Skip to main content
Top
Published in: Molecular Cancer 1/2018

Open Access 01-12-2018 | Research

Wnt-signaling enhances neural crest migration of melanoma cells and induces an invasive phenotype

Authors: Tobias Sinnberg, Mitchell P. Levesque, Jelena Krochmann, Phil F. Cheng, Kristian Ikenberg, Francisco Meraz-Torres, Heike Niessner, Claus Garbe, Christian Busch

Published in: Molecular Cancer | Issue 1/2018

Login to get access

Abstract

Background

During embryonic development Wnt family members and bone morphogenetic proteins (BMPs) cooperatively induce epithelial-mesenchymal transition (EMT) in the neural crest. Wnt and BMPs are reactivated during malignant transformation in melanoma. We previously demonstrated that the BMP-antagonist noggin blocked the EMT phenotype of melanoma cells in the neural crest and malignant invasion of melanoma cells in the chick embryo; vice-versa, malignant invasion was induced in human melanocytes in vivo by pre-treatment with BMP-2.

Results

Although there are conflicting results in the literature about the role of β-catenin for invasion of melanoma cells, we found Wnt/β-catenin signaling to be analogously important for the EMT-like phenotype of human metastatic melanoma cells in the neural crest and during invasion: β-catenin was frequently expressed at the invasive front of human primary melanomas and Wnt3a expression was inversely correlated with survival of melanoma patients. Accordingly, cytoplasmic β-catenin levels were increased during invasion of melanoma cells in the rhombencephalon of the chick embryo. Fibroblast derived Wnt3a reduced melanoma cell adhesion and enhanced migration, while the β-catenin inhibitor PKF115–584 increased adhesion and reduced migration in vitro and in the chick embryonic neural crest environment in vivo. Similarly, knockdown of β-catenin impaired intradermal melanoma cell invasion and PKF115–584 efficiently reduced liver metastasis in a chick chorioallantoic membrane model. Our observations were accompanied by specific alterations in gene expression which are linked to overall survival of melanoma patients.

Conclusion

We present a novel role for Wnt-signaling in neural crest like melanoma cell invasion and metastasis, stressing the crucial role of embryonic EMT-inducing neural crest signaling for the spreading of malignant melanoma.
Appendix
Available only for authorised users
Literature
1.
go back to reference Steventon B, Araya C, Linker C, Kuriyama S, Mayor R. Differential requirements of BMP and Wnt signalling during gastrulation and neurulation define two steps in neural crest induction. Development. 2009;136:771–9.CrossRefPubMedPubMedCentral Steventon B, Araya C, Linker C, Kuriyama S, Mayor R. Differential requirements of BMP and Wnt signalling during gastrulation and neurulation define two steps in neural crest induction. Development. 2009;136:771–9.CrossRefPubMedPubMedCentral
2.
go back to reference Endo Y, Ishiwata-Endo H, Yamada KM. Extracellular matrix protein anosmin promotes neural crest formation and regulates FGF, BMP, and WNT activities. Dev Cell. 2012;23:305–16.CrossRefPubMedPubMedCentral Endo Y, Ishiwata-Endo H, Yamada KM. Extracellular matrix protein anosmin promotes neural crest formation and regulates FGF, BMP, and WNT activities. Dev Cell. 2012;23:305–16.CrossRefPubMedPubMedCentral
4.
go back to reference Garcia-Castro MI, Marcelle C, Bronner-Fraser M. Ectodermal Wnt function as a neural crest inducer. Sci (80- ). 2002;297:848–51. Garcia-Castro MI, Marcelle C, Bronner-Fraser M. Ectodermal Wnt function as a neural crest inducer. Sci (80- ). 2002;297:848–51.
5.
go back to reference Mica Y, Lee G, Chambers SM, Tomishima MJ, Studer L. Modeling neural crest induction, melanocyte specification, and disease-related pigmentation defects in hESCs and patient-specific iPSCs. Cell Rep. 2013;3:1140–52.CrossRefPubMedPubMedCentral Mica Y, Lee G, Chambers SM, Tomishima MJ, Studer L. Modeling neural crest induction, melanocyte specification, and disease-related pigmentation defects in hESCs and patient-specific iPSCs. Cell Rep. 2013;3:1140–52.CrossRefPubMedPubMedCentral
6.
go back to reference Greenburg G, Hay ED. Epithelia suspended in collagen gels can lose polarity and express characteristics of migrating mesenchymal cells. J Cell Biol. 1982;95:333–9.CrossRefPubMed Greenburg G, Hay ED. Epithelia suspended in collagen gels can lose polarity and express characteristics of migrating mesenchymal cells. J Cell Biol. 1982;95:333–9.CrossRefPubMed
8.
go back to reference Schriek G, Oppitz M, Busch C, Just L, Drews U. Human SK-Mel 28 melanoma cells resume neural crest cell migration after transplantation into the chick embryo. Melanoma Res. 2005;15:225–34.CrossRefPubMed Schriek G, Oppitz M, Busch C, Just L, Drews U. Human SK-Mel 28 melanoma cells resume neural crest cell migration after transplantation into the chick embryo. Melanoma Res. 2005;15:225–34.CrossRefPubMed
9.
go back to reference Oppitz M, Pintaske J, Kehlbach R, Schick F, Schriek G, Busch C. Magnetic resonance imaging of iron-oxide labeled SK-Mel 28 human melanoma cells in the chick embryo using a clinical whole body MRI scanner. MAGMA. 2007;20:1–9.CrossRefPubMed Oppitz M, Pintaske J, Kehlbach R, Schick F, Schriek G, Busch C. Magnetic resonance imaging of iron-oxide labeled SK-Mel 28 human melanoma cells in the chick embryo using a clinical whole body MRI scanner. MAGMA. 2007;20:1–9.CrossRefPubMed
10.
go back to reference Oppitz M, Busch C, Schriek G, Metzger M, Just L, Drews U. Non-malignant migration of B16 mouse melanoma cells in the neural crest and invasive growth in the eye cup of the chick embryo. Melanoma Res. 2007;17:17–30.CrossRefPubMed Oppitz M, Busch C, Schriek G, Metzger M, Just L, Drews U. Non-malignant migration of B16 mouse melanoma cells in the neural crest and invasive growth in the eye cup of the chick embryo. Melanoma Res. 2007;17:17–30.CrossRefPubMed
11.
go back to reference Busch C, Oppitz M, Sailer MH, Just L, Metzger M, Drews U. BMP-2-dependent integration of adult mouse subventricular stem cells into the neural crest of chick and quail embryos. J Cell Sci. 2006;119:4467–74.CrossRefPubMed Busch C, Oppitz M, Sailer MH, Just L, Metzger M, Drews U. BMP-2-dependent integration of adult mouse subventricular stem cells into the neural crest of chick and quail embryos. J Cell Sci. 2006;119:4467–74.CrossRefPubMed
12.
go back to reference Rothhammer T, Poser I, Soncin F, Bataille F, Moser M, Bosserhoff A-K. Bone morphogenic proteins are overexpressed in malignant melanoma and promote cell invasion and migration. Cancer Res. 2005;65:448–56.PubMed Rothhammer T, Poser I, Soncin F, Bataille F, Moser M, Bosserhoff A-K. Bone morphogenic proteins are overexpressed in malignant melanoma and promote cell invasion and migration. Cancer Res. 2005;65:448–56.PubMed
13.
go back to reference Busch C, Drews U, Garbe C, Eisele SR, Oppitz M. Neural crest cell migration of mouse B16-F1 melanoma cells transplanted into the chick embryo is inhibited by the BMP-antagonist noggin. Int J Oncol. 2007;31:1367–78.PubMed Busch C, Drews U, Garbe C, Eisele SR, Oppitz M. Neural crest cell migration of mouse B16-F1 melanoma cells transplanted into the chick embryo is inhibited by the BMP-antagonist noggin. Int J Oncol. 2007;31:1367–78.PubMed
14.
go back to reference Busch C, Drews U, Eisele SR, Garbe C, Oppitz M. Noggin blocks invasive growth of murine B16-F1 melanoma cells in the optic cup of the chick embryo. Int J Cancer. 2008;122:526–33.CrossRefPubMed Busch C, Drews U, Eisele SR, Garbe C, Oppitz M. Noggin blocks invasive growth of murine B16-F1 melanoma cells in the optic cup of the chick embryo. Int J Cancer. 2008;122:526–33.CrossRefPubMed
15.
16.
go back to reference Sinnberg T, Menzel M, Kaesler S, Biedermann T, Sauer B, Nahnsen S, et al. Suppression of casein kinase 1alpha in melanoma cells induces a switch in beta-catenin signaling to promote metastasis. Cancer Res. 2010;70:6999–7009.CrossRefPubMed Sinnberg T, Menzel M, Kaesler S, Biedermann T, Sauer B, Nahnsen S, et al. Suppression of casein kinase 1alpha in melanoma cells induces a switch in beta-catenin signaling to promote metastasis. Cancer Res. 2010;70:6999–7009.CrossRefPubMed
17.
go back to reference Sinnberg T, Menzel M, Ewerth D, Sauer B, Schwarz M, Schaller M, et al. β-catenin signaling increases during melanoma progression and promotes tumor cell survival and chemoresistance. PLoS One. 2011;6:e23429. Gottardi C, editorCrossRefPubMedPubMedCentral Sinnberg T, Menzel M, Ewerth D, Sauer B, Schwarz M, Schaller M, et al. β-catenin signaling increases during melanoma progression and promotes tumor cell survival and chemoresistance. PLoS One. 2011;6:e23429. Gottardi C, editorCrossRefPubMedPubMedCentral
18.
go back to reference White RM, Cech J, Ratanasirintrawoot S, Lin CY, Rahl PB, Burke CJ, et al. DHODH modulates transcriptional elongation in the neural crest and melanoma. Nature. 2011;471:518–22.CrossRefPubMedPubMedCentral White RM, Cech J, Ratanasirintrawoot S, Lin CY, Rahl PB, Burke CJ, et al. DHODH modulates transcriptional elongation in the neural crest and melanoma. Nature. 2011;471:518–22.CrossRefPubMedPubMedCentral
19.
go back to reference Mathew LK, Sengupta SS, Ladu J, Andreasen EA, Tanguay RL. Crosstalk between AHR and Wnt signaling through R-Spondin1 impairs tissue regeneration in zebrafish. FASEB J. 2008;22:3087–96.CrossRefPubMedPubMedCentral Mathew LK, Sengupta SS, Ladu J, Andreasen EA, Tanguay RL. Crosstalk between AHR and Wnt signaling through R-Spondin1 impairs tissue regeneration in zebrafish. FASEB J. 2008;22:3087–96.CrossRefPubMedPubMedCentral
20.
go back to reference O’Donnell EF, Saili KS, Koch DC, Kopparapu PR, Farrer D, Bisson WH, et al. The anti-inflammatory drug leflunomide is an agonist of the aryl hydrocarbon receptor. PLoS One. 2010;5:e13128. Rodrigues-Lima F, editorCrossRefPubMedPubMedCentral O’Donnell EF, Saili KS, Koch DC, Kopparapu PR, Farrer D, Bisson WH, et al. The anti-inflammatory drug leflunomide is an agonist of the aryl hydrocarbon receptor. PLoS One. 2010;5:e13128. Rodrigues-Lima F, editorCrossRefPubMedPubMedCentral
21.
go back to reference Chen Y, Huang Q, Zhou H, Wang Y, Hu X, Li T. Inhibition of canonical WNT/β-catenin signaling is involved in leflunomide (LEF)-mediated cytotoxic effects on renal carcinoma cells. Oncotarget. 2016;7:50401–16.PubMedPubMedCentral Chen Y, Huang Q, Zhou H, Wang Y, Hu X, Li T. Inhibition of canonical WNT/β-catenin signaling is involved in leflunomide (LEF)-mediated cytotoxic effects on renal carcinoma cells. Oncotarget. 2016;7:50401–16.PubMedPubMedCentral
22.
go back to reference Kawajiri K, Kobayashi Y, Ohtake F, Ikuta T, Matsushima Y, Mimura J, et al. Aryl hydrocarbon receptor suppresses intestinal carcinogenesis in ApcMin/+ mice with natural ligands. Proc Natl Acad Sci U S A. 2009;106:13481–6.CrossRefPubMedPubMedCentral Kawajiri K, Kobayashi Y, Ohtake F, Ikuta T, Matsushima Y, Mimura J, et al. Aryl hydrocarbon receptor suppresses intestinal carcinogenesis in ApcMin/+ mice with natural ligands. Proc Natl Acad Sci U S A. 2009;106:13481–6.CrossRefPubMedPubMedCentral
23.
go back to reference Lucero OM, Dawson DW, Moon RT, Chien AJ. A re-evaluation of the “oncogenic” nature of Wnt/beta-catenin signaling in melanoma and other cancers. Curr Oncol Rep. 2010;12:314–8.CrossRefPubMedPubMedCentral Lucero OM, Dawson DW, Moon RT, Chien AJ. A re-evaluation of the “oncogenic” nature of Wnt/beta-catenin signaling in melanoma and other cancers. Curr Oncol Rep. 2010;12:314–8.CrossRefPubMedPubMedCentral
24.
go back to reference Sharma A, Sen JM. Molecular basis for the tissue specificity of β-catenin oncogenesis. Oncogene. 2013;32:1901–9.CrossRefPubMed Sharma A, Sen JM. Molecular basis for the tissue specificity of β-catenin oncogenesis. Oncogene. 2013;32:1901–9.CrossRefPubMed
26.
go back to reference Arozarena I, Bischof H, Gilby D, Belloni B, Dummer R, Wellbrock C. In melanoma, beta-catenin is a suppressor of invasion. Oncogene. 2011;30:4531–43. Arozarena I, Bischof H, Gilby D, Belloni B, Dummer R, Wellbrock C. In melanoma, beta-catenin is a suppressor of invasion. Oncogene. 2011;30:4531–43.
27.
go back to reference Chien AJ, Moore EC, Lonsdorf AS, Kulikauskas RM, Rothberg BG, Berger AJ, et al. Activated Wnt/beta-catenin signaling in melanoma is associated with decreased proliferation in patient tumors and a murine melanoma model. Proc Natl Acad Sci U S A. 2009;106:1193–8.CrossRefPubMedPubMedCentral Chien AJ, Moore EC, Lonsdorf AS, Kulikauskas RM, Rothberg BG, Berger AJ, et al. Activated Wnt/beta-catenin signaling in melanoma is associated with decreased proliferation in patient tumors and a murine melanoma model. Proc Natl Acad Sci U S A. 2009;106:1193–8.CrossRefPubMedPubMedCentral
28.
go back to reference Kageshita T, Hamby CV, Ishihara T, Matsumoto K, Saida T, Ono T. Loss of beta-catenin expression associated with disease progression in malignant melanoma. Br J Dermatol. 2001;145:210–6.CrossRefPubMed Kageshita T, Hamby CV, Ishihara T, Matsumoto K, Saida T, Ono T. Loss of beta-catenin expression associated with disease progression in malignant melanoma. Br J Dermatol. 2001;145:210–6.CrossRefPubMed
29.
go back to reference Maelandsmo GM, Holm R, Nesland JM, Fodstad O, Florenes VA. Reduced beta-catenin expression in the cytoplasm of advanced-stage superficial spreading malignant melanoma. Clin Cancer Res. 2003;9:3383–8.PubMed Maelandsmo GM, Holm R, Nesland JM, Fodstad O, Florenes VA. Reduced beta-catenin expression in the cytoplasm of advanced-stage superficial spreading malignant melanoma. Clin Cancer Res. 2003;9:3383–8.PubMed
30.
go back to reference Damsky WE, Curley DP, Santhanakrishnan M, Rosenbaum LE, Platt JT, Gould Rothberg BE, et al. β-catenin signaling controls metastasis in Braf-activated Pten-deficient melanomas. Cancer Cell. 2011;20:741–54.CrossRefPubMedPubMedCentral Damsky WE, Curley DP, Santhanakrishnan M, Rosenbaum LE, Platt JT, Gould Rothberg BE, et al. β-catenin signaling controls metastasis in Braf-activated Pten-deficient melanomas. Cancer Cell. 2011;20:741–54.CrossRefPubMedPubMedCentral
31.
go back to reference Murakami T, Toda S, Fujimoto M, Ohtsuki M, Byers HR, Etoh T, et al. Constitutive activation of Wnt/beta-catenin signaling pathway in migration-active melanoma cells: role of LEF-1 in melanoma with increased metastatic potential. Biochem Biophys Res Commun. 2001;288:8–15.CrossRefPubMed Murakami T, Toda S, Fujimoto M, Ohtsuki M, Byers HR, Etoh T, et al. Constitutive activation of Wnt/beta-catenin signaling pathway in migration-active melanoma cells: role of LEF-1 in melanoma with increased metastatic potential. Biochem Biophys Res Commun. 2001;288:8–15.CrossRefPubMed
32.
go back to reference Zuidervaart W, Pavey S, van Nieuwpoort FA, Packer L, Out C, Maat W, et al. Expression of Wnt5a and its downstream effector beta-catenin in uveal melanoma. Melanoma Res. 2007;17:380–6.CrossRefPubMed Zuidervaart W, Pavey S, van Nieuwpoort FA, Packer L, Out C, Maat W, et al. Expression of Wnt5a and its downstream effector beta-catenin in uveal melanoma. Melanoma Res. 2007;17:380–6.CrossRefPubMed
33.
go back to reference Grossmann AH, Yoo JH, Clancy J, Sorensen LK, Sedgwick A, Tong Z, et al. The small GTPase ARF6 stimulates β-catenin transcriptional activity during WNT5A-mediated melanoma invasion and metastasis. Sci Signal. 2013;6:ra14.CrossRefPubMedPubMedCentral Grossmann AH, Yoo JH, Clancy J, Sorensen LK, Sedgwick A, Tong Z, et al. The small GTPase ARF6 stimulates β-catenin transcriptional activity during WNT5A-mediated melanoma invasion and metastasis. Sci Signal. 2013;6:ra14.CrossRefPubMedPubMedCentral
34.
go back to reference Vaid M, Prasad R, Sun Q, Katiyar SK. Silymarin Targets ?-catenin signaling in blocking migration/invasion of human melanoma cells. PLoS One. 2011;6:e23000.CrossRefPubMedPubMedCentral Vaid M, Prasad R, Sun Q, Katiyar SK. Silymarin Targets ?-catenin signaling in blocking migration/invasion of human melanoma cells. PLoS One. 2011;6:e23000.CrossRefPubMedPubMedCentral
35.
go back to reference Wang W, Runkle KB, Terkowski SM, Ekaireb RI, Witze ES. Protein Depalmitoylation is induced by Wnt5a and promotes polarized cell behavior. J Biol Chem. 2015;290:15707–16.CrossRefPubMedPubMedCentral Wang W, Runkle KB, Terkowski SM, Ekaireb RI, Witze ES. Protein Depalmitoylation is induced by Wnt5a and promotes polarized cell behavior. J Biol Chem. 2015;290:15707–16.CrossRefPubMedPubMedCentral
36.
go back to reference Weeraratna AT, Jiang Y, Hostetter G, Rosenblatt K, Duray P, Bittner M, et al. Wnt5a signaling directly affects cell motility and invasion of metastatic melanoma. Cancer Cell. 2002;1:279–88.CrossRefPubMed Weeraratna AT, Jiang Y, Hostetter G, Rosenblatt K, Duray P, Bittner M, et al. Wnt5a signaling directly affects cell motility and invasion of metastatic melanoma. Cancer Cell. 2002;1:279–88.CrossRefPubMed
37.
go back to reference Dissanayake SK, Wade M, Johnson CE, O’Connell MP, Leotlela PD, French AD, et al. The Wnt5A/protein kinase C pathway mediates motility in melanoma cells via the inhibition of metastasis suppressors and initiation of an epithelial to mesenchymal transition. J Biol Chem. 2007;282:17259–71.CrossRefPubMedPubMedCentral Dissanayake SK, Wade M, Johnson CE, O’Connell MP, Leotlela PD, French AD, et al. The Wnt5A/protein kinase C pathway mediates motility in melanoma cells via the inhibition of metastasis suppressors and initiation of an epithelial to mesenchymal transition. J Biol Chem. 2007;282:17259–71.CrossRefPubMedPubMedCentral
38.
go back to reference Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature. 2015;523:231–5.CrossRefPubMed Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature. 2015;523:231–5.CrossRefPubMed
39.
go back to reference Busch C, Krochmann J, Drews U. Human melanoma cells in the rhombencephalon of the chick embryo: a novel model for brain metastasis. Exp Dermatol. 2012;21:944–7.CrossRefPubMed Busch C, Krochmann J, Drews U. Human melanoma cells in the rhombencephalon of the chick embryo: a novel model for brain metastasis. Exp Dermatol. 2012;21:944–7.CrossRefPubMed
40.
go back to reference Krochmann J, Sinnberg T, Meier F, Garbe C, Busch C. Melanoma cells in distinct growth phases retain specific invasive qualities during brain metastasis in vivo. Pigment Cell Melanoma Res. 2012;25:113–4.CrossRefPubMed Krochmann J, Sinnberg T, Meier F, Garbe C, Busch C. Melanoma cells in distinct growth phases retain specific invasive qualities during brain metastasis in vivo. Pigment Cell Melanoma Res. 2012;25:113–4.CrossRefPubMed
41.
go back to reference Ossowski L, Reich E. Experimental model for quantitative study of metastasis. Cancer Res. 1980;40:2300–9.PubMed Ossowski L, Reich E. Experimental model for quantitative study of metastasis. Cancer Res. 1980;40:2300–9.PubMed
43.
go back to reference Aschermann I, Noor S, Venturelli S, Sinnberg T, Mnich CD, Busch C. Extracorporal shock waves activate migration, proliferation and inflammatory pathways in fibroblasts and Keratinocytes, and improve wound healing in an open-label, single-arm study in patients with therapy-refractory chronic leg ulcers. Cell Physiol Biochem. 2017;41:890–906.CrossRefPubMed Aschermann I, Noor S, Venturelli S, Sinnberg T, Mnich CD, Busch C. Extracorporal shock waves activate migration, proliferation and inflammatory pathways in fibroblasts and Keratinocytes, and improve wound healing in an open-label, single-arm study in patients with therapy-refractory chronic leg ulcers. Cell Physiol Biochem. 2017;41:890–906.CrossRefPubMed
44.
go back to reference Reischauer S, Levesque MP, Nüsslein-Volhard C, Sonawane M. Lgl2 executes its function as a tumor suppressor by regulating ErbB signaling in the zebrafish epidermis. PLoS Genet. 2009;5:e1000720. Mullins MC, editorCrossRefPubMedPubMedCentral Reischauer S, Levesque MP, Nüsslein-Volhard C, Sonawane M. Lgl2 executes its function as a tumor suppressor by regulating ErbB signaling in the zebrafish epidermis. PLoS Genet. 2009;5:e1000720. Mullins MC, editorCrossRefPubMedPubMedCentral
45.
go back to reference Drews U, Ebensperger C, Wolf U. An in vitro model of gonad differentiation in the chick embryo. Roller cultures in gas permeable biofoil bags. Anat Embryol (Berl). 1988;178:529–36.CrossRef Drews U, Ebensperger C, Wolf U. An in vitro model of gonad differentiation in the chick embryo. Roller cultures in gas permeable biofoil bags. Anat Embryol (Berl). 1988;178:529–36.CrossRef
46.
go back to reference Sinnberg T, Makino E, Krueger MA, Velic A, Macek B, Rothbauer U, et al. A nexus consisting of Beta-catenin and Stat3 attenuates BRAF inhibitor efficacy and mediates acquired resistance to Vemurafenib. EBioMedicine. 2016;8:132–49.CrossRefPubMedPubMedCentral Sinnberg T, Makino E, Krueger MA, Velic A, Macek B, Rothbauer U, et al. A nexus consisting of Beta-catenin and Stat3 attenuates BRAF inhibitor efficacy and mediates acquired resistance to Vemurafenib. EBioMedicine. 2016;8:132–49.CrossRefPubMedPubMedCentral
47.
go back to reference Hamburger V, Hamilton HL. A series of normal stages in the development of the chick embryo. 1951. Dev Dyn. 1992;195:231–72.CrossRefPubMed Hamburger V, Hamilton HL. A series of normal stages in the development of the chick embryo. 1951. Dev Dyn. 1992;195:231–72.CrossRefPubMed
48.
go back to reference Hoek KS, Schlegel NC, Brafford P, Sucker A, Ugurel S, Kumar R, et al. Metastatic potential of melanomas defined by specific gene expression profiles with no BRAF signature. Pigment Cell Res. 2006;19:290–302.CrossRefPubMed Hoek KS, Schlegel NC, Brafford P, Sucker A, Ugurel S, Kumar R, et al. Metastatic potential of melanomas defined by specific gene expression profiles with no BRAF signature. Pigment Cell Res. 2006;19:290–302.CrossRefPubMed
49.
go back to reference Cheng PF, Dummer R, Levesque MP. Data mining the cancer genome atlas in the era of precision cancer medicine. Swiss Med Wkly. 2015;145:w14183.PubMed Cheng PF, Dummer R, Levesque MP. Data mining the cancer genome atlas in the era of precision cancer medicine. Swiss Med Wkly. 2015;145:w14183.PubMed
50.
go back to reference Cancer Genome Atlas Network {fname}. Genomic classification of Cutaneous melanoma. Cell. 2015;161:1681–96.CrossRef Cancer Genome Atlas Network {fname}. Genomic classification of Cutaneous melanoma. Cell. 2015;161:1681–96.CrossRef
51.
go back to reference Bald T, Quast T, Landsberg J, Rogava M, Glodde N, Lopez-Ramos D, et al. Ultraviolet-radiation-induced inflammation promotes angiotropism and metastasis in melanoma. Nature. 2014;507:109–13.CrossRefPubMed Bald T, Quast T, Landsberg J, Rogava M, Glodde N, Lopez-Ramos D, et al. Ultraviolet-radiation-induced inflammation promotes angiotropism and metastasis in melanoma. Nature. 2014;507:109–13.CrossRefPubMed
52.
go back to reference Qi J, Chen N, Wang J, Siu C-H. Transendothelial migration of melanoma cells involves N-cadherin-mediated adhesion and activation of the beta-catenin signaling pathway. Mol Biol Cell. 2005;16:4386–97.CrossRefPubMedPubMedCentral Qi J, Chen N, Wang J, Siu C-H. Transendothelial migration of melanoma cells involves N-cadherin-mediated adhesion and activation of the beta-catenin signaling pathway. Mol Biol Cell. 2005;16:4386–97.CrossRefPubMedPubMedCentral
53.
go back to reference Søndergaard JN, Nazarian R, Wang Q, Guo D, Hsueh T, Mok S, et al. Differential sensitivity of melanoma cell lines with BRAFV600E mutation to the specific Raf inhibitor PLX4032. J Transl Med. 2010;8:39.CrossRefPubMedPubMedCentral Søndergaard JN, Nazarian R, Wang Q, Guo D, Hsueh T, Mok S, et al. Differential sensitivity of melanoma cell lines with BRAFV600E mutation to the specific Raf inhibitor PLX4032. J Transl Med. 2010;8:39.CrossRefPubMedPubMedCentral
54.
go back to reference Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–23.CrossRefPubMedPubMedCentral Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–23.CrossRefPubMedPubMedCentral
55.
go back to reference Hamid O, Robert C, Daud A, Hodi FS, Hwu W-J, Kefford R, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 2013;369:134–44.CrossRefPubMedPubMedCentral Hamid O, Robert C, Daud A, Hodi FS, Hwu W-J, Kefford R, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 2013;369:134–44.CrossRefPubMedPubMedCentral
56.
go back to reference Forschner A, Eichner F, Amaral T, Keim U, Garbe C, Eigentler TK. Improvement of overall survival in stage IV melanoma patients during 2011-2014: analysis of real-world data in 441 patients of the German central malignant melanoma registry (CMMR). J Cancer Res Clin Oncol. 2017;143:533–40.CrossRefPubMed Forschner A, Eichner F, Amaral T, Keim U, Garbe C, Eigentler TK. Improvement of overall survival in stage IV melanoma patients during 2011-2014: analysis of real-world data in 441 patients of the German central malignant melanoma registry (CMMR). J Cancer Res Clin Oncol. 2017;143:533–40.CrossRefPubMed
57.
go back to reference Poulikakos PI, Zhang C, Bollag G, Shokat KM, Rosen N. RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF. Nature. 2010;464:427–30.CrossRefPubMedPubMedCentral Poulikakos PI, Zhang C, Bollag G, Shokat KM, Rosen N. RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF. Nature. 2010;464:427–30.CrossRefPubMedPubMedCentral
58.
go back to reference Richard G, Dalle S, Monet M-A, Ligier M, Boespflug A, Pommier RM, et al. ZEB1-mediated melanoma cell plasticity enhances resistance to MAPK inhibitors. EMBO Mol Med. 2016;8:1143–61.CrossRefPubMedPubMedCentral Richard G, Dalle S, Monet M-A, Ligier M, Boespflug A, Pommier RM, et al. ZEB1-mediated melanoma cell plasticity enhances resistance to MAPK inhibitors. EMBO Mol Med. 2016;8:1143–61.CrossRefPubMedPubMedCentral
59.
go back to reference Garrido C, Paco L, Romero I, Berruguilla E, Stefansky J, Collado A, et al. MHC class I molecules act as tumor suppressor genes regulating the cell cycle gene expression, invasion and intrinsic tumorigenicity of melanoma cells. Carcinogenesis. 2012;33:687–93.CrossRefPubMed Garrido C, Paco L, Romero I, Berruguilla E, Stefansky J, Collado A, et al. MHC class I molecules act as tumor suppressor genes regulating the cell cycle gene expression, invasion and intrinsic tumorigenicity of melanoma cells. Carcinogenesis. 2012;33:687–93.CrossRefPubMed
60.
go back to reference Forschner A, Niessner H, Möller Y, Horak P, Fröhlich M, Warsow G, et al. Genomics of immunotherapy-associated Hyperprogressors—letter. Clin Cancer Res. 2017;23:6374–5.CrossRefPubMed Forschner A, Niessner H, Möller Y, Horak P, Fröhlich M, Warsow G, et al. Genomics of immunotherapy-associated Hyperprogressors—letter. Clin Cancer Res. 2017;23:6374–5.CrossRefPubMed
61.
go back to reference Newgreen DF, Minichiello J. Control of epitheliomesenchymal transformation. I. Events in the onset of neural crest cell migration are separable and inducible by protein kinase inhibitors. Dev Biol. 1995;170:91–101.CrossRefPubMed Newgreen DF, Minichiello J. Control of epitheliomesenchymal transformation. I. Events in the onset of neural crest cell migration are separable and inducible by protein kinase inhibitors. Dev Biol. 1995;170:91–101.CrossRefPubMed
62.
go back to reference Newgreen DF, Minichiello J. Control of epitheliomesenchymal transformation. II. Cross-modulation of cell adhesion and cytoskeletal systems in embryonic neural cells. Dev Biol. 1996;176:300–12.CrossRefPubMed Newgreen DF, Minichiello J. Control of epitheliomesenchymal transformation. II. Cross-modulation of cell adhesion and cytoskeletal systems in embryonic neural cells. Dev Biol. 1996;176:300–12.CrossRefPubMed
63.
go back to reference Brown K, Yang P, Salvador D, Kulikauskas R, Ruohola-Baker H, Robitaille AM, et al. WNT/β-catenin signaling regulates mitochondrial activity to alter the oncogenic potential of melanoma in a PTEN-dependent manner. Oncogene. 2017;36:3119–36.CrossRefPubMedPubMedCentral Brown K, Yang P, Salvador D, Kulikauskas R, Ruohola-Baker H, Robitaille AM, et al. WNT/β-catenin signaling regulates mitochondrial activity to alter the oncogenic potential of melanoma in a PTEN-dependent manner. Oncogene. 2017;36:3119–36.CrossRefPubMedPubMedCentral
64.
go back to reference Dobroff AS, Wang H, Melnikova VO, Villares GJ, Zigler M, Huang L, et al. Silencing cAMP-response element-binding protein (CREB) identifies CYR61 as a tumor suppressor gene in melanoma. J Biol Chem. 2009;284:26194–206.CrossRefPubMedPubMedCentral Dobroff AS, Wang H, Melnikova VO, Villares GJ, Zigler M, Huang L, et al. Silencing cAMP-response element-binding protein (CREB) identifies CYR61 as a tumor suppressor gene in melanoma. J Biol Chem. 2009;284:26194–206.CrossRefPubMedPubMedCentral
65.
66.
go back to reference Wellbrock C, Arozarena I. Microphthalmia-associated transcription factor in melanoma development and MAP-kinase pathway targeted therapy. Pigment Cell Melanoma Res. 2015;28:390–406.CrossRefPubMedPubMedCentral Wellbrock C, Arozarena I. Microphthalmia-associated transcription factor in melanoma development and MAP-kinase pathway targeted therapy. Pigment Cell Melanoma Res. 2015;28:390–406.CrossRefPubMedPubMedCentral
67.
go back to reference Fane ME, Chhabra Y, Hollingsworth DEJ, Simmons JL, Spoerri L, Oh TG, et al. NFIB mediates BRN2 driven melanoma cell migration and invasion through regulation of EZH2 and MITF. EBioMedicine. 2017;16:63–75.CrossRefPubMedPubMedCentral Fane ME, Chhabra Y, Hollingsworth DEJ, Simmons JL, Spoerri L, Oh TG, et al. NFIB mediates BRN2 driven melanoma cell migration and invasion through regulation of EZH2 and MITF. EBioMedicine. 2017;16:63–75.CrossRefPubMedPubMedCentral
68.
go back to reference Wang X, Liu Y, Chen H, Mei L, He C, Jiang L, et al. LEF-1 regulates Tyrosinase gene transcription in vitro. PLoS One. Public Libr Sci. 2015;10:e0143142.CrossRefPubMedPubMedCentral Wang X, Liu Y, Chen H, Mei L, He C, Jiang L, et al. LEF-1 regulates Tyrosinase gene transcription in vitro. PLoS One. Public Libr Sci. 2015;10:e0143142.CrossRefPubMedPubMedCentral
Metadata
Title
Wnt-signaling enhances neural crest migration of melanoma cells and induces an invasive phenotype
Authors
Tobias Sinnberg
Mitchell P. Levesque
Jelena Krochmann
Phil F. Cheng
Kristian Ikenberg
Francisco Meraz-Torres
Heike Niessner
Claus Garbe
Christian Busch
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2018
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-018-0773-5

Other articles of this Issue 1/2018

Molecular Cancer 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