Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2018

Open Access 01-12-2018 | Research

Supervillin promotes epithelial-mesenchymal transition and metastasis of hepatocellular carcinoma in hypoxia via activation of the RhoA/ROCK-ERK/p38 pathway

Authors: Xueran Chen, Shangrong Zhang, Zhen Wang, Fengsong Wang, Xinwang Cao, Quan Wu, Chenggang Zhao, Huihui Ma, Fang Ye, Hongzhi Wang, Zhiyou Fang

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2018

Login to get access

Abstract

Background

Hepatocellular carcinoma (HCC) is one of the most common malignant tumors in the world and metastasis is the leading cause of death associated with HCC. Hypoxia triggers the epithelial-mesenchymal transition (EMT) of cancer cells, which enhances their malignant character and elevates metastatic risk. Supervillin associates tightly with the membrane and cytoskeleton, promoting cell motility, invasiveness, and cell survival. However, the roles of supervillin in HCC metastasis remain unclear.

Methods

Tissue microarray technology was used to immunohistochemically stain for supervillin antibody in 173 HCC tissue specimens and expression levels correlated with the clinicopathological variables. Tumor cell motility and invasiveness, as well as changes in the mRNA expression levels of genes associated with cancer cell EMT, were investigated. The relationship between supervillin and Rho GTPases was examined using Co-IP and GST pull-down.

Results

Hypoxia-induced upregulation of supervillin promoted cancer cell migration and invasion via the activation of the ERK/p38 pathway downstream of RhoA/ROCK signaling. Furthermore, supervillin regulated the expression of EMT genes during hypoxia and accelerated the metastasis of HCC in vivo.

Conclusions

Hypoxia-induced increase in supervillin expression is a significant and independent predictor of cancer metastasis, which leads to poor survival in HCC patients. Our results suggest that supervillin may be a candidate prognostic factor for HCC and a valuable target for therapy.
Appendix
Available only for authorised users
Literature
1.
go back to reference Mendizabal M, Reddy KR. Current management of hepatocellular carcinoma. Med Clin North Am. 2009;93:885–900.CrossRefPubMed Mendizabal M, Reddy KR. Current management of hepatocellular carcinoma. Med Clin North Am. 2009;93:885–900.CrossRefPubMed
2.
go back to reference Kim Y, Ejaz A, Tayal A, Spolverato G, Bridges JF, Anders RA, et al. Temporal trends in population-based death rates associated with chronic liver disease and liver cancer in the United States over the last 30 years. Cancer. 2014;120:3058–65.CrossRefPubMedPubMedCentral Kim Y, Ejaz A, Tayal A, Spolverato G, Bridges JF, Anders RA, et al. Temporal trends in population-based death rates associated with chronic liver disease and liver cancer in the United States over the last 30 years. Cancer. 2014;120:3058–65.CrossRefPubMedPubMedCentral
6.
go back to reference Guro H, Cho JY, Han HS, Yoon YS, Choi Y, Periyasamy M. Current status of laparoscopic liver resection for hepatocellular carcinoma. Clin Mol Hepatol. 2016;22:212–8.CrossRefPubMedPubMedCentral Guro H, Cho JY, Han HS, Yoon YS, Choi Y, Periyasamy M. Current status of laparoscopic liver resection for hepatocellular carcinoma. Clin Mol Hepatol. 2016;22:212–8.CrossRefPubMedPubMedCentral
7.
go back to reference Chen XP, Qiu FZ, Wu ZD, Zhang ZW, Huang ZY, Chen YF, et al. Effects of location and extension of portal vein tumor thrombus on long-term outcomes of surgical treatment for hepatocellular carcinoma. Ann Surg Oncol. 2006;13:940–6.CrossRefPubMed Chen XP, Qiu FZ, Wu ZD, Zhang ZW, Huang ZY, Chen YF, et al. Effects of location and extension of portal vein tumor thrombus on long-term outcomes of surgical treatment for hepatocellular carcinoma. Ann Surg Oncol. 2006;13:940–6.CrossRefPubMed
8.
go back to reference Jiang JF, Lao YC, Yuan BH, Yin J, Liu X, Chen L, et al. Treatment of hepatocellular carcinoma with portal vein tumor thrombus: advances and challenges. Oncotarget. 2017;8:33911–21.PubMedPubMedCentral Jiang JF, Lao YC, Yuan BH, Yin J, Liu X, Chen L, et al. Treatment of hepatocellular carcinoma with portal vein tumor thrombus: advances and challenges. Oncotarget. 2017;8:33911–21.PubMedPubMedCentral
9.
go back to reference Yuki K, Hirohashi S, Sakamoto M, Kanai T, Shimosato Y. Growth and spread of hepatocellular carcinoma. A review of 240 consecutive autopsy cases. Cancer. 1990;66:2174–9.CrossRefPubMed Yuki K, Hirohashi S, Sakamoto M, Kanai T, Shimosato Y. Growth and spread of hepatocellular carcinoma. A review of 240 consecutive autopsy cases. Cancer. 1990;66:2174–9.CrossRefPubMed
12.
go back to reference Talbot LJ, Bhattacharya SD, Kuo PC. Epithelial-mesenchymal transition, the tumor microenvironment, and metastatic behavior of epithelial malignancies. Int J Biochem Mol Biol. 2012;3:117–36.PubMedPubMedCentral Talbot LJ, Bhattacharya SD, Kuo PC. Epithelial-mesenchymal transition, the tumor microenvironment, and metastatic behavior of epithelial malignancies. Int J Biochem Mol Biol. 2012;3:117–36.PubMedPubMedCentral
14.
go back to reference Garvalov BK, Acker T. Implications of oxygen homeostasis for tumor biology and treatment. Adv Exp Med Biol. 2016;903:169–85.CrossRefPubMed Garvalov BK, Acker T. Implications of oxygen homeostasis for tumor biology and treatment. Adv Exp Med Biol. 2016;903:169–85.CrossRefPubMed
15.
go back to reference Somerset H, Witt JP, Kleinschmidt-Demasters BK. Hepatocellular carcinoma metastases to the epidural space. Arch Pathol Lab Med. 2009;133:1975–80.PubMed Somerset H, Witt JP, Kleinschmidt-Demasters BK. Hepatocellular carcinoma metastases to the epidural space. Arch Pathol Lab Med. 2009;133:1975–80.PubMed
16.
go back to reference Giannelli G, Koudelkova P, Dituri F, Mikulits W. Role of epithelial to mesenchymal transition in hepatocellular carcinoma. J Hepatol. 2016;65:798–808.CrossRefPubMed Giannelli G, Koudelkova P, Dituri F, Mikulits W. Role of epithelial to mesenchymal transition in hepatocellular carcinoma. J Hepatol. 2016;65:798–808.CrossRefPubMed
17.
go back to reference Yilmaz M, Christofori G. EMT, the cytoskeleton, and cancer cell invasion. Cancer Metastasis Rev. 2009;28:15–33.CrossRefPubMed Yilmaz M, Christofori G. EMT, the cytoskeleton, and cancer cell invasion. Cancer Metastasis Rev. 2009;28:15–33.CrossRefPubMed
18.
go back to reference Gilkes DM, Xiang L, Lee SJ, Chaturvedi P, Hubbi ME, Wirtz D, et al. Hypoxia-inducible factors mediate coordinated RhoA-ROCK1 expression and signaling in breast cancer cells. Proc Natl Acad Sci U S A. 2014;111:E384–93.CrossRefPubMed Gilkes DM, Xiang L, Lee SJ, Chaturvedi P, Hubbi ME, Wirtz D, et al. Hypoxia-inducible factors mediate coordinated RhoA-ROCK1 expression and signaling in breast cancer cells. Proc Natl Acad Sci U S A. 2014;111:E384–93.CrossRefPubMed
19.
go back to reference Turcotte S, Desrosiers RR, Beliveau R. HIF-1alpha mRNA and protein upregulation involves rho GTPase expression during hypoxia in renal cell carcinoma. J Cell Sci. 2003;116:2247–60.CrossRefPubMed Turcotte S, Desrosiers RR, Beliveau R. HIF-1alpha mRNA and protein upregulation involves rho GTPase expression during hypoxia in renal cell carcinoma. J Cell Sci. 2003;116:2247–60.CrossRefPubMed
20.
go back to reference Heasman SJ, Ridley AJ. Mammalian rho GTPases: new insights into their functions from in vivo studies. Nat Rev Mol Cell Biol. 2008;9:690–701.CrossRefPubMed Heasman SJ, Ridley AJ. Mammalian rho GTPases: new insights into their functions from in vivo studies. Nat Rev Mol Cell Biol. 2008;9:690–701.CrossRefPubMed
21.
go back to reference Aslam M, Schluter KD, Rohrbach S, Rafiq A, Nazli S, Piper HM, et al. Hypoxia-reoxygenation-induced endothelial barrier failure: role of RhoA, Rac1 and myosin light chain kinase. J Physiol. 2013;591:461–73.CrossRefPubMed Aslam M, Schluter KD, Rohrbach S, Rafiq A, Nazli S, Piper HM, et al. Hypoxia-reoxygenation-induced endothelial barrier failure: role of RhoA, Rac1 and myosin light chain kinase. J Physiol. 2013;591:461–73.CrossRefPubMed
22.
go back to reference Qi H, Wang P, Liu C, Li M, Wang S, Huang Y, et al. Involvement of HIF-1alpha in MLCK-dependent endothelial barrier dysfunction in hypoxia. Cell Physiol Biochem. 2011;27:251–62.CrossRefPubMed Qi H, Wang P, Liu C, Li M, Wang S, Huang Y, et al. Involvement of HIF-1alpha in MLCK-dependent endothelial barrier dysfunction in hypoxia. Cell Physiol Biochem. 2011;27:251–62.CrossRefPubMed
23.
go back to reference Vogel S, Wottawa M, Farhat K, Zieseniss A, Schnelle M, Le-Huu S, et al. Prolyl hydroxylase domain (PHD) 2 affects cell migration and F-actin formation via RhoA/rho-associated kinase-dependent cofilin phosphorylation. J Biol Chem. 2010;285:33756–63.CrossRefPubMedPubMedCentral Vogel S, Wottawa M, Farhat K, Zieseniss A, Schnelle M, Le-Huu S, et al. Prolyl hydroxylase domain (PHD) 2 affects cell migration and F-actin formation via RhoA/rho-associated kinase-dependent cofilin phosphorylation. J Biol Chem. 2010;285:33756–63.CrossRefPubMedPubMedCentral
24.
go back to reference Kusuyama J, Bandow K, Shamoto M, Kakimoto K, Ohnishi T, Matsuguchi T. Low intensity pulsed ultrasound (LIPUS) influences the multilineage differentiation of mesenchymal stem and progenitor cell lines through ROCK-cot/Tpl2-MEK-ERK signaling pathway. J Biol Chem. 2014;289:10330–44.CrossRefPubMedPubMedCentral Kusuyama J, Bandow K, Shamoto M, Kakimoto K, Ohnishi T, Matsuguchi T. Low intensity pulsed ultrasound (LIPUS) influences the multilineage differentiation of mesenchymal stem and progenitor cell lines through ROCK-cot/Tpl2-MEK-ERK signaling pathway. J Biol Chem. 2014;289:10330–44.CrossRefPubMedPubMedCentral
25.
go back to reference Robinson JD, Pitcher JA. G protein-coupled receptor kinase 2 (GRK2) is a rho-activated scaffold protein for the ERK MAP kinase cascade. Cell Signal. 2013;25:2831–9.CrossRefPubMed Robinson JD, Pitcher JA. G protein-coupled receptor kinase 2 (GRK2) is a rho-activated scaffold protein for the ERK MAP kinase cascade. Cell Signal. 2013;25:2831–9.CrossRefPubMed
26.
go back to reference El Azreq MA, Kadiri M, Boisvert M, Page N, Tessier PA, Aoudjit F. Discoidin domain receptor 1 promotes Th17 cell migration by activating the RhoA/ROCK/MAPK/ERK signaling pathway. Oncotarget. 2016;7:44975–90.CrossRefPubMed El Azreq MA, Kadiri M, Boisvert M, Page N, Tessier PA, Aoudjit F. Discoidin domain receptor 1 promotes Th17 cell migration by activating the RhoA/ROCK/MAPK/ERK signaling pathway. Oncotarget. 2016;7:44975–90.CrossRefPubMed
27.
go back to reference Kim EK, Choi EJ. Compromised MAPK signaling in human diseases: an update. Arch Toxicol. 2015;89:867–82.CrossRefPubMed Kim EK, Choi EJ. Compromised MAPK signaling in human diseases: an update. Arch Toxicol. 2015;89:867–82.CrossRefPubMed
28.
go back to reference Kim EK, Choi EJ. Pathological roles of MAPK signaling pathways in human diseases. Biochim Biophys Acta. 2010;1802:396–405.CrossRefPubMed Kim EK, Choi EJ. Pathological roles of MAPK signaling pathways in human diseases. Biochim Biophys Acta. 2010;1802:396–405.CrossRefPubMed
29.
go back to reference Bhuwania R, Cornfine S, Fang Z, Kruger M, Luna EJ, Linder S. Supervillin couples myosin-dependent contractility to podosomes and enables their turnover. J Cell Sci. 2012;125:2300–14.CrossRefPubMedPubMedCentral Bhuwania R, Cornfine S, Fang Z, Kruger M, Luna EJ, Linder S. Supervillin couples myosin-dependent contractility to podosomes and enables their turnover. J Cell Sci. 2012;125:2300–14.CrossRefPubMedPubMedCentral
30.
go back to reference Pestonjamasp KN, Pope RK, Wulfkuhle JD, Luna EJ. Supervillin (p205): a novel membrane-associated, F-actin-binding protein in the villin/gelsolin superfamily. J Cell Biol. 1997;139:1255–69.CrossRefPubMedPubMedCentral Pestonjamasp KN, Pope RK, Wulfkuhle JD, Luna EJ. Supervillin (p205): a novel membrane-associated, F-actin-binding protein in the villin/gelsolin superfamily. J Cell Biol. 1997;139:1255–69.CrossRefPubMedPubMedCentral
31.
go back to reference Crowley JL, Smith TC, Fang Z, Takizawa N, Luna EJ. Supervillin reorganizes the actin cytoskeleton and increases invadopodial efficiency. Mol Biol Cell. 2009;20:948–62.CrossRefPubMedPubMedCentral Crowley JL, Smith TC, Fang Z, Takizawa N, Luna EJ. Supervillin reorganizes the actin cytoskeleton and increases invadopodial efficiency. Mol Biol Cell. 2009;20:948–62.CrossRefPubMedPubMedCentral
32.
go back to reference Pope RK, Pestonjamasp KN, Smith KP, Wulfkuhle JD, Strassel CP, Lawrence JB, et al. Cloning, characterization, and chromosomal localization of human superillin (SVIL). Genomics. 1998;52:342–51.CrossRefPubMed Pope RK, Pestonjamasp KN, Smith KP, Wulfkuhle JD, Strassel CP, Lawrence JB, et al. Cloning, characterization, and chromosomal localization of human superillin (SVIL). Genomics. 1998;52:342–51.CrossRefPubMed
33.
go back to reference Oh SW, Pope RK, Smith KP, Crowley JL, Nebl T, Lawrence JB, et al. Archvillin, a muscle-specific isoform of supervillin, is an early expressed component of the costameric membrane skeleton. J Cell Sci. 2003;116:2261–75.CrossRefPubMed Oh SW, Pope RK, Smith KP, Crowley JL, Nebl T, Lawrence JB, et al. Archvillin, a muscle-specific isoform of supervillin, is an early expressed component of the costameric membrane skeleton. J Cell Sci. 2003;116:2261–75.CrossRefPubMed
34.
go back to reference Gangopadhyay SS, Takizawa N, Gallant C, Barber AL, Je HD, Smith TC, et al. Smooth muscle archvillin: a novel regulator of signaling and contractility in vascular smooth muscle. J Cell Sci. 2004;117:5043–57.CrossRefPubMed Gangopadhyay SS, Takizawa N, Gallant C, Barber AL, Je HD, Smith TC, et al. Smooth muscle archvillin: a novel regulator of signaling and contractility in vascular smooth muscle. J Cell Sci. 2004;117:5043–57.CrossRefPubMed
36.
go back to reference Chen X, Yang H, Zhang S, Wang Z, Ye F, Liang C, et al. A novel splice variant of supervillin, SV5, promotes carcinoma cell proliferation and cell migration. Biochem Biophys Res Commun. 2017;482:43–9.CrossRefPubMed Chen X, Yang H, Zhang S, Wang Z, Ye F, Liang C, et al. A novel splice variant of supervillin, SV5, promotes carcinoma cell proliferation and cell migration. Biochem Biophys Res Commun. 2017;482:43–9.CrossRefPubMed
37.
go back to reference Chen Y, Takizawa N, Crowley JL, Oh SW, Gatto CL, Kambara T, et al. F-actin and myosin II binding domains in supervillin. J Biol Chem. 2003;278:46094–106.CrossRefPubMed Chen Y, Takizawa N, Crowley JL, Oh SW, Gatto CL, Kambara T, et al. F-actin and myosin II binding domains in supervillin. J Biol Chem. 2003;278:46094–106.CrossRefPubMed
38.
go back to reference Takizawa N, Smith TC, Nebl T, Crowley JL, Palmieri SJ, Lifshitz LM, et al. Supervillin modulation of focal adhesions involving TRIP6/ZRP-1. J Cell Biol. 2006;174:447–58.CrossRefPubMedPubMedCentral Takizawa N, Smith TC, Nebl T, Crowley JL, Palmieri SJ, Lifshitz LM, et al. Supervillin modulation of focal adhesions involving TRIP6/ZRP-1. J Cell Biol. 2006;174:447–58.CrossRefPubMedPubMedCentral
39.
go back to reference Takizawa N, Ikebe R, Ikebe M, Luna EJ. Supervillin slows cell spreading by facilitating myosin II activation at the cell periphery. J Cell Sci. 2007;120:3792–803.CrossRefPubMed Takizawa N, Ikebe R, Ikebe M, Luna EJ. Supervillin slows cell spreading by facilitating myosin II activation at the cell periphery. J Cell Sci. 2007;120:3792–803.CrossRefPubMed
40.
go back to reference Fang Z, Takizawa N, Wilson KA, Smith TC, Delprato A, Davidson MW, et al. The membrane-associated protein, supervillin, accelerates F-actin-dependent rapid integrin recycling and cell motility. Traffic. 2010;11:782–99.CrossRefPubMedPubMedCentral Fang Z, Takizawa N, Wilson KA, Smith TC, Delprato A, Davidson MW, et al. The membrane-associated protein, supervillin, accelerates F-actin-dependent rapid integrin recycling and cell motility. Traffic. 2010;11:782–99.CrossRefPubMedPubMedCentral
41.
go back to reference Son K, Smith TC, Luna EJ. Supervillin binds the Rac/rho-GEF trio and increases trio-mediated Rac1 activation. Cytoskeleton (Hoboken). 2015;72:47–64.CrossRefPubMedCentral Son K, Smith TC, Luna EJ. Supervillin binds the Rac/rho-GEF trio and increases trio-mediated Rac1 activation. Cytoskeleton (Hoboken). 2015;72:47–64.CrossRefPubMedCentral
42.
go back to reference Ye QH, Zhu WW, Zhang JB, Qin Y, Lu M, Lin GL, et al. GOLM1 modulates EGFR/RTK cell-surface recycling to drive hepatocellular carcinoma metastasis. Cancer Cell. 2016;30:444–58.CrossRefPubMedPubMedCentral Ye QH, Zhu WW, Zhang JB, Qin Y, Lu M, Lin GL, et al. GOLM1 modulates EGFR/RTK cell-surface recycling to drive hepatocellular carcinoma metastasis. Cancer Cell. 2016;30:444–58.CrossRefPubMedPubMedCentral
43.
go back to reference Chen X, Ma H, Wang Z, Zhang S, Yang H, Fang Z. EZH2 Palmitoylation mediated by ZDHHC5 in p53-mutant glioma drives malignant development and progression. Cancer Res. 2017;77:4998–5010.CrossRefPubMed Chen X, Ma H, Wang Z, Zhang S, Yang H, Fang Z. EZH2 Palmitoylation mediated by ZDHHC5 in p53-mutant glioma drives malignant development and progression. Cancer Res. 2017;77:4998–5010.CrossRefPubMed
44.
go back to reference Yamazaki D, Kurisu S, Takenawa T. Regulation of cancer cell motility through actin reorganization. Cancer Sci. 2005;96:379–86.CrossRefPubMed Yamazaki D, Kurisu S, Takenawa T. Regulation of cancer cell motility through actin reorganization. Cancer Sci. 2005;96:379–86.CrossRefPubMed
45.
go back to reference Ibrahim AA, Schmithals C, Kowarz E, Koberle V, Kakoschky B, Pleli T, et al. Hypoxia causes downregulation of dicer in hepatocellular carcinoma, which is required for upregulation of hypoxia-inducible factor 1alpha and epithelial-mesenchymal transition. Clin Cancer Res. 2017;23:3896–905.CrossRefPubMed Ibrahim AA, Schmithals C, Kowarz E, Koberle V, Kakoschky B, Pleli T, et al. Hypoxia causes downregulation of dicer in hepatocellular carcinoma, which is required for upregulation of hypoxia-inducible factor 1alpha and epithelial-mesenchymal transition. Clin Cancer Res. 2017;23:3896–905.CrossRefPubMed
46.
go back to reference Ye LY, Chen W, Bai XL, Xu XY, Zhang Q, Xia XF, et al. Hypoxia-induced epithelial-to-mesenchymal transition in hepatocellular carcinoma induces an immunosuppressive tumor microenvironment to promote metastasis. Cancer Res. 2016;76:818–30.CrossRefPubMed Ye LY, Chen W, Bai XL, Xu XY, Zhang Q, Xia XF, et al. Hypoxia-induced epithelial-to-mesenchymal transition in hepatocellular carcinoma induces an immunosuppressive tumor microenvironment to promote metastasis. Cancer Res. 2016;76:818–30.CrossRefPubMed
47.
go back to reference Gangopadhyay SS, Kengni E, Appel S, Gallant C, Kim HR, Leavis P, et al. Smooth muscle archvillin is an ERK scaffolding protein. J Biol Chem. 2009;284:17607–15.CrossRefPubMedPubMedCentral Gangopadhyay SS, Kengni E, Appel S, Gallant C, Kim HR, Leavis P, et al. Smooth muscle archvillin is an ERK scaffolding protein. J Biol Chem. 2009;284:17607–15.CrossRefPubMedPubMedCentral
48.
go back to reference Smith TC, Fridy PC, Li Y, Basil S, Arjun S, Friesen RM, et al. Supervillin binding to myosin II and synergism with anillin are required for cytokinesis. Mol Biol Cell. 2013;24:3603–19.CrossRefPubMedPubMedCentral Smith TC, Fridy PC, Li Y, Basil S, Arjun S, Friesen RM, et al. Supervillin binding to myosin II and synergism with anillin are required for cytokinesis. Mol Biol Cell. 2013;24:3603–19.CrossRefPubMedPubMedCentral
49.
go back to reference Pawlak G, Helfman DM. Cytoskeletal changes in cell transformation and tumorigenesis. Curr Opin Genet Dev. 2001;11:41–7.CrossRefPubMed Pawlak G, Helfman DM. Cytoskeletal changes in cell transformation and tumorigenesis. Curr Opin Genet Dev. 2001;11:41–7.CrossRefPubMed
50.
51.
go back to reference Stroka KM, Wong BS. Shriver M, Phillip JM, Wirtz D, Kontrogianni-Konstantopoulos a, Konstantopoulos K. Loss of giant obscurins alters breast epithelial cell mechanosensing of matrix stiffness. Oncotarget. 2016;8(33):54004–20.PubMedPubMedCentral Stroka KM, Wong BS. Shriver M, Phillip JM, Wirtz D, Kontrogianni-Konstantopoulos a, Konstantopoulos K. Loss of giant obscurins alters breast epithelial cell mechanosensing of matrix stiffness. Oncotarget. 2016;8(33):54004–20.PubMedPubMedCentral
52.
go back to reference Hetmanski JH, Zindy E, Schwartz JM, Caswell PT. A MAPK-driven feedback loop suppresses Rac activity to promote RhoA-driven Cancer cell invasion. PLoS Comput Biol. 2016;12:e1004909.CrossRefPubMedPubMedCentral Hetmanski JH, Zindy E, Schwartz JM, Caswell PT. A MAPK-driven feedback loop suppresses Rac activity to promote RhoA-driven Cancer cell invasion. PLoS Comput Biol. 2016;12:e1004909.CrossRefPubMedPubMedCentral
53.
go back to reference Chen SH, Hung WC, Wang P, Paul C, Konstantopoulos K. Mesothelin binding to CA125/MUC16 promotes pancreatic cancer cell motility and invasion via MMP-7 activation. Sci Rep. 2013;3:1870.CrossRefPubMedPubMedCentral Chen SH, Hung WC, Wang P, Paul C, Konstantopoulos K. Mesothelin binding to CA125/MUC16 promotes pancreatic cancer cell motility and invasion via MMP-7 activation. Sci Rep. 2013;3:1870.CrossRefPubMedPubMedCentral
54.
go back to reference Wang P, Chen SH, Hung WC, Paul C, Zhu F, Guan PP, Huso DL, Kontrogianni- Konstantopoulos A, Konstantopoulos K. Fluid shear promotes chondrosarcoma cell invasion by activating matrix metalloproteinase 12 via IGF-2 and VEGF signaling pathways. Oncogene. 2015;34(35):4558–69.CrossRefPubMed Wang P, Chen SH, Hung WC, Paul C, Zhu F, Guan PP, Huso DL, Kontrogianni- Konstantopoulos A, Konstantopoulos K. Fluid shear promotes chondrosarcoma cell invasion by activating matrix metalloproteinase 12 via IGF-2 and VEGF signaling pathways. Oncogene. 2015;34(35):4558–69.CrossRefPubMed
Metadata
Title
Supervillin promotes epithelial-mesenchymal transition and metastasis of hepatocellular carcinoma in hypoxia via activation of the RhoA/ROCK-ERK/p38 pathway
Authors
Xueran Chen
Shangrong Zhang
Zhen Wang
Fengsong Wang
Xinwang Cao
Quan Wu
Chenggang Zhao
Huihui Ma
Fang Ye
Hongzhi Wang
Zhiyou Fang
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2018
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-018-0787-2

Other articles of this Issue 1/2018

Journal of Experimental & Clinical Cancer Research 1/2018 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine