open access

Vol 26, No 1 (2019)
Original articles — Basic science and experimental cardiology
Submitted: 2017-06-06
Accepted: 2018-01-15
Published online: 2018-01-25
Get Citation

Silencing of c-Ski augments TGF-b1-induced epithelial-mesenchymal transition in cardiomyocyte H9C2 cells

Jia Ling1, Zhenrong Cai1, Wei Jin2, Xiaohua Zhuang1, Lihong Kan1, Fei Wang1, Xiaolei Ye3
·
Pubmed: 29570207
·
Cardiol J 2019;26(1):66-76.
Affiliations
  1. Department of Medical Oncology, Fudan University Pudong Hospital, Shanghai, China
  2. The First People’s Hospital of Shanghai, China
  3. Department of Pharmacology, Ningbo Institute of Medical Sciences, Ningbo University, Ningbo, China

open access

Vol 26, No 1 (2019)
Original articles — Basic science and experimental cardiology
Submitted: 2017-06-06
Accepted: 2018-01-15
Published online: 2018-01-25

Abstract

Background: The shRNA lentiviral vector was constructed to silence c-Ski expression in cardiac mus- 

cle cells, with the aim of exploring the role of c-Ski in transforming growth factor b1 (TGF-b1)-induced epithelial-mesenchymal transitions (EMT) in H9C2 cells.
Methods: Real-time polymerase chain reaction (RT-PCR) and western blot were used to detect c-Ski ex- pression at protein and messenger ribonucleic acid (mRNA) levels in 5 different cell lines. Then, lentiviral vector was constructed to silence or overexpress c-Ski in H9C2 cells. MTT and/or soft agar assay and tran- swell assay were used to detect cell proliferation and migration, respectively. The expression levels of c-Ski under different concentrations of TGF-b1 stimulation were detected by RT-qPCR and immunocytochemi- cal analysis. In the presence or absence of TGF-b1 stimulation, the proteins’ expression levels of a-SMA, FN and E-cadherin, which are closely correlated with the process of EMT, were measured by western blot after c-Ski silencing or overexpression. Meanwhile, the effect of c-Ski on Samd3 phosphorylation with TGF-b1 stimulation was investigated. 

Results: There is a high expression of c-Ski at protein and mRNA levels in H9C2 cell line, which first demonstrated the presence of c-Ski expression in H9C2 cells. Overexpression of c-Ski significantly increased H9C2 cell proliferation. The ability of c-Ski gene silencing to suppress cell proliferation was gradually enhanced, and inhibition efficiency was the highest after 6 to 7 d of transfection. Moreover, H9C2 cells with c-Ski knockdown gained significantly aggressive invasive potential when compared with the control group. TGF-b1 stimulation could dose-independently reduce c-Ski expression in H9C2 cells and lead to obvious down-regulated expression of E-cadherin. Interestingly, c-Ski could restore E-cadherin expression while suppressing a-SMA and/or FN expression stimulated by TGF-b1. How- ever, shRNA-induced c-Ski knockdown aggravated only the TGF-b1-induced EMT. Moreover, c-Ski- -shRNA also promoted the phosphorylation of Samd3 induced by TGF-b1. 

Conclusions: c-Ski expression in cardiac muscle cells could be down-regulated by TGF-b1. Silencing of c-Ski gene was accompanied by down-regulation of E-cadherin, up-regulation of a-SMA and/or FN and Smad3 phosphorylation induced by TGF-b1, promoting EMT process. Therefore, c-Ski may be closely associated with TGF-b1-induced EMT and play an important role in cardiac fibrosis develop- ment and progression. 

Abstract

Background: The shRNA lentiviral vector was constructed to silence c-Ski expression in cardiac mus- 

cle cells, with the aim of exploring the role of c-Ski in transforming growth factor b1 (TGF-b1)-induced epithelial-mesenchymal transitions (EMT) in H9C2 cells.
Methods: Real-time polymerase chain reaction (RT-PCR) and western blot were used to detect c-Ski ex- pression at protein and messenger ribonucleic acid (mRNA) levels in 5 different cell lines. Then, lentiviral vector was constructed to silence or overexpress c-Ski in H9C2 cells. MTT and/or soft agar assay and tran- swell assay were used to detect cell proliferation and migration, respectively. The expression levels of c-Ski under different concentrations of TGF-b1 stimulation were detected by RT-qPCR and immunocytochemi- cal analysis. In the presence or absence of TGF-b1 stimulation, the proteins’ expression levels of a-SMA, FN and E-cadherin, which are closely correlated with the process of EMT, were measured by western blot after c-Ski silencing or overexpression. Meanwhile, the effect of c-Ski on Samd3 phosphorylation with TGF-b1 stimulation was investigated. 

Results: There is a high expression of c-Ski at protein and mRNA levels in H9C2 cell line, which first demonstrated the presence of c-Ski expression in H9C2 cells. Overexpression of c-Ski significantly increased H9C2 cell proliferation. The ability of c-Ski gene silencing to suppress cell proliferation was gradually enhanced, and inhibition efficiency was the highest after 6 to 7 d of transfection. Moreover, H9C2 cells with c-Ski knockdown gained significantly aggressive invasive potential when compared with the control group. TGF-b1 stimulation could dose-independently reduce c-Ski expression in H9C2 cells and lead to obvious down-regulated expression of E-cadherin. Interestingly, c-Ski could restore E-cadherin expression while suppressing a-SMA and/or FN expression stimulated by TGF-b1. How- ever, shRNA-induced c-Ski knockdown aggravated only the TGF-b1-induced EMT. Moreover, c-Ski- -shRNA also promoted the phosphorylation of Samd3 induced by TGF-b1. 

Conclusions: c-Ski expression in cardiac muscle cells could be down-regulated by TGF-b1. Silencing of c-Ski gene was accompanied by down-regulation of E-cadherin, up-regulation of a-SMA and/or FN and Smad3 phosphorylation induced by TGF-b1, promoting EMT process. Therefore, c-Ski may be closely associated with TGF-b1-induced EMT and play an important role in cardiac fibrosis develop- ment and progression. 

Get Citation

Keywords

cardiac muscle cells; c-Ski; proliferation; migration; TGF-b1-induced epithelial-mesenchymal transition

About this article
Title

Silencing of c-Ski augments TGF-b1-induced epithelial-mesenchymal transition in cardiomyocyte H9C2 cells

Journal

Cardiology Journal

Issue

Vol 26, No 1 (2019)

Pages

66-76

Published online

2018-01-25

Page views

6584

Article views/downloads

1812

DOI

10.5603/CJ.a2018.0009

Pubmed

29570207

Bibliographic record

Cardiol J 2019;26(1):66-76.

Keywords

cardiac muscle cells
c-Ski
proliferation
migration
TGF-b1-induced epithelial-mesenchymal transition

Authors

Jia Ling
Zhenrong Cai
Wei Jin
Xiaohua Zhuang
Lihong Kan
Fei Wang
Xiaolei Ye

References (31)
  1. Yu W, Liu Z, An S, et al. The endothelial-mesenchymal transition (EndMT) and tissue regeneration. Curr Stem Cell Res Ther. 2014; 9(3): 196–204.
  2. Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009; 119(6): 1420–1428.
  3. Arciniegas E, Frid MG, Douglas IS, et al. Perspectives on endothelial-to-mesenchymal transition: potential contribution to vascular remodeling in chronic pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol. 2007; 293(1): L1–L8.
  4. Ghosh AK, Bradham WS, Gleaves LA, et al. Genetic deficiency of plasminogen activator inhibitor-1 promotes cardiac fibrosis in aged mice: involvement of constitutive transforming growth factor-beta signaling and endothelial-to-mesenchymal transition. Circulation. 2010; 122(12): 1200–1209.
  5. Li Y, Turck CM, Teumer JK, et al. Unique sequence, ski, in Sloan-Kettering avian retroviruses with properties of a new cell-derived oncogene. J Virol. 1986; 57(3): 1065–1072.
  6. Wong CC, Poon WH, Tsim TY, et al. Gene expressions during the development and sexual differentiation of the olfactory bulb in rats. Brain Res Dev Brain Res. 2000; 119(2): 187–194.
  7. Wang L, Hou Y, Sun Y, et al. c-Ski activates cancer-associated fibroblasts to regulate breast cancer cell invasion. Mol Oncol. 2013; 7(6): 1116–1128.
  8. Cooley BC, Nevado J, Mellad J, et al. TGF-β signaling mediates endothelial-to-mesenchymal transition (EndMT) during vein graft remodeling. Sci Transl Med. 2014; 6(227): 227ra34.
  9. Kong P, Christia P, Frangogiannis NG. The pathogenesis of cardiac fibrosis. Cell Mol Life Sci. 2014; 71(4): 549–574.
  10. Li J, Zhao Li, Yang T, et al. c-Ski inhibits autophagy of vascular smooth muscle cells induced by oxLDL and PDGF. PLoS One. 2014; 9(6): e98902.
  11. Yasui T, Kadono Y, Nakamura M, et al. Regulation of RANKL-induced osteoclastogenesis by TGF-β through molecular interaction between Smad3 and Traf6. J Bone Miner Res. 2011; 26(7): 1447–1456.
  12. Nomura N, Sasamoto S, Ishii S, et al. Isolation of human cDNA clones of ski and the ski-related gene, sno. Nucleic Acids Res. 1989; 17(14): 5489–5500.
  13. Fumagalli S, Doneda L, Nomura N, et al. Expression of the c-ski proto-oncogene in human melanoma cell lines. Melanoma Res. 1993; 3(1): 23–27.
  14. Suzuki H, Yagi K, Kondo M, et al. c-Ski inhibits the TGF-beta signaling pathway through stabilization of inactive Smad complexes on Smad-binding elements. Oncogene. 2004; 23(29): 5068–5076.
  15. Cunnington RH, Nazari M, Dixon IMC. c-Ski, Smurf2, and Arkadia as regulators of TGF-beta signaling: new targets for managing myofibroblast function and cardiac fibrosis. Can J Physiol Pharmacol. 2009; 87(10): 764–772.
  16. Luo K, Stroschein SL, Wang W, et al. The Ski oncoprotein interacts with the Smad proteins to repress TGFbeta signaling. Genes Dev. 1999; 13(17): 2196–2206.
  17. Xu W, Angelis K, Danielpour D, et al. Ski acts as a co-repressor with Smad2 and Smad3 to regulate the response to type beta transforming growth factor. Proc Natl Acad Sci U S A. 2000; 97(11): 5924–5929.
  18. Chen M, Shi J, Zhang W, et al. MiR-23b controls TGF-β1 induced airway smooth muscle cell proliferation via direct targeting of Smad3. Pulm Pharmacol Ther. 2017; 42: 33–42.
  19. Zhu H, Gui Q, Hui X, et al. TGF-β1/Smad3 Signaling Pathway Suppresses Cell Apoptosis in Cerebral Ischemic Stroke Rats. Med Sci Monit. 2017; 23: 366–376.
  20. Liu Z, Gu H, Gan Lu, et al. Reducing Smad3/ATF4 was essential for Sirt1 inhibiting ER stress-induced apoptosis in mice brown adipose tissue. Oncotarget. 2017; 8(6): 9267–9279.
  21. Biernacka A, Cavalera M, Wang J, et al. TGF-β signaling in fibrosis. Growth Factors. 2011; 29(5): 196–202.
  22. Dobaczewski M, Chen W, Frangogiannis NG. Transforming growth factor (TGF)-β signaling in cardiac remodeling. J Mol Cell Cardiol. 2011; 51(4): 600–606.
  23. Li RK, Li G, Mickle DA, et al. Overexpression of transforming growth factor-beta1 and insulin-like growth factor-I in patients with idiopathic hypertrophic cardiomyopathy. Circulation. 1997; 96(3): 874–881.
  24. Li M, Luan F, Zhao Y, et al. Epithelial-mesenchymal transition: An emerging target in tissue fibrosis. Exp Biol Med (Maywood). 2016; 241(1): 1–13.
  25. Liu R, Wang Y, Xiao Y, et al. SnoN as a key regulator of the high glucose-induced epithelial-mesenchymal transition in cells of the proximal tubule. Kidney Blood Press Res. 2012; 35(6): 517–528.
  26. Xiao C, Ogle SA, Schumacher MA, et al. Hedgehog signaling regulates E-cadherin expression for the maintenance of the actin cytoskeleton and tight junctions. Am J Physiol Gastrointest Liver Physiol. 2010; 299(6): G1252–G1265.
  27. Yang H, Zhan L, Yang T, et al. Ski prevents TGF-β-induced EMT and cell invasion by repressing SMAD-dependent signaling in non-small cell lung cancer. Oncol Rep. 2015; 34(1): 87–94.
  28. Choi SS, Diehl AM. Epithelial-to-mesenchymal transitions in the liver. Hepatology. 2009; 50(6): 2007–2013.
  29. Moustakas A, Heldin CH. Signaling networks guiding epithelial-mesenchymal transitions during embryogenesis and cancer progression. Cancer Sci. 2007; 98(10): 1512–1520.
  30. Izaguirre MF, Casco VH. E-cadherin roles in animal biology: A perspective on thyroid hormone-influence. Cell Commun Signal. 2016; 14(1): 27.
  31. Ashrafian H, McKenna WJ, Watkins H. Disease pathways and novel therapeutic targets in hypertrophic cardiomyopathy. Circ Res. 2011; 109(1): 86–96.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

By VM Media Group sp. z o.o., Grupa Via Medica, ul. Świętokrzyska 73, 80–180 Gdańsk, Poland
tel.:+48 58 320 94 94, fax:+48 58 320 94 60, e-mail: viamedica@viamedica.pl