidney, liver and lung (26). Moreover, our former study found that the Notch signaling pathway is upregulated via canonical TGFβ2/Smad signaling in LECs EMT, while blockade of the Notch pathway with DAPT markedly reverses TGFβ2-induced EMT. In this study, we have shown that U0126 attenuates the TGFβ2-induced upregulation of Jagged-1, Notch-1 and Notch-2, as well as TGFβ2-induced Notch target genes Hes-1 and Hey-1 expression. These results suggest that non-canonical ERK1/2 signaling also contributes to the TGFβ2-induced activation of the Notch pathway in LECs. Inactivation of ERK1/2 with U0126 abrogates TGFβ2-induced EMT partly by suppressing the Jagged/Notch pathway. Furthermore, we observed that blockade of the Notch pathway by DAPT inhibits the TGFβ2-induced activation of the ERK1/2 pathway. This means non-canonical TGFβ/ERK1/2 signaling can be mediated by the Notch pathway inversely in LECs. Collectively, these data indicate that there is a crosstalk between the ERK1/2 signaling and the Notch pathway in LECs EMT.

In summary, our results provide evidence that the TGFβ2-induced activation of ERK1/2 is independent of canonical TGFβ/Smad signaling in human LECs. Inactivation of ERK1/2 signaling with U0126 completely inhibits TGFβ2-induced EMT in LECs. In addition, the blockade of ERK1/2 signaling inhibits the canonical Smad signaling pathway, as well as the Jagged/Notch pathway. We also found that non-canonical TGFβ/ERK1/2 signaling can be mediated by the Notch pathway conversely. Thus, findings of this study suggest that ERK1/2 signaling cross-interacts with the canonical TGFβ/Smad and the Jagged/Notch signaling pathways, thus mediating EMT in LECs. Therefore, ERK inhibitor may have therapeutic value in the prevention and treatment of ASC and PCO.

Acknowledgements

We would like to thank Professor Fu Shang for kindly providing the SRA01/04 human LEC line for this study. The study was funded by the grant from the Guangdong Natural Science Foundation (S2012020010878).

References

1 

McCarty CA and Taylor HR: Recent developments in vision research: light damage in cataract. Invest Ophthalmol Vis Sci. 37:1720–1723. 1996.PubMed/NCBI

2 

Nathu Z, Dwivedi DJ, Reddan JR, Sheardown H, Margetts PJ and West-Mays JA: Temporal changes in MMP mRNA expression in the lens epithelium during anterior subcapsular cataract formation. Exp Eye Res. 88:323–330. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Shin EH, Basson MA, Robinson ML, McAvoy JW and Lovicu FJ: Sprouty is a negative regulator of transforming growth factor β-induced epithelial-to-mesenchymal transition and cataract. Mol Med. 18:861–873. 2012.PubMed/NCBI

4 

Apple DJ, Solomon KD, Tetz MR, et al: Posterior capsule opacification. Surv Ophthalmol. 37:73–116. 1992. View Article : Google Scholar

5 

Hodge WG: Posterior capsule opacification after cataract surgery. Ophthalmology. 105:943–944. 1998. View Article : Google Scholar : PubMed/NCBI

6 

Srinivasan Y, Lovicu FJ and Overbeek PA: Lens-specific expression of transforming growth factor beta1 in transgenic mice causes anterior subcapsular cataracts. J Clin Invest. 101:625–634. 1998. View Article : Google Scholar : PubMed/NCBI

7 

de Iongh RU, Wederell E, Lovicu FJ and McAvoy JW: Transforming growth factor-beta-induced epithelial-mesenchymal transition in the lens: a model for cataract formation. Cells Tissues Organs. 179:43–55. 2005.PubMed/NCBI

8 

Wallentin N, Wickström K and Lundberg C: Effect of cataract surgery on aqueous TGF-beta and lens epithelial cell proliferation. Invest Ophthalmol Vis Sci. 39:1410–1418. 1998.PubMed/NCBI

9 

Meacock WR, Spalton DJ and Stanford MR: Role of cytokines in the pathogenesis of posterior capsule opacification. Br J Ophthalmol. 84:332–336. 2000. View Article : Google Scholar : PubMed/NCBI

10 

Awasthi N, Guo S and Wagner BJ: Posterior capsular opacification: a problem reduced but not yet eradicated. Arch Ophthalmol. 127:555–562. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Allen JB, Davidson MG, Nasisse MP, Fleisher LN and McGahan MC: The lens influences aqueous humor levels of transforming growth factor-beta 2. Graefes Arch Clin Exp Ophthalmol. 236:305–311. 1998. View Article : Google Scholar : PubMed/NCBI

12 

Eldred JA, Dawes LJ and Wormstone IM: The lens as a model for fibrotic disease. Philos Trans R Soc Lond B Biol Sci. 366:1301–1319. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Akhurst RJ and Hata A: Targeting the TGFβ signalling pathway in disease. Nat Rev Drug Discov. 11:790–811. 2012.

14 

Li J, Tang X and Chen X: Comparative effects of TGF-β2/Smad2 and TGF-β2/Smad3 signaling pathways on proliferation, migration, and extracellular matrix production in a human lens cell line. Exp Eye Res. 92:173–179. 2011.

15 

Dawes LJ, Sleeman MA, Anderson IK, Reddan JR and Wormstone IM: TGFbeta/Smad4-dependent and -independent regulation of human lens epithelial cells. Invest Ophthalmol Vis Sci. 50:5318–5327. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Chung EJ, Chun JN, Jung SA, Cho JW and Lee JH: TGF-β-stimulated aberrant expression of class III β-tubulin via the ERK signaling pathway in cultured retinal pigment epithelial cells. Biochem Biophys Res Commun. 415:367–372. 2011.

17 

Chen XF, Zhang HJ, Wang HB, et al: Transforming growth factor-β1 induces epithelial-to-mesenchymal transition in human lung cancer cells via PI3K/Akt and MEK/Erk1/2 signaling pathways. Mol Biol Rep. 39:3549–3556. 2012.

18 

Aomatsu K, Arao T, Sugioka K, et al: TGF-β induces sustained upregulation of SNAI1 and SNAI2 through Smad and non-Smad pathways in a human corneal epithelial cell line. Invest Ophthalmol Vis Sci. 52:2437–2443. 2011.

19 

Choi J, Park SY and Joo CK: Transforming growth factor-beta1 represses E-cadherin production via slug expression in lens epithelial cells. Invest Ophthalmol Vis Sci. 48:2708–2718. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Xie L, Law BK, Chytil AM, Brown KA, Aakre ME and Moses HL: Activation of the Erk pathway is required for TGF-beta1-induced EMT in vitro. Neoplasia. 6:603–610. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Wang Z, Li Y, Kong D and Sarkar FH: The role of Notch signaling pathway in epithelial-mesenchymal transition (EMT) during development and tumor aggressiveness. Curr Drug Targets. 11:745–751. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Neuzillet C, Tijeras-Raballand A, de Mestier L, Cros J, Faivre S and Raymond E: MEK in cancer and cancer therapy. Pharmacol Ther. 141:160–171. 2013. View Article : Google Scholar

23 

Tanahashi T, Osada S, Yamada A, et al: Extracellular signal-regulated kinase and Akt activation play a critical role in the process of hepatocyte growth factor-induced epithelial-mesenchymal transition. Int J Oncol. 42:556–564. 2013.

24 

Pacheco-Domínguez RL, Palma-Nicolas JP, López E and López-Colomé AM: The activation of MEK-ERK1/2 by glutamate receptor-stimulation is involved in the regulation of RPE proliferation and morphologic transformation. Exp Eye Res. 86:207–219. 2008.PubMed/NCBI

25 

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

26 

Leask A: Targeting the jagged/notch pathway: a new treatment for fibrosis? J Cell Commun Signal. 4:197–198. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2014
Volume 33 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen X, Ye S, Xiao W, Wang W, Luo L and Liu Y: ERK1/2 pathway mediates epithelial-mesenchymal transition by cross-interacting with TGFβ/Smad and Jagged/Notch signaling pathways in lens epithelial cells. Int J Mol Med 33: 1664-1670, 2014
APA
Chen, X., Ye, S., Xiao, W., Wang, W., Luo, L., & Liu, Y. (2014). ERK1/2 pathway mediates epithelial-mesenchymal transition by cross-interacting with TGFβ/Smad and Jagged/Notch signaling pathways in lens epithelial cells. International Journal of Molecular Medicine, 33, 1664-1670. https://doi.org/10.3892/ijmm.2014.1723
MLA
Chen, X., Ye, S., Xiao, W., Wang, W., Luo, L., Liu, Y."ERK1/2 pathway mediates epithelial-mesenchymal transition by cross-interacting with TGFβ/Smad and Jagged/Notch signaling pathways in lens epithelial cells". International Journal of Molecular Medicine 33.6 (2014): 1664-1670.
Chicago
Chen, X., Ye, S., Xiao, W., Wang, W., Luo, L., Liu, Y."ERK1/2 pathway mediates epithelial-mesenchymal transition by cross-interacting with TGFβ/Smad and Jagged/Notch signaling pathways in lens epithelial cells". International Journal of Molecular Medicine 33, no. 6 (2014): 1664-1670. https://doi.org/10.3892/ijmm.2014.1723