Skip to main content
Top
Published in: Molecular Cancer 1/2015

Open Access 01-12-2015 | Research

RETRACTED ARTICLE: HGF and TGFβ1 differently influenced Wwox regulatory function on Twist program for mesenchymal-epithelial transition in bone metastatic versus parental breast carcinoma cells

Authors: Paola Bendinelli, Paola Maroni, Emanuela Matteucci, Maria Alfonsina Desiderio

Published in: Molecular Cancer | Issue 1/2015

Login to get access

Abstract

Background

Much effort has been devoted to determining how metastatic cells and microenvironment reciprocally interact. However, the role of biological stimuli of microenvironment in controlling molecular events in bone metastasis from breast carcinoma for mesenchymal-epithelial transition (MET) is largely unknown. The purpose of the present paper was to clarify (1) the influence of hepatocyte-growth factor (HGF) and transforming growth factorβ1 (TGFβ1) on the phenotype of bone-metastatic 1833 and parental MDA-MB231 cells; (2) the hierarchic response of Twist and Snail controlled by Wwox co-factor, that might be critical for the control of 1833-adhesive properties via E-cadherin.

Methods

We studied under HGF and TGFβ1 the gene profiles—responsible for epithelial-mesenchymal transition (EMT), versus the revertant MET phenotype—making the correspondence with 1833 morphology and the relation to HGF-dependent control of TGFβ1 signalling. In particular, the activation of Twist program and the underlying molecular mechanisms were investigated, considering the role of endogenous and exogenous Wwox with siRNAWWOX and the expression vector transfection, to clarify whether Twist affected E-cadherin transactivation through a network of transcription factors and regulators.

Results

HGF and TGFβ1 oppositely affected the expression of Wwox in 1833 cells. Under HGF, endogenous Wwox decreased concomitant with Twist access to nuclei and its phosphorylation via PI3K/Akt pathway. Twist activated by HGF did not influence the gene profile through an E-box mechanism, but participated in the interplay of PPARγ/Ets1/NF-kB-transcription factors, triggering E-cadherin transactivation. Altogether, HGF conferred MET phenotype to 1833 cells, even if this was transient since followed by TGFβ1-signalling activation. TGFβ1 induced Snail in both the cell lines, with E-cadherin down-regulation only in 1833 cells because in MDA-MB231 cells E-cadherin was practically absent. Exogenous Wwox activated metastatic HIF-1, with Twist as co-factor.

Conclusions

HGF and TGFβ1 of bone-metastasis microenvironment acted co-ordinately, influencing non redundant pathways regulated by Twist program or Snail-transcription factor, with reversible MET switch. This process implicated different roles for Wwox in the various steps of the metastatic process including colonization, with microenvironmental/exogenous Wwox that activated HIF-1, important for E-cadherin expression. Interfering with the Twist program by targeting the pre-metastatic niche stimuli could be an effective anti-bone metastasis therapy.
Appendix
Available only for authorised users
Literature
1.
go back to reference Lowery FJ, Yu D. Growth factor signaling in metastasis: current understanding and future opportunities. Cancer Metastasis Rev. 2012;31:479–91.PubMed Lowery FJ, Yu D. Growth factor signaling in metastasis: current understanding and future opportunities. Cancer Metastasis Rev. 2012;31:479–91.PubMed
2.
go back to reference Previdi S, Maroni P, Matteucci E, Broggini M, Bendinelli P, Desiderio MA. Interaction between human-breast cancer metastasis and bone microenvironment through activated hepatocyte growth factor/Met and β-catenin/Wnt pathways. Eur J Cancer. 2010;46:1679–91.PubMed Previdi S, Maroni P, Matteucci E, Broggini M, Bendinelli P, Desiderio MA. Interaction between human-breast cancer metastasis and bone microenvironment through activated hepatocyte growth factor/Met and β-catenin/Wnt pathways. Eur J Cancer. 2010;46:1679–91.PubMed
3.
go back to reference Sceneay J, Smyth MJ, Möller A. The pre-metastatic niche: finding common ground. Cancer Metastasis Rev. 2013;32:449–64.PubMed Sceneay J, Smyth MJ, Möller A. The pre-metastatic niche: finding common ground. Cancer Metastasis Rev. 2013;32:449–64.PubMed
4.
go back to reference Gao D, Vahdat LT, Wong S, Chang JC, Mittal V. Microenvironmental regulation of epithelial-mesenchymal transitions in cancer. Cancer Res 2012;72:4883–89. Gao D, Vahdat LT, Wong S, Chang JC, Mittal V. Microenvironmental regulation of epithelial-mesenchymal transitions in cancer. Cancer Res 2012;72:4883–89.
5.
go back to reference Olechnowicz SW, Edwards CM. Contributions of the host microenvironment to cancer-induced bone disease. Cancer Res. 2014;74:1625–31.PubMedPubMedCentral Olechnowicz SW, Edwards CM. Contributions of the host microenvironment to cancer-induced bone disease. Cancer Res. 2014;74:1625–31.PubMedPubMedCentral
6.
go back to reference Caramel J, Papadogeorgakis E, Hill L, Browne GJ, Richard G, Wierinckx A, et al. A switch in the expression of embryonic EMT-inducers drives the development of malignant melanoma. Cancer Cell. 2013;24:466–80.PubMed Caramel J, Papadogeorgakis E, Hill L, Browne GJ, Richard G, Wierinckx A, et al. A switch in the expression of embryonic EMT-inducers drives the development of malignant melanoma. Cancer Cell. 2013;24:466–80.PubMed
7.
go back to reference De Craene B, Berx G. Regulatory networks defining EMT during cancer initiation and progression. Nature Rev Cancer. 2013;13:97–110. De Craene B, Berx G. Regulatory networks defining EMT during cancer initiation and progression. Nature Rev Cancer. 2013;13:97–110.
8.
go back to reference Yang MH, Wu MZ, Chiou SH, Chen PM, Chang SY, Liu CJ, et al. Direct regulation of TWIST by HIF-1alpha promotes metastasis. Nat Cell Biol. 2008;10:295–305.PubMed Yang MH, Wu MZ, Chiou SH, Chen PM, Chang SY, Liu CJ, et al. Direct regulation of TWIST by HIF-1alpha promotes metastasis. Nat Cell Biol. 2008;10:295–305.PubMed
9.
go back to reference Birchmeier W, Behrens J. Cadherin expression in carcinomas: role in the formation of cell junctions and prevention of invasiveness. Biochim Biophys Acta. 1994;1198:11–26.PubMed Birchmeier W, Behrens J. Cadherin expression in carcinomas: role in the formation of cell junctions and prevention of invasiveness. Biochim Biophys Acta. 1994;1198:11–26.PubMed
10.
go back to reference Vleminckx K, Vakaet Jr L, Mareel M, Fiers W, van Roy F. Genetic manipulation of E-cadherin expression by epithelial tumor cells reveals an invasion suppressor role. Cell. 1991;66:107–19.PubMed Vleminckx K, Vakaet Jr L, Mareel M, Fiers W, van Roy F. Genetic manipulation of E-cadherin expression by epithelial tumor cells reveals an invasion suppressor role. Cell. 1991;66:107–19.PubMed
11.
go back to reference Gheldof A, Berx G. Cadherins and epithelial-to-mesenchymal transition. Prog Mol Biol Transl. 2013;116:317–36. Gheldof A, Berx G. Cadherins and epithelial-to-mesenchymal transition. Prog Mol Biol Transl. 2013;116:317–36.
12.
go back to reference Stankic M, Pavlovic S, Chin Y, Brogi E, Padua D, Norton L, et al. TGF-β-Id1 signaling opposes Twist1 and promotes metastatic colonization via a mesenchymal-to-epithelial transition. Cell Rep. 2013;5:1228–42.PubMedPubMedCentral Stankic M, Pavlovic S, Chin Y, Brogi E, Padua D, Norton L, et al. TGF-β-Id1 signaling opposes Twist1 and promotes metastatic colonization via a mesenchymal-to-epithelial transition. Cell Rep. 2013;5:1228–42.PubMedPubMedCentral
13.
go back to reference Tsai JH, Donaher JL, Murphy DA, Chau S, Yang J. Spatiotemporal regulation of epithelial-mesenchymal transition is essential for squamous cell carcinoma metastasis. Cancer Cell. 2012;22:725–36.PubMedPubMedCentral Tsai JH, Donaher JL, Murphy DA, Chau S, Yang J. Spatiotemporal regulation of epithelial-mesenchymal transition is essential for squamous cell carcinoma metastasis. Cancer Cell. 2012;22:725–36.PubMedPubMedCentral
14.
go back to reference Joyce JA, Pollard JW. Microenvironmental regulation of metastasis. Nat Rev Cancer. 2009;9:239–52.PubMed Joyce JA, Pollard JW. Microenvironmental regulation of metastasis. Nat Rev Cancer. 2009;9:239–52.PubMed
15.
go back to reference Waning DL, Guise TA. Molecular mechanisms of bone metastasis and associated muscle weakness. Clin Cancer Res. 2014;20:3071–7.PubMedPubMedCentral Waning DL, Guise TA. Molecular mechanisms of bone metastasis and associated muscle weakness. Clin Cancer Res. 2014;20:3071–7.PubMedPubMedCentral
16.
go back to reference Li DM, Feng YM. Signaling mechanism of cell adhesion molecules in breast cancer metastasis: potential therapeutic targets. Breast Cancer Res Treat. 2011;128:7–21.PubMed Li DM, Feng YM. Signaling mechanism of cell adhesion molecules in breast cancer metastasis: potential therapeutic targets. Breast Cancer Res Treat. 2011;128:7–21.PubMed
17.
go back to reference Wang H, Yu C, Gao X, Welte T, Muscarella AM, Tian L, et al. The osteogenic niche promotes early-stage bone colonization of disseminated breast cancer cells. Cancer Cell. 2015;27:193–210.PubMedPubMedCentral Wang H, Yu C, Gao X, Welte T, Muscarella AM, Tian L, et al. The osteogenic niche promotes early-stage bone colonization of disseminated breast cancer cells. Cancer Cell. 2015;27:193–210.PubMedPubMedCentral
18.
go back to reference Mousa SA. Cell adhesion molecules: potential therapeutic & diagnostic implications. Mol Biotechnol. 2008;38:33–40.PubMed Mousa SA. Cell adhesion molecules: potential therapeutic & diagnostic implications. Mol Biotechnol. 2008;38:33–40.PubMed
19.
go back to reference Matteucci E, Maroni P, Luzzati A, Perrucchini G, Bendinelli P, Desiderio MA. Bone metastatic process of breast cancer involves methylation state affecting E-cadherin expression through TAZ and WWOX nuclear effectors. Eur J Cancer. 2013;49:231–44.PubMed Matteucci E, Maroni P, Luzzati A, Perrucchini G, Bendinelli P, Desiderio MA. Bone metastatic process of breast cancer involves methylation state affecting E-cadherin expression through TAZ and WWOX nuclear effectors. Eur J Cancer. 2013;49:231–44.PubMed
20.
go back to reference Kang Y, Siegel PM, Shu W, Drobnjak M, Kakonen SM, Cordón-Cardo C, et al. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell. 2003;3:537–49.PubMed Kang Y, Siegel PM, Shu W, Drobnjak M, Kakonen SM, Cordón-Cardo C, et al. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell. 2003;3:537–49.PubMed
21.
go back to reference Abdeen SK, Salah Z, Maly B, Smith Y, Tufail R, Abu-Odeh M, et al. Wwox inactivation enhances mammary tumorigenesis. Oncogene. 2011;30:3900–6.PubMed Abdeen SK, Salah Z, Maly B, Smith Y, Tufail R, Abu-Odeh M, et al. Wwox inactivation enhances mammary tumorigenesis. Oncogene. 2011;30:3900–6.PubMed
22.
go back to reference Chang JY, He RY, Lin HP, Hsu LJ, Lai FJ, Hong Q, et al. Signaling from membrane receptors to tumor suppressor WW domain-containing oxidoreductase. Exp Biol Med. 2010;235:796–804. Chang JY, He RY, Lin HP, Hsu LJ, Lai FJ, Hong Q, et al. Signaling from membrane receptors to tumor suppressor WW domain-containing oxidoreductase. Exp Biol Med. 2010;235:796–804.
23.
go back to reference Li MY, Lai FJ, Hsu LJ, Lo CP, Cheng CL, Lin SR, et al. Dramatic co-activation of WWOX/WOX1 with CREB and NFkB in delayed loss of small dorsal root ganglion neurons upon sciatic nerve transection in rats. PLoS One. 2009;4, e7820.PubMedPubMedCentral Li MY, Lai FJ, Hsu LJ, Lo CP, Cheng CL, Lin SR, et al. Dramatic co-activation of WWOX/WOX1 with CREB and NFkB in delayed loss of small dorsal root ganglion neurons upon sciatic nerve transection in rats. PLoS One. 2009;4, e7820.PubMedPubMedCentral
24.
go back to reference Matteucci E, Bendinelli P, Desiderio MA. Nuclear localization of active HGF receptor Met in aggressive MDA-MB231 breast carcinoma cells. Carcinogenesis. 2009;30:937–45.PubMed Matteucci E, Bendinelli P, Desiderio MA. Nuclear localization of active HGF receptor Met in aggressive MDA-MB231 breast carcinoma cells. Carcinogenesis. 2009;30:937–45.PubMed
25.
go back to reference Maroni P, Bendinelli P, Matteucci E, Locatelli A, Nakamura T, Scita G, et al. Osteolytic bone metastasis is hampered by impinging on the interplay among autophagy, anoikis and ossification. Cell Death Dis. 2014;5, e1005.PubMedPubMedCentral Maroni P, Bendinelli P, Matteucci E, Locatelli A, Nakamura T, Scita G, et al. Osteolytic bone metastasis is hampered by impinging on the interplay among autophagy, anoikis and ossification. Cell Death Dis. 2014;5, e1005.PubMedPubMedCentral
26.
go back to reference Franco HL, Casasnovas J, Rodríguez-Medina JR, Cadilla CL. Redundant or separate entities?-roles of Twist1 and Twist2 as molecular switches during gene transcription. Nucleic Acids Res. 2010;39:1177–86.PubMedPubMedCentral Franco HL, Casasnovas J, Rodríguez-Medina JR, Cadilla CL. Redundant or separate entities?-roles of Twist1 and Twist2 as molecular switches during gene transcription. Nucleic Acids Res. 2010;39:1177–86.PubMedPubMedCentral
27.
go back to reference Croset M, Goehrig D, Frackowiak A, Bonnelye E, Ansieau S, Puisieux A, et al. TWIST1 expression in breast cancer cells facilitates bone metastasis formation. J Bone Mineral Res. 2014;29:1886–99. Croset M, Goehrig D, Frackowiak A, Bonnelye E, Ansieau S, Puisieux A, et al. TWIST1 expression in breast cancer cells facilitates bone metastasis formation. J Bone Mineral Res. 2014;29:1886–99.
28.
go back to reference Xue G, Hemmings BA. Phosphorylation of basic helix-loop-helix transcription factor Twist in development and disease. Biochem Soc Trans. 2012;40:90–3.PubMed Xue G, Hemmings BA. Phosphorylation of basic helix-loop-helix transcription factor Twist in development and disease. Biochem Soc Trans. 2012;40:90–3.PubMed
29.
go back to reference Zhang K, Corsa CA, Ponik SM, Prior JL, Piwnica-Worms D, Eliceiri KW, et al. The collagen receptor discoidin domain receptor 2 stabilizes SNAIL1 to facilitate breast cancer metastasis. Nat Cell Biol. 2013;15:677–87.PubMedPubMedCentral Zhang K, Corsa CA, Ponik SM, Prior JL, Piwnica-Worms D, Eliceiri KW, et al. The collagen receptor discoidin domain receptor 2 stabilizes SNAIL1 to facilitate breast cancer metastasis. Nat Cell Biol. 2013;15:677–87.PubMedPubMedCentral
30.
go back to reference Wrana JL, Attisano L, Carcamo J, Zentella A, Doody J, Laiho M, et al. TGF beta signals through a heteromeric protein kinase receptor complex. Cell. 1992;13:1003–14. Wrana JL, Attisano L, Carcamo J, Zentella A, Doody J, Laiho M, et al. TGF beta signals through a heteromeric protein kinase receptor complex. Cell. 1992;13:1003–14.
31.
go back to reference Cui Q, Fu S, Li Z. Hepatocyte growth factor inhibits TGF-β1-induced myofibroblast differentiation in tendon fibroblasts: role of AMPK signaling pathway. J Physiol Sci. 2013;63:163–70.PubMed Cui Q, Fu S, Li Z. Hepatocyte growth factor inhibits TGF-β1-induced myofibroblast differentiation in tendon fibroblasts: role of AMPK signaling pathway. J Physiol Sci. 2013;63:163–70.PubMed
32.
go back to reference Bendinelli P, Maroni P, Matteucci E, Luzzati A, Perrucchini G, Desiderio MA. Hypoxia inducible factor-1 is activated by transcriptional co-activator with PDZ binding motif (TAZ) versus WWdomain-containing oxidoreductase (WWOX) in hypoxic microenvironment of bone metastasis from breast cancer. Eur J Cancer. 2013;49:2608–18.PubMed Bendinelli P, Maroni P, Matteucci E, Luzzati A, Perrucchini G, Desiderio MA. Hypoxia inducible factor-1 is activated by transcriptional co-activator with PDZ binding motif (TAZ) versus WWdomain-containing oxidoreductase (WWOX) in hypoxic microenvironment of bone metastasis from breast cancer. Eur J Cancer. 2013;49:2608–18.PubMed
33.
go back to reference Dang CV, Semenza GL. Oncogenic alterations of metabolism. TIBS. 1999;24:68–72.PubMed Dang CV, Semenza GL. Oncogenic alterations of metabolism. TIBS. 1999;24:68–72.PubMed
34.
go back to reference Presser LD, McRae S, Waris G. Activation of TGF-β1 promoter by hepatitis C virus-induced AP-1 and Sp1: role of TGF-β1 in hepatic stellate cell activation and invasion. PLoS One. 2013;8, e56367.PubMedPubMedCentral Presser LD, McRae S, Waris G. Activation of TGF-β1 promoter by hepatitis C virus-induced AP-1 and Sp1: role of TGF-β1 in hepatic stellate cell activation and invasion. PLoS One. 2013;8, e56367.PubMedPubMedCentral
35.
go back to reference Gunasinghe NP, Wells A, Thompson EW, Hugo HJ. Mesenchymal-epithelial transition (MET) as a mechanism for metastatic colonization in breast cancer. Cancer Metastasis. 2012;31:469–78. Gunasinghe NP, Wells A, Thompson EW, Hugo HJ. Mesenchymal-epithelial transition (MET) as a mechanism for metastatic colonization in breast cancer. Cancer Metastasis. 2012;31:469–78.
36.
go back to reference Zhang Y, Hassan MQ, Li ZY, Stein JL, Lian JB, van Wijnen AJ, et al. Intricate gene regulatory networks of helix-loop-helix (HLH) proteins support regulation of bone-tissue related genes during osteoblast differentiation. J Cell Biochem. 2008;105:487–96.PubMedPubMedCentral Zhang Y, Hassan MQ, Li ZY, Stein JL, Lian JB, van Wijnen AJ, et al. Intricate gene regulatory networks of helix-loop-helix (HLH) proteins support regulation of bone-tissue related genes during osteoblast differentiation. J Cell Biochem. 2008;105:487–96.PubMedPubMedCentral
37.
go back to reference Shamir ER, Pappalardo E, Jorgens DM, Coutinho K, Tsai WT, Aziz K, et al. Twist1-induced dissemination preserves epithelial identity and requires E-cadherin. J Cell Biol. 2014;204:839–56.PubMedPubMedCentral Shamir ER, Pappalardo E, Jorgens DM, Coutinho K, Tsai WT, Aziz K, et al. Twist1-induced dissemination preserves epithelial identity and requires E-cadherin. J Cell Biol. 2014;204:839–56.PubMedPubMedCentral
38.
go back to reference Grotegut S, von Schweinitz D, Christofori G, Lehembre F. Hepatocyte growth factor induces cell scattering through MAPK/Erg-1-mediated upregulation of Snail. EMBO J. 2006;25:3534–45. Grotegut S, von Schweinitz D, Christofori G, Lehembre F. Hepatocyte growth factor induces cell scattering through MAPK/Erg-1-mediated upregulation of Snail. EMBO J. 2006;25:3534–45.
39.
go back to reference Kowalczyk AP, Nanes BA. Adherens junction turnover: regulating adhesion through cadherin Endocytosis, degradation, and recycling. In: Harris T, editor. Adherens junctions: from molecular mechanisms to tissue development and disease, Subcellular biochemistry, vol. 60. Dordrecht, The Netherlands: Springer Science- Business Media; 2012. p. 197–222. Kowalczyk AP, Nanes BA. Adherens junction turnover: regulating adhesion through cadherin Endocytosis, degradation, and recycling. In: Harris T, editor. Adherens junctions: from molecular mechanisms to tissue development and disease, Subcellular biochemistry, vol. 60. Dordrecht, The Netherlands: Springer Science- Business Media; 2012. p. 197–222.
40.
go back to reference Maroni P, Matteucci E, Drago L, Banfi G, Bendinelli P, Desiderio MA. Hypoxia induced E-cadherin involving regulators of Hippo pathway due to HIF-1α stabilization/nuclear translocation in bone metastasis from breast carcinoma. Exp Cell Res. 2015;330:287–99.PubMed Maroni P, Matteucci E, Drago L, Banfi G, Bendinelli P, Desiderio MA. Hypoxia induced E-cadherin involving regulators of Hippo pathway due to HIF-1α stabilization/nuclear translocation in bone metastasis from breast carcinoma. Exp Cell Res. 2015;330:287–99.PubMed
41.
go back to reference Jin C, Ge L, Ding X, Chen Y, Zhu H, Ward T, et al. PKA-mediated protein phosphorylation regulates ezrin-WWOX interaction. Biochem Biophys Res Commun. 2006;341:784–91.PubMed Jin C, Ge L, Ding X, Chen Y, Zhu H, Ward T, et al. PKA-mediated protein phosphorylation regulates ezrin-WWOX interaction. Biochem Biophys Res Commun. 2006;341:784–91.PubMed
42.
go back to reference Kingsley LA, Fournier PGJ, Chirgwin JM, Guise TA. Molecular biology of bone metastasis. Mol Cancer Ther. 2007;6:2609–17.PubMed Kingsley LA, Fournier PGJ, Chirgwin JM, Guise TA. Molecular biology of bone metastasis. Mol Cancer Ther. 2007;6:2609–17.PubMed
43.
go back to reference Käkönen S-M, Selander KS, Chirgwin JM, Yin JJ, Burns S, Rankin WA, et al. Transforming growth factor-β stimulates parathyroid hormone-related protein and osteolytic metastases via Smad and mitogen-activated protein kinase signaling pathways. J Biol Chem. 2002;277:24571–8.PubMed Käkönen S-M, Selander KS, Chirgwin JM, Yin JJ, Burns S, Rankin WA, et al. Transforming growth factor-β stimulates parathyroid hormone-related protein and osteolytic metastases via Smad and mitogen-activated protein kinase signaling pathways. J Biol Chem. 2002;277:24571–8.PubMed
44.
go back to reference Chiechi A, Waning DL, Stayrook KR, Buijs JT, Guise TA, Mohammad KS. Role of TGF-β in breast cancer bone metastases. Adv Biosci Biotechnol. 2013;4:15–30.PubMedPubMedCentral Chiechi A, Waning DL, Stayrook KR, Buijs JT, Guise TA, Mohammad KS. Role of TGF-β in breast cancer bone metastases. Adv Biosci Biotechnol. 2013;4:15–30.PubMedPubMedCentral
45.
go back to reference Ikushima H, Miyazano K. TGFbeta signaling: a complex web in cancer progression. Nat Rev Cancer. 2010;10:415–24.PubMed Ikushima H, Miyazano K. TGFbeta signaling: a complex web in cancer progression. Nat Rev Cancer. 2010;10:415–24.PubMed
46.
go back to reference Ridolfi E, Matteucci E, Maroni P, Desiderio MA. Inhibitory effect of HGF on invasiveness of aggressive MDA-MB231 breast carcinoma cells, and role of HDACs. Br J Cancer. 2008;99:937–45. Ridolfi E, Matteucci E, Maroni P, Desiderio MA. Inhibitory effect of HGF on invasiveness of aggressive MDA-MB231 breast carcinoma cells, and role of HDACs. Br J Cancer. 2008;99:937–45.
47.
go back to reference Maroni P, Matteucci E, Luzzati A, Perrucchini G, Bendinelli P, Desiderio MA. Nuclear co-localization and functional interaction of COX2 and HIF-1α characterize bone metastasis of human breast carcinoma. Breast Cancer Res Treat. 2011;129:433–50.PubMed Maroni P, Matteucci E, Luzzati A, Perrucchini G, Bendinelli P, Desiderio MA. Nuclear co-localization and functional interaction of COX2 and HIF-1α characterize bone metastasis of human breast carcinoma. Breast Cancer Res Treat. 2011;129:433–50.PubMed
48.
go back to reference Zhu M, Doshi S, Gisleskog PO, Oliner KS, Perez Ruixo JJ, Loh E, et al. Population pharmacokinetics of rilotumumab, a fully human monoclonal antibody against hepatocyte growth factor, in cancer patients. J Pharm Sci. 2014;103:328–36.PubMed Zhu M, Doshi S, Gisleskog PO, Oliner KS, Perez Ruixo JJ, Loh E, et al. Population pharmacokinetics of rilotumumab, a fully human monoclonal antibody against hepatocyte growth factor, in cancer patients. J Pharm Sci. 2014;103:328–36.PubMed
49.
go back to reference Kong FM, Anscher MS, Murase T, Abbott BD, Iglehart JD, Jirtle RL. Elevated plasma transforming growth factor-beta 1 levels in breast cancer patients decrease after surgical removal of the tumor. Annals Surgery. 1995;222:155–62. Kong FM, Anscher MS, Murase T, Abbott BD, Iglehart JD, Jirtle RL. Elevated plasma transforming growth factor-beta 1 levels in breast cancer patients decrease after surgical removal of the tumor. Annals Surgery. 1995;222:155–62.
50.
go back to reference Peyton KJ, Yu Y, Yates B, Shebib AR, Liu X-M, Wang H, et al. Compound C inhibits vascular smooth muscle cell proliferation and migration via an AMP-activated protein kinase-independent fashion. J Pharmacol Exp Ther. 2011;338:476–84.PubMedPubMedCentral Peyton KJ, Yu Y, Yates B, Shebib AR, Liu X-M, Wang H, et al. Compound C inhibits vascular smooth muscle cell proliferation and migration via an AMP-activated protein kinase-independent fashion. J Pharmacol Exp Ther. 2011;338:476–84.PubMedPubMedCentral
51.
go back to reference Cheng J-C, Klausen C, Leung PCK. Hypoxia-inducible factor 1 alpha mediates epidermal growth factor-induced down-regulation of E-cadherin expression and cell invasion in human ovarian cancer cells. Cancer Lett. 2013;329:197–206.PubMed Cheng J-C, Klausen C, Leung PCK. Hypoxia-inducible factor 1 alpha mediates epidermal growth factor-induced down-regulation of E-cadherin expression and cell invasion in human ovarian cancer cells. Cancer Lett. 2013;329:197–206.PubMed
Metadata
Title
RETRACTED ARTICLE: HGF and TGFβ1 differently influenced Wwox regulatory function on Twist program for mesenchymal-epithelial transition in bone metastatic versus parental breast carcinoma cells
Authors
Paola Bendinelli
Paola Maroni
Emanuela Matteucci
Maria Alfonsina Desiderio
Publication date
01-12-2015
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2015
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-015-0389-y

Other articles of this Issue 1/2015

Molecular Cancer 1/2015 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