A Framework of Major Tumor-Promoting Signal Transduction Pathways Implicated in Melanoma-Fibroblast Dialogue
Abstract
:Simple Summary
Abstract
1. Introduction
2. Transformation of Normal Fibroblasts to Melanoma-Associated Fibroblasts
3. Significance of Altered Intracellular Signal Transduction Pathways in Melanoma and Associated Fibroblasts
3.1. Wnt Signaling
3.2. Hippo Signaling
3.3. TGF-β Signaling
3.4. MAPK Signaling
4. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Miller, A.J.; Mihm, M.C., Jr. Melanoma. N. Engl. J. Med. 2006, 355, 51–65. [Google Scholar] [CrossRef]
- Houghton, A.N.; Polsky, D. Focus on Melanoma. Cancer Cell. 2002, 2, 275–278. [Google Scholar] [CrossRef] [Green Version]
- Arnold, M.; Holterhues, C.; Hollestein, L.M.; Coebergh, J.W.; Nijsten, T.; Pukkala, E.; Holleczek, B.; Tryggvadóttir, L.; Comber, H.; Bento, M.J.; et al. Trends in incidence and predictions of cutaneous melanoma across Europe up to 2015. J. Eur. Acad. Dermatol. Venereol. 2014, 28, 1170–1178. [Google Scholar] [CrossRef] [PubMed]
- Sun, X.; Zhang, N.; Yin, C.; Zhu, B.; Li, X. Ultraviolet radiation and melanomagenesis: From mechanism to immunotherapy. Front. Oncol. 2020, 10, 951. [Google Scholar] [CrossRef] [PubMed]
- Palmieri, G.; Colombino, M.; Casula, M.; Manca, A.; Mandalà, M.; Cossu, A. Italian Melanoma Intergroup (IMI). Molecular pathways in melanomagenesis: What we learned from next-generation sequencing approaches. Curr. Oncol. Rep. 2018, 20, 86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shain, A.H.; Bastian, B.C. From melanocytes to melanomas. Nat. Rev. Cancer 2016, 16, 345–358. [Google Scholar] [CrossRef] [PubMed]
- Eggermont, A.M.; Spatz, A.; Robert, C. Cutaneous melanoma. Lancet 2014, 383, 816–827. [Google Scholar] [CrossRef]
- Hawryluk, E.B.; Tsao, H. Melanoma: Clinical features and genomic insights. Cold Spring Harb Perspect. Med. 2014, 4, a015388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Landi, M.T.; Kanetsky, P.A.; Tsang, S.; Gold, B.; Munroe, D.; Rebbeck, T.; Swoyer, J.; Ter-Minassian, M.; Hedayati, M.; Grossman, L.; et al. MC1R, ASIP, and DNA repair in sporadic and familial melanoma in a mediterranean population. J. Natl. Cancer Inst. 2005, 97, 998–1007. [Google Scholar] [CrossRef] [Green Version]
- Sample, A.; He, Y.Y. Mechanisms and prevention of UV-induced melanoma. Photodermatol. Photoimmunol. Photomed. 2018, 34, 13–24. [Google Scholar] [CrossRef]
- Schulman, J.M.; Fisher, D.E. Indoor ultraviolet tanning and skin cancer: Health risks and opportunities. Curr. Opin. Oncol. 2009, 21, 144–149. [Google Scholar] [CrossRef] [PubMed]
- Sarkar, S.; Gaddameedhi, S. Solar ultraviolet-induced DNA damage response: Melanocytes story in transformation to environmental melanomagenesis. Environ. Mol. Mutagen. 2020, 61, 736–751. [Google Scholar] [CrossRef] [PubMed]
- Cichorek, M.; Wachulska, M.; Stasiewicz, A.; Tymińska, A. Skin melanocytes: Biology and development. Postepy Dermatol. Alergol. 2013, 30, 30–41. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.T.; Choi, B.; Tang, M.S. Melanocytes are deficient in repair of oxidative DNA damage and UV-induced photoproducts. Proc. Natl. Acad. Sci. USA 2010, 107, 12180–12185. [Google Scholar] [CrossRef] [Green Version]
- Khan, A.Q.; Travers, J.B.; Kemp, M.G. Roles of UVA radiation and DNA damage responses in melanoma pathogenesis. Env. Mol. Mutagen. 2018, 59, 438–460. [Google Scholar] [CrossRef]
- Mo, X.; Preston, S.; Zaidi, M.R. Macroenvironment-gene-microenvironment interactions in ultraviolet radiation-induced melanomagenesis. Adv. Cancer Res. 2019, 144, 1–54. [Google Scholar]
- Zhang, S.; Duan, E. Fighting against skin aging: The way from bench to bedside. Cell Transplant. 2018, 27, 729–738. [Google Scholar] [CrossRef]
- Naylor, E.C.; Watson, R.E.; Sherratt, M.J. Molecular aspects of skin ageing. Maturitas 2011, 69, 249–256. [Google Scholar] [CrossRef]
- Yaar, M.; Gilchrest, B.A. Photoageing: Mechanism, prevention and therapy. Br. J. Dermatol. 2007, 157, 874–887. [Google Scholar] [CrossRef]
- Chen, Y.; Chen, Y.; Huang, L.; Yu, J. Evaluation of heparanase and matrix metalloproteinase-9 in patients with cutaneous malignant melanoma. J. Dermatol. 2012, 39, 339–343. [Google Scholar] [CrossRef]
- Rotte, A.; Martinka, M.; Li, G. MMP2 expression is a prognostic marker for primary melanoma patients. Cell. Oncol. (Dordr.) 2012, 35, 207–216. [Google Scholar] [CrossRef]
- Berking, C.; Takemoto, R.; Schaider, H.; Showe, L.; Satyamoorthy, K.; Robbins, P.; Herlyn, M. Transforming growth factor-beta1 increases survival of human melanoma through stroma remodeling. Cancer Res. 2001, 61, 8306–8316. [Google Scholar]
- Kirkpatrick, S.J.; Wang, R.K.; Duncan, D.D.; Kulesz-Martin, M.; Lee, K. Imaging the mechanical stiffness of skin lesions by in vivo acousto-optical elastography. Opt. Express 2006, 14, 9770–9779. [Google Scholar] [CrossRef] [Green Version]
- Miskolczi, Z.; Smith, M.P.; Rowling, E.J.; Ferguson, J.; Barriuso, J.; Wellbrock, C. Collagen abundance controls melanoma phenotypes through lineage-specific microenvironment sensing. Oncogene 2018, 37, 3166–3182. [Google Scholar] [CrossRef] [Green Version]
- Natarajan, V.T.; Ganju, P.; Ramkumar, A.; Grover, R.; Gokhale, R.S. Multifaceted pathways protect human skin from UV radiation. Nat. Chem. Biol. 2014, 10, 542–551. [Google Scholar] [CrossRef]
- Wang, J.X.; Fukunaga-Kalabis, M.; Herlyn, M. Crosstalk in skin: Melanocytes, keratinocytes, stem cells, and melanoma. J. Cell. Commun. Signal. 2016, 10, 191–196. [Google Scholar] [CrossRef] [Green Version]
- Maresca, V.; Flori, E.; Bellei, B.; Aspite, N.; Kovacs, D.; Picardo, M. MC1R stimulation by alpha-MSH induces catalase and promotes its Re-distribution to the cell periphery and dendrites. Pigment Cell. Melanoma Res. 2010, 23, 263–275. [Google Scholar] [CrossRef]
- Brenner, M.; Degitz, K.; Besch, R.; Berking, C. Differential expression of melanoma-associated growth factors in keratinocytes and fibroblasts by ultraviolet A and ultraviolet B radiation. Br. J. Dermatol. 2005, 153, 733–739. [Google Scholar] [CrossRef]
- Berking, C.; Takemoto, R.; Satyamoorthy, K.; Elenitsas, R.; Herlyn, M. Basic fibroblast growth factor and ultraviolet B transform melanocytes in human skin. Am. J. Pathol. 2001, 158, 943–953. [Google Scholar] [CrossRef] [Green Version]
- Kovacs, D.; Cardinali, G.; Aspite, N.; Cota, C.; Luzi, F.; Bellei, B.; Briganti, S.; Amantea, A.; Torrisi, M.R.; Picardo, M. Role of fibroblast-derived growth factors in regulating hyperpigmentation of solar lentigo. Br. J. Dermatol. 2010, 163, 1020–1027. [Google Scholar] [CrossRef]
- Lin, C.B.; Hu, Y.; Rossetti, D.; Chen, N.; David, C.; Slominski, A.; Seiberg, M. Immuno-histochemical evaluation of solar lentigines: The association of KGF/KGFR and other factors with lesion development. J. Dermatol. Sci. 2010, 59, 91–97. [Google Scholar] [CrossRef]
- Passeron, T.; Picardo, M. Melasma, a photoaging disorder. Pigment Cell. Melanoma Res. 2018, 31, 461–465. [Google Scholar] [CrossRef] [Green Version]
- Imokawa, G. Autocrine and paracrine regulation of melanocytes in human skin and in pigmentary disorders. Pigment Cell Res. 2004, 17, 96–110. [Google Scholar] [CrossRef]
- Mellone, M.; Hanley, C.J.; Thirdborough, S.; Mellows, T.; Garcia, E.; Woo, J.; Tod, J.; Frampton, S.; Jenei, V.; Moutasim, K.A.; et al. Induction of fibroblast senescence generates a non-fibrogenic myofibroblast phenotype that differentially impacts on cancer prognosis. Aging (Albany N. Y.) 2016, 9, 114–132. [Google Scholar] [CrossRef] [Green Version]
- Ahmadi, A.; Najafi, M.; Farhood, B.; Mortezaee, K. Transforming growth factor-β signaling: Tumorigenesis and targeting for cancer therapy. J. Cell. Physiol. 2019, 234, 12173–12187. [Google Scholar] [CrossRef]
- Miyazono, K.; Katsuno, Y.; Koinuma, D.; Ehata, S.; Morikawa, M. Intracellular and extracellular TGF-β signaling in cancer: Some recent topics. Front. Med. 2018, 12, 387–411. [Google Scholar] [CrossRef] [Green Version]
- Alspach, E.; Fu, Y.; Stewart, S.A. Senescence and the pro-tumorigenic stroma. Crit. Rev. Oncog. 2013, 18, 549–558. [Google Scholar] [CrossRef] [Green Version]
- Kim, E.; Rebecca, V.; Fedorenko, I.V.; Messina, J.L.; Mathew, R.; Maria-Engler, S.S.; Basanta, D.; Smalley, K.S.; Anderson, A.R. Senescent fibroblasts in melanoma initiation and progression: An integrated theoretical, experimental, and clinical approach. Cancer Res. 2013, 73, 6874–6885. [Google Scholar] [CrossRef] [Green Version]
- Moinfar, F.; Beham, A.; Friedrich, G.; Deutsch, A.; Hrzenjak, A.; Luschin, G.; Tavassoli, F.A. Macro-environment of breast carcinoma: Frequent genetic alterations in the normal appearing skins of patients with breast cancer. Mod. Pathol. 2008, 21, 639–646. [Google Scholar] [CrossRef]
- Balch, C.M.; Soong, S.J.; Gershenwald, J.E.; Thompson, J.F.; Reintgen, D.S.; Cascinelli, N.; Urist, M.; McMasters, K.M.; Ross, M.I.; Kirkwood, J.M.; et al. Prognostic factors analysis of 17,600 melanoma patients: Validation of the american joint committee on cancer melanoma staging system. J. Clin. Oncol. 2001, 19, 3622–3634. [Google Scholar] [CrossRef]
- Coppé, J.P.; Desprez, P.Y.; Krtolica, A.; Campisi, J. The senescence-associated secretory phenotype: The dark side of tumor suppression. Annu. Rev. Pathol. 2010, 5, 99–118. [Google Scholar] [CrossRef] [Green Version]
- Cirri, P.; Chiarugi, P. Cancer associated fibroblasts: The dark side of the coin. Am. J. Cancer Res. 2011, 1, 482–497. [Google Scholar]
- Guido, C.; Whitaker-Menezes, D.; Capparelli, C.; Balliet, R.; Lin, Z.; Pestell, R.G.; Howell, A.; Aquila, S.; Andò, S.; Martinez-Outschoorn, U.; et al. Metabolic reprogramming of cancer-associated fibroblasts by TGF-β drives tumor growth: Connecting TGF-β signaling with "warburg-like" cancer metabolism and L-lactate production. Cell. Cycle 2012, 11, 3019–3035. [Google Scholar] [CrossRef] [Green Version]
- Micke, P.; Ostman, A. Exploring the tumour environment: Cancer-associated fibroblasts as targets in cancer therapy. Expert Opin. Ther. Targets 2005, 9, 1217–1233. [Google Scholar] [CrossRef]
- Bellei, B.; Picardo, M. Premature cell senescence in human skin: Dual face in chronic acquired pigmentary disorders. Ageing Res. Rev. 2020, 57, 100981. [Google Scholar] [CrossRef]
- Almeida, F.V.; Douglass, S.M.; Fane, M.E.; Weeraratna, A.T. Bad Company: Microenvironmentally mediated resistance to targeted therapy in melanoma. Pigment Cell. Melanoma Res. 2019, 32, 237–247. [Google Scholar] [CrossRef]
- Tsang, M.; Quesnel, K.; Vincent, K.; Hutchenreuther, J.; Postovit, L.M.; Leask, A. Insights into fibroblast plasticity: Cellular communication network 2 is required for activation of cancer-associated fibroblasts in a murine model of melanoma. Am. J. Pathol. 2020, 190, 206–221. [Google Scholar] [CrossRef]
- Danen, E.H.; de Vries, T.J.; Morandini, R.; Ghanem, G.G.; Ruiter, D.J.; van Muijen, G.N. E-cadherin expression in human melanoma. Melanoma Res. 1996, 6, 127–131. [Google Scholar] [CrossRef]
- Hsu, M.Y.; Meier, F.E.; Nesbit, M.; Hsu, J.Y.; Van Belle, P.; Elder, D.E.; Herlyn, M. E-cadherin expression in melanoma cells restores keratinocyte-mediated growth control and down-regulates expression of invasion-related adhesion receptors. Am. J. Pathol. 2000, 156, 1515–1525. [Google Scholar] [CrossRef] [Green Version]
- Silye, R.; Karayiannakis, A.J.; Syrigos, K.N.; Poole, S.; van Noorden, S.; Batchelor, W.; Regele, H.; Sega, W.; Boesmueller, H.; Krausz, T.; et al. E-cadherin/catenin complex in benign and malignant melanocytic lesions. J. Pathol. 1998, 186, 350–355. [Google Scholar] [CrossRef]
- Hsu, M.Y.; Meier, F.; Herlyn, M. Melanoma development and progression: A conspiracy between tumor and host. Differentiation 2002, 70, 522–536. [Google Scholar] [CrossRef] [PubMed]
- Sanders, D.S.; Blessing, K.; Hassan, G.A.; Bruton, R.; Marsden, J.R.; Jankowski, J. Alterations in cadherin and catenin expression during the biological progression of melanocytic tumours. Mol. Pathol. 1999, 52, 151–157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haass, N.K.; Smalley, K.S.; Li, L.; Herlyn, M. Adhesion, migration and communication in melanocytes and melanoma. Pigment Cell Res. 2005, 18, 150–159. [Google Scholar] [CrossRef] [PubMed]
- Tang, A.; Eller, M.S.; Hara, M.; Yaar, M.; Hirohashi, S.; Gilchrest, B.A. E-cadherin is the major mediator of human melanocyte adhesion to keratinocytes in vitro. J. Cell. Sci. 1994, 107, 983–992. [Google Scholar]
- Li, G.; Satyamoorthy, K.; Herlyn, M. N-cadherin-mediated intercellular interactions promote survival and migration of melanoma cells. Cancer Res. 2001, 61, 3819–3825. [Google Scholar]
- Li, G.; Satyamoorthy, K.; Meier, F.; Berking, C.; Bogenrieder, T.; Herlyn, M. Function and regulation of melanoma-stromal fibroblast interactions: When seeds meet soil. Oncogene 2003, 22, 3162–3171. [Google Scholar] [CrossRef] [Green Version]
- Kuphal, S.; Poser, I.; Jobin, C.; Hellerbrand, C.; Bosserhoff, A.K. Loss of E-cadherin leads to upregulation of NFkappaB activity in malignant melanoma. Oncogene 2004, 23, 8509–8519. [Google Scholar] [CrossRef] [Green Version]
- McGary, E.C.; Lev, D.C.; Bar-Eli, M. Cellular adhesion pathways and metastatic potential of human melanoma. Cancer Biol. Ther. 2002, 1, 459–465. [Google Scholar] [CrossRef] [Green Version]
- Hazan, R.B.; Phillips, G.R.; Qiao, R.F.; Norton, L.; Aaronson, S.A. Exogenous expression of N-cadherin in breast cancer cells induces cell migration, invasion, and metastasis. J. Cell Biol. 2000, 148, 779–790. [Google Scholar] [CrossRef] [Green Version]
- Raposo, G.; Marks, M.S. Melanosomes—dark organelles enlighten endosomal membrane transport. Nat. Rev. Mol. Cell Biol. 2007, 8, 786–797. [Google Scholar] [CrossRef] [Green Version]
- Dror, S.; Sander, L.; Schwartz, H.; Sheinboim, D.; Barzilai, A.; Dishon, Y.; Apcher, S.; Golan, T.; Greenberger, S.; Barshack, I.; et al. Melanoma miRNA trafficking controls tumour primary niche formation. Nat. Cell Biol. 2016, 18, 1006–1017. [Google Scholar] [CrossRef] [PubMed]
- Anderberg, C.; Pietras, K. On the origin of cancer-associated fibroblasts. Cell. Cycle 2009, 8, 1461–1462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hauge, A.; Rofstad, E.K. Antifibrotic therapy to normalize the tumor microenvironment. J. Transl. Med. 2020, 18, 207–218. [Google Scholar] [CrossRef]
- Otranto, M.; Sarrazy, V.; Bonté, F.; Hinz, B.; Gabbiani, G.; Desmoulière, A. The role of the myofibroblast in tumor stroma remodeling. Cell. Adh. Migr. 2012, 6, 203–219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shimoda, M.; Mellody, K.T.; Orimo, A. Carcinoma-associated fibroblasts are a rate-limiting determinant for tumour progression. Semin. Cell Dev. Biol. 2010, 21, 19–25. [Google Scholar] [CrossRef] [PubMed]
- Érsek, B.; Silló, P.; Cakir, U.; Molnár, V.; Bencsik, A.; Mayer, B.; Mezey, E.; Kárpáti, S.; Pós, Z.; Németh, K. Melanoma-associated fibroblasts impair CD8+ T cell function and modify expression of immune checkpoint regulators via increased arginase activity. Cell Mol. Life Sci. 2020, 77, 1–13. [Google Scholar]
- Zhou, L.; Yang, K.; Andl, T.; Wickett, R.R.; Zhang, Y. Perspective of targeting cancer-associated fibroblasts in melanoma. J. Cancer 2015, 6, 717–726. [Google Scholar] [CrossRef] [Green Version]
- García-Silva, S.; Peinado, H. Melanosomes foster a tumour niche by activating CAFs. Nat. Cell Biol. 2016, 18, 911–913. [Google Scholar] [CrossRef]
- Gieniec, K.A.; Butler, L.M.; Worthley, D.L.; Woods, S.L. Cancer-associated fibroblasts-heroes or villains? Br. J. Cancer 2019, 121, 293–302. [Google Scholar] [CrossRef]
- Zhou, L.; Yang, K.; Randall Wickett, R.; Zhang, Y. Dermal fibroblasts induce cell cycle arrest and block epithelial-mesenchymal transition to inhibit the early stage melanoma development. Cancer Med. 2016, 5, 1566–1579. [Google Scholar] [CrossRef]
- Cornil, I.; Theodorescu, D.; Man, S.; Herlyn, M.; Jambrosic, J.; Kerbel, R.S. Fibroblast cell interactions with human melanoma cells affect tumor cell growth as a function of tumor progression. Proc. Natl. Acad. Sci. USA 1991, 88, 6028–6032. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, L.; Yang, K.; Wickett, R.R.; Kadekaro, A.L.; Zhang, Y. Targeted deactivation of cancer-associated fibroblasts by β-catenin ablation suppresses melanoma growth. Tumour Biol. 2016, 37, 14235–14248. [Google Scholar] [CrossRef] [PubMed]
- Madar, S.; Brosh, R.; Buganim, Y.; Ezra, O.; Goldstein, I.; Solomon, H.; Kogan, I.; Goldfinger, N.; Klocker, H.; Rotter, V. Modulated expression of WFDC1 during carcinogenesis and cellular senescence. Carcinogenesis 2009, 30, 20–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liberato, T.; Pessotti, D.S.; Fukushima, I.; Kitano, E.S.; Serrano, S.M.T.; Zelanis, A. Signatures of protein expression revealed by secretome analyses of cancer associated fibroblasts and melanoma cell lines. J. Proteomics 2018, 174, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Pessotti, D.S.; Andrade-Silva, D.; Serrano, S.M.T.; Zelanis, A. Heterotypic signaling between dermal fibroblasts and melanoma cells induces phenotypic plasticity and proteome rearrangement in malignant cells. Biochim. Biophys. Acta Proteins Proteom. 2020, 1868, 140525. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; Dragulev, B.; Zigrino, P.; Mauch, C.; Fox, J.W. The invasive potential of human melanoma cell lines correlates with their ability to alter fibroblast gene expression in vitro and the stromal microenvironment in vivo. Int. J. Cancer 2009, 125, 1796–1804. [Google Scholar] [CrossRef]
- Lu, C.; Vickers, M.F.; Kerbel, R.S. Interleukin 6: A fibroblast-derived growth inhibitor of human melanoma cells from early but not advanced stages of tumor progression. Proc. Natl. Acad. Sci. USA 1992, 89, 9215–9219. [Google Scholar] [CrossRef] [Green Version]
- Jobe, N.P.; Rösel, D.; Dvořánková, B.; Kodet, O.; Lacina, L.; Mateu, R.; Smetana, K.; Brábek, J. Simultaneous blocking of IL-6 and IL-8 is sufficient to fully inhibit CAF-induced human melanoma cell invasiveness. Histochem. Cell Biol. 2016, 146, 205–217. [Google Scholar] [CrossRef]
- Balsamo, M.; Scordamaglia, F.; Pietra, G.; Manzini, C.; Cantoni, C.; Boitano, M.; Queirolo, P.; Vermi, W.; Facchetti, F.; Moretta, A.; et al. Melanoma-associated fibroblasts modulate NK cell phenotype and antitumor cytotoxicity. Proc. Natl. Acad. Sci. USA 2009, 106, 20847–20852. [Google Scholar] [CrossRef] [Green Version]
- Goldstein, L.J.; Chen, H.; Bauer, R.J.; Bauer, S.M.; Velazquez, O.C. Normal human fibroblasts enable melanoma cells to induce angiogenesis in type I collagen. Surgery 2005, 138, 439–449. [Google Scholar] [CrossRef]
- Rhim, A.D.; Oberstein, P.E.; Thomas, D.H.; Mirek, E.T.; Palermo, C.F.; Sastra, S.A.; Dekleva, E.N.; Saunders, T.; Becerra, C.P.; Tattersall, I.W.; et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell 2014, 25, 735–747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Özdemir, B.C.; Pentcheva-Hoang, T.; Carstens, J.L.; Zheng, X.; Wu, C.C.; Simpson, T.R.; Laklai, H.; Sugimoto, H.; Kahlert, C.; Novitskiy, S.V.; et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell 2015, 28, 831–833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kikuchi, A.; Yamamoto, H.; Sato, A.; Matsumoto, S. New insights into the mechanism of Wnt signaling pathway activation. Int. Rev. Cell. Mol. Biol. 2011, 291, 21–71. [Google Scholar] [PubMed]
- van Amerongen, R.; Nusse, R. Towards an integrated view of Wnt signaling in development. Development 2009, 136, 3205–3214. [Google Scholar] [CrossRef] [Green Version]
- MacDonald, B.T.; Tamai, K.; He, X. Wnt/beta-catenin signaling: Components, mechanisms, and diseases. Dev. Cell. 2009, 17, 9–26. [Google Scholar] [CrossRef] [Green Version]
- Gloushankova, N.A.; Rubtsova, S.N.; Zhitnyak, I.Y. Cadherin-mediated cell-cell interactions in normal and cancer cells. Tissue Barriers 2017, 5, e1356900. [Google Scholar] [CrossRef] [Green Version]
- Tafrihi, M.; Nakhaei Sistani, R. E-cadherin/β-catenin complex: A target for anticancer and antimetastasis plants/plant-derived compounds. Nutr. Cancer 2017, 69, 702–722. [Google Scholar] [CrossRef]
- Behrens, J.; Jerchow, B.A.; Würtele, M.; Grimm, J.; Asbrand, C.; Wirtz, R.; Kühl, M.; Wedlich, D.; Birchmeier, W. Functional interaction of an axin homolog, conductin, with beta-catenin, APC, and GSK3beta. Science 1998, 280, 596–599. [Google Scholar] [CrossRef]
- Hrckulak, D.; Kolar, M.; Strnad, H.; Korinek, V. TCF/LEF transcription factors: An update from the internet resources. Cancers 2016, 8, 70. [Google Scholar] [CrossRef] [Green Version]
- Dorsky, R.I.; Raible, D.W.; Moon, R.T. Direct regulation of nacre, a zebrafish MITF homolog required for pigment cell formation, by the Wnt pathway. Genes Dev. 2000, 14, 158–162. [Google Scholar]
- Schepsky, A.; Bruser, K.; Gunnarsson, G.J.; Goodall, J.; Hallsson, J.H.; Goding, C.R.; Steingrimsson, E.; Hecht, A. The microphthalmia-associated transcription factor Mitf interacts with beta-catenin to determine target gene expression. Mol. Cell. Biol. 2006, 26, 8914–8927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saito, H.; Yasumoto, K.; Takeda, K.; Takahashi, K.; Yamamoto, H.; Shibahara, S. Microphthalmia-associated transcription factor in the Wnt signaling pathway. Pigment Cell Res. 2003, 16, 261–265. [Google Scholar] [CrossRef] [PubMed]
- Larue, L.; Delmas, V. The WNT/beta-catenin pathway in melanoma. Front. Biosci. 2006, 11, 733–742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bellei, B.; Flori, E.; Izzo, E.; Maresca, V.; Picardo, M. GSK3beta inhibition promotes melanogenesis in mouse B16 melanoma cells and normal human melanocytes. Cell. Signal. 2008, 20, 1750–1761. [Google Scholar] [CrossRef] [PubMed]
- Bellei, B.; Pitisci, A.; Catricalà, C.; Larue, L.; Picardo, M. Wnt/β-catenin signaling is stimulated by α-melanocyte-stimulating hormone in melanoma and melanocyte cells: Implication in cell differentiation. Pigment Cell. Melanoma Res. 2011, 24, 309–325. [Google Scholar] [CrossRef]
- Larue, L.; Kumasaka, M.; Goding, C.R. Beta-catenin in the melanocyte lineage. Pigment Cell Res. 2003, 16, 312–317. [Google Scholar] [CrossRef]
- Hari, L.; Brault, V.; Kléber, M.; Lee, H.Y.; Ille, F.; Leimeroth, R.; Paratore, C.; Suter, U.; Kemler, R.; Sommer, L. Lineage-specific requirements of beta-catenin in neural crest development. J. Cell Biol. 2002, 159, 867–880. [Google Scholar] [CrossRef] [Green Version]
- De Melker, A.A.; Desban, N.; Duband, J.L. Cellular localization and signaling activity of beta-catenin in migrating neural crest cells. Dev. Dyn. 2004, 230, 708–726. [Google Scholar] [CrossRef]
- Yamada, T.; Hasegawa, S.; Inoue, Y.; Date, Y.; Yamamoto, N.; Mizutani, H.; Nakata, S.; Matsunaga, K.; Akamatsu, H. Wnt/β-catenin and kit signaling sequentially regulate melanocyte stem cell differentiation in UVB-induced epidermal pigmentation. J. Investig. Dermatol. 2013, 133, 2753–2762. [Google Scholar] [CrossRef] [Green Version]
- Goldstein, N.B.; Koster, M.I.; Jones, K.L.; Gao, B.; Hoaglin, L.G.; Robinson, S.E.; Wright, M.J.; Birlea, S.I.; Luman, A.; Lambert, K.A.; et al. Repigmentation of human vitiligo skin by NBUVB is controlled by transcription of GLI1 and activation of the β-catenin pathway in the hair follicle bulge stem cells. J. Investig. Dermatol. 2018, 138, 657–668. [Google Scholar] [CrossRef] [Green Version]
- Bellei, B.; Pacchiarotti, A.; Perez, M.; Faraggiana, T. Frequent beta-catenin overexpression without exon 3 mutation in cutaneous lymphomas. Mod. Pathol. 2004, 17, 1275–1281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Clevers, H. Wnt/beta-catenin signaling in development and disease. Cell 2006, 127, 469–480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ring, A.; Kim, Y.M.; Kahn, M. Wnt/catenin signaling in adult stem cell physiology and disease. Stem Cell. Rev. Rep. 2014, 10, 512–525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Galluzzi, L.; Spranger, S.; Fuchs, E.; López-Soto, A. WNT signaling in cancer immunosurveillance. Trends Cell Biol. 2019, 29, 44–65. [Google Scholar] [CrossRef] [PubMed]
- Sinnberg, T.; Menzel, M.; Ewerth, D.; Sauer, B.; Schwarz, M.; Schaller, M.; Garbe, C.; Schittek, B. Β-catenin signaling increases during melanoma progression and promotes tumor cell survival and chemoresistance. PLoS ONE 2011, 6, e23429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rimm, D.L.; Caca, K.; Hu, G.; Harrison, F.B.; Fearon, E.R. Frequent nuclear/cytoplasmic localization of beta-catenin without exon 3 mutations in malignant melanoma. Am. J. Pathol. 1999, 154, 325–329. [Google Scholar] [CrossRef]
- Omholt, K.; Platz, A.; Ringborg, U.; Hansson, J. Cytoplasmic and nuclear accumulation of beta-catenin is rarely caused by CTNNB1 exon 3 mutations in cutaneous malignant melanoma. Int. J. Cancer 2001, 92, 839–842. [Google Scholar] [CrossRef]
- Demunter, A.; Libbrecht, L.; Degreef, H.; De Wolf-Peeters, C.; van den Oord, J.J. Loss of membranous expression of beta-catenin is associated with tumor progression in cutaneous melanoma and rarely caused by exon 3 mutations. Mod. Pathol. 2002, 15, 454–461. [Google Scholar] [CrossRef] [Green Version]
- Worm, J.; Christensen, C.; Grønbaek, K.; Tulchinsky, E.; Guldberg, P. Genetic and epigenetic alterations of the APC gene in malignant melanoma. Oncogene 2004, 23, 5215–5226. [Google Scholar] [CrossRef] [Green Version]
- Reifenberger, J.; Knobbe, C.B.; Wolter, M.; Blaschke, B.; Schulte, K.W.; Pietsch, T.; Ruzicka, T.; Reifenberger, G. Molecular genetic analysis of malignant melanomas for aberrations of the WNT signaling pathway genes CTNNB1, APC, ICAT and BTRC. Int. J. Cancer 2002, 100, 549–556. [Google Scholar] [CrossRef]
- Castiglia, D.; Bernardini, S.; Alvino, E.; Pagani, E.; De Luca, N.; Falcinelli, S.; Pacchiarotti, A.; Bonmassar, E.; Zambruno, G.; D’Atri, S. Concomitant activation of Wnt pathway and loss of mismatch repair function in human melanoma. Genes Chromosomes Cancer 2008, 47, 614–624. [Google Scholar] [CrossRef] [PubMed]
- Rubinfeld, B.; Robbins, P.; El-Gamil, M.; Albert, I.; Porfiri, E.; Polakis, P. Stabilization of beta-catenin by genetic defects in melanoma cell lines. Science 1997, 275, 1790–1792. [Google Scholar] [CrossRef] [PubMed]
- Kuphal, S.; Lodermeyer, S.; Bataille, F.; Schuierer, M.; Hoang, B.H.; Bosserhoff, A.K. Expression of dickkopf genes is strongly reduced in malignant melanoma. Oncogene 2006, 25, 5027–5036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huynh, K.T.; Takei, Y.; Kuo, C.; Scolyer, R.A.; Murali, R.; Chong, K.; Takeshima, L.; Sim, M.S.; Morton, D.L.; Turner, R.R.; et al. Aberrant hypermethylation in primary tumours and sentinel lymph node metastases in paediatric patients with cutaneous melanoma. Br. J. Dermatol. 2012, 166, 1319–1326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Massi, D.; Romano, E.; Rulli, E.; Merelli, B.; Nassini, R.; De Logu, F.; Bieche, I.; Baroni, G.; Cattaneo, L.; Xue, G.; et al. Baseline β-catenin, programmed death-ligand 1 expression and tumour-infiltrating lymphocytes predict response and poor prognosis in BRAF inhibitor-treated melanoma patients. Eur. J. Cancer 2017, 78, 70–81. [Google Scholar] [CrossRef]
- Gajos-Michniewicz, A.; Czyz, M. WNT signaling in melanoma. Int. J. Mol. Sci. 2020, 21, 4852. [Google Scholar] [CrossRef]
- Lucero, O.M.; Dawson, D.W.; Moon, R.T.; Chien, A.J. A re-evaluation of the “oncogenic” nature of Wnt/beta-catenin signaling in melanoma and other cancers. Curr. Oncol. Rep. 2010, 12, 314–318. [Google Scholar] [CrossRef] [Green Version]
- Larue, L.; Delmas, V. Secrets to developing Wnt-age melanoma revealed. Pigment Cell. Melanoma Res. 2009, 22, 520–521. [Google Scholar] [CrossRef]
- Widlund, H.R.; Horstmann, M.A.; Price, E.R.; Cui, J.; Lessnick, S.L.; Wu, M.; He, X.; Fisher, D.E. Beta-Catenin-induced melanoma growth requires the downstream target microphthalmia-associated transcription factor. J. Cell Biol. 2002, 158, 1079–1087. [Google Scholar] [CrossRef] [Green Version]
- Chien, A.J.; Moore, E.C.; Lonsdorf, A.S.; Kulikauskas, R.M.; Rothberg, B.G.; Berger, A.J.; Major, M.B.; Hwang, S.T.; Rimm, D.L.; Moon, R.T. Activated Wnt/beta-catenin signaling in melanoma is associated with decreased proliferation in patient tumors and a murine melanoma model. Proc. Natl. Acad. Sci. USA 2009, 106, 1193–1198. [Google Scholar] [CrossRef] [Green Version]
- Arozarena, I.; Bischof, H.; Gilby, D.; Belloni, B.; Dummer, R.; Wellbrock, C. In melanoma, beta-catenin is a suppressor of invasion. Oncogene 2011, 30, 4531–4543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gallagher, S.J.; Rambow, F.; Kumasaka, M.; Champeval, D.; Bellacosa, A.; Delmas, V.; Larue, L. Beta-catenin inhibits melanocyte migration but induces melanoma metastasis. Oncogene 2013, 32, 2230–2238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bachmann, I.M.; Straume, O.; Puntervoll, H.E.; Kalvenes, M.B.; Akslen, L.A. Importance of p-cadherin, beta-catenin, and Wnt5a/frizzled for progression of melanocytic tumors and prognosis in cutaneous melanoma. Clin. Cancer Res. 2005, 11, 8606–8614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kageshita, T.; Hamby, C.V.; 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, 210–216. [Google Scholar] [CrossRef]
- De la Fouchardière, A.; Caillot, C.; Jacquemus, J.; Durieux, E.; Houlier, A.; Haddad, V.; Pissaloux, D. Β-catenin nuclear expression discriminates deep penetrating nevi from other cutaneous melanocytic tumors. Virchows Arch. 2019, 474, 539–550. [Google Scholar] [CrossRef]
- Hoek, K.S.; Eichhoff, O.M.; Schlegel, N.C.; Döbbeling, U.; Kobert, N.; Schaerer, L.; Hemmi, S.; Dummer, R. In vivo switching of human melanoma cells between proliferative and invasive states. Cancer Res. 2008, 68, 650–656. [Google Scholar] [CrossRef] [Green Version]
- Eichhoff, O.M.; Zipser, M.C.; Xu, M.; Weeraratna, A.T.; Mihic, D.; Dummer, R.; Hoek, K.S. The immunohistochemistry of invasive and proliferative phenotype switching in melanoma: A case report. Melanoma Res. 2010, 20, 349–355. [Google Scholar] [CrossRef] [Green Version]
- Kovacs, D.; Migliano, E.; Muscardin, L.; Silipo, V.; Catricalà, C.; Picardo, M.; Bellei, B. The role of Wnt/β-catenin signaling pathway in melanoma epithelial-to-mesenchymal-like switching: Evidences from patients-derived cell lines. Oncotarget 2016, 7, 43295–43314. [Google Scholar] [CrossRef] [Green Version]
- Webster, M.R.; Kugel, C.H., III; Weeraratna, A.T. The Wnts of change: How Wnts regulate phenotype switching in melanoma. Biochim. Biophys. Acta 2015, 1856, 244–251. [Google Scholar] [CrossRef] [Green Version]
- Spranger, S.; Bao, R.; Gajewski, T.F. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 2015, 523, 231–235. [Google Scholar] [CrossRef]
- Nsengimana, J.; Laye, J.; Filia, A.; O’Shea, S.; Muralidhar, S.; Poźniak, J.; Droop, A.; Chan, M.; Walker, C.; Parkinson, L.; et al. Β-catenin-mediated immune evasion pathway frequently operates in primary cutaneous melanomas. J. Clin. Investig. 2018, 128, 2048–2063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, X.; Xiang, Y.; Li, F.; Yin, C.; Li, B.; Ke, X. WNT/β-catenin signaling pathway regulating T cell-inflammation in the tumor microenvironment. Front. Immunol. 2019, 10, 2293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luke, J.J.; Bao, R.; Sweis, R.F.; Spranger, S.; Gajewski, T.F. WNT/β-catenin pathway activation correlates with immune exclusion across human cancers. Clin. Cancer Res. 2019, 25, 3074–3083. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ganesh, S.; Shui, X.; Craig, K.P.; Park, J.; Wang, W.; Brown, B.D.; Abrams, M.T. RNAi-mediated β-catenin inhibition promotes T cell infiltration and antitumor activity in combination with immune checkpoint blockade. Mol. Ther. 2018, 26, 2567–2579. [Google Scholar] [CrossRef]
- Biechele, T.L.; Kulikauskas, R.M.; Toroni, R.A.; Lucero, O.M.; Swift, R.D.; James, R.G.; Robin, N.C.; Dawson, D.W.; Moon, R.T.; Chien, A.J. Wnt/β-catenin signaling and AXIN1 regulate apoptosis triggered by inhibition of the mutant kinase BRAFV600E in human melanoma. Sci. Signal. 2012, 5, ra3. [Google Scholar] [CrossRef] [Green Version]
- Conrad, W.H.; Swift, R.D.; Biechele, T.L.; Kulikauskas, R.M.; Moon, R.T.; Chien, A.J. Regulating the response to targeted MEK inhibition in melanoma: Enhancing apoptosis in NRAS- and BRAF-mutant melanoma cells with Wnt/β-catenin activation. Cell. Cycle 2012, 11, 3724–3730. [Google Scholar] [CrossRef] [Green Version]
- Chien, A.J.; Haydu, L.E.; Biechele, T.L.; Kulikauskas, R.M.; Rizos, H.; Kefford, R.F.; Scolyer, R.A.; Moon, R.T.; Long, G.V. Targeted BRAF inhibition impacts survival in melanoma patients with high levels of Wnt/β-catenin signaling. PLoS ONE 2014, 9, e94748. [Google Scholar] [CrossRef]
- Fodde, R.; Brabletz, T. Wnt/beta-catenin signaling in cancer stemness and malignant behavior. Curr. Opin. Cell Biol. 2007, 19, 150–158. [Google Scholar] [CrossRef]
- Unterleuthner, D.; Neuhold, P.; Schwarz, K.; Janker, L.; Neuditschko, B.; Nivarthi, H.; Crncec, I.; Kramer, N.; Unger, C.; Hengstschläger, M.; et al. Cancer-associated fibroblast-derived WNT2 increases tumor angiogenesis in colon cancer. Angiogenesis 2020, 23, 159–177. [Google Scholar] [CrossRef] [Green Version]
- Kramer, N.; Schmöllerl, J.; Unger, C.; Nivarthi, H.; Rudisch, A.; Unterleuthner, D.; Scherzer, M.; Riedl, A.; Artaker, M.; Crncec, I.; et al. Autocrine WNT2 signaling in fibroblasts promotes colorectal cancer progression. Oncogene 2017, 36, 5460–5472. [Google Scholar] [CrossRef]
- Rasola, A.; Fassetta, M.; De Bacco, F.; D’Alessandro, L.; Gramaglia, D.; Di Renzo, M.F.; Comoglio, P.M. A positive feedback loop between hepatocyte growth factor receptor and beta-catenin sustains colorectal cancer cell invasive growth. Oncogene 2007, 26, 1078–1087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, L.; Lin, C.; Liu, Z.R. P68 RNA helicase mediates PDGF-induced epithelial mesenchymal transition by displacing axin from beta-catenin. Cell 2006, 127, 139–155. [Google Scholar] [CrossRef] [Green Version]
- Kaur, A.; Webster, M.R.; Marchbank, K.; Behera, R.; Ndoye, A.; Kugel, C.H., III; Dang, V.M.; Appleton, J.; O’Connell, M.P.; Cheng, P.; et al. SFRP2 in the aged microenvironment drives melanoma metastasis and therapy resistance. Nature 2016, 532, 250–254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bittner, M.; Meltzer, P.; Chen, Y.; Jiang, Y.; Seftor, E.; Hendrix, M.; Radmacher, M.; Simon, R.; Yakhini, Z.; Ben-Dor, A.; et al. Molecular classification of cutaneous malignant melanoma by gene expression profiling. Nature 2000, 406, 536–540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weeraratna, A.T.; Jiang, Y.; Hostetter, G.; Rosenblatt, K.; Duray, P.; Bittner, M.; Trent, J.M. Wnt5a signaling directly affects cell motility and invasion of metastatic melanoma. Cancer Cell 2002, 1, 279–288. [Google Scholar] [CrossRef] [Green Version]
- Pham, K.; Milovanovic, T.; Barr, R.J.; Truong, T.; Holcombe, R.F. Wnt ligand expression in malignant melanoma: Pilot study indicating correlation with histopathological features. Mol. Pathol. 2003, 56, 280–285. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.; Ye, Y.C.; Chen, Y.; Zhao, J.L.; Gao, C.C.; Han, H.; Liu, W.C.; Qin, H.Y. Crosstalk between hepatic tumor cells and macrophages via Wnt/β-catenin signaling promotes M2-like macrophage polarization and reinforces tumor malignant behaviors. Cell. Death Dis. 2018, 9, 793. [Google Scholar] [CrossRef]
- Katoh, M. Multi-layered prevention and treatment of chronic inflammation, organ fibrosis and cancer associated with canonical WNT/β-catenin signaling activation (review). Int. J. Mol. Med. 2018, 42, 713–725. [Google Scholar] [CrossRef]
- Baarsma, H.A.; Spanjer, A.I.; Haitsma, G.; Engelbertink, L.H.; Meurs, H.; Jonker, M.R.; Timens, W.; Postma, D.S.; Kerstjens, H.A.; Gosens, R. Activation of WNT/β-catenin signaling in pulmonary fibroblasts by TGF-β1 is increased in chronic obstructive pulmonary disease. PLoS ONE 2011, 6, e25450. [Google Scholar] [CrossRef] [Green Version]
- Chilosi, M.; Poletti, V.; Zamò, A.; Lestani, M.; Montagna, L.; Piccoli, P.; Pedron, S.; Bertaso, M.; Scarpa, A.; Murer, B.; et al. Aberrant Wnt/beta-catenin pathway activation in idiopathic pulmonary fibrosis. Am. J. Pathol. 2003, 162, 1495–1502. [Google Scholar] [CrossRef]
- Cheon, H.; Boyle, D.L.; Firestein, G.S. Wnt1 inducible signaling pathway protein-3 regulation and microsatellite structure in arthritis. J. Rheumatol. 2004, 31, 2106–2114. [Google Scholar] [PubMed]
- Cheon, S.; Poon, R.; Yu, C.; Khoury, M.; Shenker, R.; Fish, J.; Alman, B.A. Prolonged beta-catenin stabilization and Tcf-dependent transcriptional activation in hyperplastic cutaneous wounds. Lab. Investig. 2005, 85, 416–425. [Google Scholar] [CrossRef] [PubMed]
- Akhmetshina, A.; Palumbo, K.; Dees, C.; Bergmann, C.; Venalis, P.; Zerr, P.; Horn, A.; Kireva, T.; Beyer, C.; Zwerina, J.; et al. Activation of canonical Wnt signalling is required for TGF-β-mediated fibrosis. Nat. Commun. 2012, 3, 735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jahoda, C.A.B.; Gilmore, A.C. What lies beneath: Wnt/β-catenin signaling and cell fate in the lower dermis. J. Investig. Dermatol. 2016, 136, 1084–1087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mastrogiannaki, M.; Lichtenberger, B.M.; Reimer, A.; Collins, C.A.; Driskell, R.R.; Watt, F.M. Β-catenin stabilization in skin fibroblasts causes fibrotic lesions by preventing adipocyte differentiation of the reticular dermis. J. Investig. Dermatol. 2016, 136, 1130–1142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, T.; Zhou, L.; Yang, K.; Iwasawa, K.; Kadekaro, A.L.; Takebe, T.; Andl, T.; Zhang, Y. The β-catenin/YAP signaling axis is a key regulator of melanoma-associated fibroblasts. Signal. Transduct. Target. Ther. 2019, 4, 63–78. [Google Scholar] [CrossRef] [Green Version]
- Zhou, L.; Yang, K.; Dunaway, S.; Abdel-Malek, Z.; Andl, T.; Kadekaro, A.L.; Zhang, Y. Suppression of MAPK signaling in BRAF-activated PTEN-deficient melanoma by blocking β-catenin signaling in cancer-associated fibroblasts. Pigment Cell. Melanoma Res. 2018, 31, 297–307. [Google Scholar] [CrossRef]
- Shao, H.; Kong, R.; Ferrari, M.L.; Radtke, F.; Capobianco, A.J.; Liu, Z.J. Notch1 pathway activity determines the regulatory role of cancer-associated fibroblasts in melanoma growth and invasion. PLoS ONE 2015, 10, e0142815. [Google Scholar] [CrossRef]
- Du, J.; Zu, Y.; Li, J.; Du, S.; Xu, Y.; Zhang, L.; Jiang, L.; Wang, Z.; Chien, S.; Yang, C. Extracellular matrix stiffness dictates Wnt expression through integrin pathway. Sci. Rep. 2016, 6, 20395. [Google Scholar] [CrossRef] [Green Version]
- Yeung, Y.T.; Guerrero-Castilla, A.; Cano, M.; Muñoz, M.F.; Ayala, A.; Argüelles, S. Dysregulation of the hippo pathway signaling in aging and cancer. Pharmacol. Res. 2019, 143, 151–165. [Google Scholar] [CrossRef]
- Yu, F.X.; Zhao, B.; Guan, K.L. Hippo pathway in organ size control, tissue homeostasis, and cancer. Cell 2015, 163, 811–828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Harvey, K.F.; Zhang, X.; Thomas, D.M. The hippo pathway and human cancer. Nat. Rev. Cancer 2013, 13, 246–257. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Tang, J.Z.; Vergara, I.A.; Zhang, Y.; Szeto, P.; Yang, L.; Mintoff, C.; Colebatch, A.; McIntosh, L.; Mitchell, K.A.; et al. Somatic hypermutation of the YAP oncogene in a human cutaneous melanoma. Mol. Cancer Res. 2019, 17, 1435–1449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Menzel, M.; Meckbach, D.; Weide, B.; Toussaint, N.C.; Schilbach, K.; Noor, S.; Eigentler, T.; Ikenberg, K.; Busch, C.; Quintanilla-Martinez, L.; et al. In melanoma, hippo signaling is affected by copy number alterations and YAP1 overexpression impairs patient survival. Pigment Cell. Melanoma Res. 2014, 27, 671–673. [Google Scholar] [CrossRef] [PubMed]
- Sarmasti Emami, S.; Zhang, D.; Yang, X. Interaction of the hippo pathway and phosphatases in tumorigenesis. Cancers 2020, 12, 2438. [Google Scholar] [CrossRef] [PubMed]
- Dey, A.; Varelas, X.; Guan, K.L. Targeting the hippo pathway in cancer, fibrosis, wound healing and regenerative medicine. Nat. Rev. Drug Discov. 2020, 19, 480–494. [Google Scholar] [CrossRef]
- Huh, H.D.; Kim, D.H.; Jeong, H.S.; Park, H.W. Regulation of TEAD transcription factors in cancer biology. Cells 2019, 8, 600. [Google Scholar] [CrossRef] [Green Version]
- Kim, M.K.; Jang, J.W.; Bae, S.C. DNA binding partners of YAP/TAZ. BMB Rep. 2018, 51, 126–133. [Google Scholar] [CrossRef] [Green Version]
- Corvaisier, M.; Bauzone, M.; Corfiotti, F.; Renaud, F.; El Amrani, M.; Monté, D.; Truant, S.; Leteurtre, E.; Formstecher, P.; Van Seuningen, I.; et al. Regulation of cellular quiescence by YAP/TAZ and cyclin E1 in colon cancer cells: Implication in chemoresistance and cancer relapse. Oncotarget 2016, 7, 56699–56712. [Google Scholar] [CrossRef] [Green Version]
- Perez, D.E.; Henle, A.M.; Amsterdam, A.; Hagen, H.R.; Lees, J.A. Uveal melanoma driver mutations in GNAQ/11 yield numerous changes in melanocyte biology. Pigment Cell. Melanoma Res. 2018, 31, 604–613. [Google Scholar] [CrossRef]
- Lyubasyuk, V.; Ouyang, H.; Yu, F.X.; Guan, K.L.; Zhang, K. YAP inhibition blocks uveal melanogenesis driven by GNAQ Or GNA11 mutations. Mol. Cell. Oncol. 2014, 2, e970957. [Google Scholar] [CrossRef] [PubMed]
- Javelaud, D.; Alexaki, V.I.; Pierrat, M.J.; Hoek, K.S.; Dennler, S.; Van Kempen, L.; Bertolotto, C.; Ballotti, R.; Saule, S.; Delmas, V.; et al. GLI2 and M-MITF transcription factors control exclusive gene expression programs and inversely regulate invasion in human melanoma cells. Pigment Cell. Melanoma Res. 2011, 24, 932–943. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Yang, L.; Szeto, P.; Abali, G.K.; Zhang, Y.; Kulkarni, A.; Amarasinghe, K.; Li, J.; Vergara, I.A.; Molania, R.; et al. The hippo pathway oncoprotein YAP promotes melanoma cell invasion and spontaneous metastasis. Oncogene 2020, 39, 5267–5281. [Google Scholar] [CrossRef]
- Nallet-Staub, F.; Marsaud, V.; Li, L.; Gilbert, C.; Dodier, S.; Bataille, V.; Sudol, M.; Herlyn, M.; Mauviel, A. Pro-invasive activity of the hippo pathway effectors YAP and TAZ in cutaneous melanoma. J. Investig. Dermatol. 2014, 134, 123–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, N.G.; Koh, E.; Chen, X.; Gumbiner, B.M. E-cadherin mediates contact inhibition of proliferation through hippo signaling-pathway components. Proc. Natl. Acad. Sci. USA 2011, 108, 11930–11935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Azzolin, L.; Zanconato, F.; Bresolin, S.; Forcato, M.; Basso, G.; Bicciato, S.; Cordenonsi, M.; Piccolo, S. Role of TAZ as mediator of Wnt signaling. Cell 2012, 151, 1443–1456. [Google Scholar] [CrossRef] [Green Version]
- Azzolin, L.; Panciera, T.; Soligo, S.; Enzo, E.; Bicciato, S.; Dupont, S.; Bresolin, S.; Frasson, C.; Basso, G.; Guzzardo, V.; et al. YAP/TAZ incorporation in the β-catenin destruction complex orchestrates the Wnt response. Cell 2014, 158, 157–170. [Google Scholar] [CrossRef] [Green Version]
- Imajo, M.; Miyatake, K.; Iimura, A.; Miyamoto, A.; Nishida, E. A molecular mechanism that links hippo signalling to the inhibition of Wnt/β-catenin signalling. EMBO J. 2012, 31, 1109–1122. [Google Scholar] [CrossRef] [Green Version]
- Kim, M.; Jho, E.H. Cross-talk between Wnt/β-catenin and hippo signaling pathways: A brief review. BMB Rep. 2014, 47, 540–545. [Google Scholar] [CrossRef] [Green Version]
- Johannessen, C.M.; Johnson, L.A.; Piccioni, F.; Townes, A.; Frederick, D.T.; Donahue, M.K.; Narayan, R.; Flaherty, K.T.; Wargo, J.A.; Root, D.E.; et al. A melanocyte lineage program confers resistance to MAP kinase pathway inhibition. Nature 2013, 504, 138–142. [Google Scholar] [CrossRef] [Green Version]
- Kim, M.H.; Kim, C.G.; Kim, S.K.; Shin, S.J.; Choe, E.A.; Park, S.H.; Shin, E.C.; Kim, J. YAP-induced PD-L1 expression drives immune evasion in BRAFi-resistant melanoma. Cancer Immunol. Res. 2018, 6, 255–266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moya, I.M.; Castaldo, S.A.; Van den Mooter, L.; Soheily, S.; Sansores-Garcia, L.; Jacobs, J.; Mannaerts, I.; Xie, J.; Verboven, E.; Hillen, H.; et al. Peritumoral activation of the hippo pathway effectors YAP and TAZ suppresses liver cancer in mice. Science 2019, 366, 1029–1034. [Google Scholar] [CrossRef] [PubMed]
- Lawlor, K.; Pérez-Montero, S.; Lima, A.; Rodríguez, T.A. Transcriptional versus metabolic control of cell fitness during cell competition. Semin. Cancer Biol. 2020, 63, 36–43. [Google Scholar] [CrossRef] [PubMed]
- Wu, M.Y.; Hill, C.S. Tgf-beta superfamily signaling in embryonic development and homeostasis. Dev. Cell. 2009, 16, 329–343. [Google Scholar] [CrossRef] [Green Version]
- Shi, Y.; Massagué, J. Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell 2003, 113, 685–700. [Google Scholar] [CrossRef] [Green Version]
- Schmierer, B.; Hill, C.S. TGFbeta-SMAD signal transduction: Molecular specificity and functional flexibility. Nat. Rev. Mol. Cell Biol. 2007, 8, 970–982. [Google Scholar] [CrossRef]
- Weber, C.E.; Li, N.Y.; Wai, P.Y.; Kuo, P.C. Epithelial-mesenchymal transition, TGF-β, and osteopontin in wound healing and tissue remodeling after injury. J. Burn Care. Res. 2012, 33, 311–318. [Google Scholar] [CrossRef] [Green Version]
- Colak, S.; Ten Dijke, P. Targeting TGF-β signaling in cancer. Trends Cancer 2017, 3, 56–71. [Google Scholar] [CrossRef]
- De Caestecker, M.P.; Piek, E.; Roberts, A.B. Role of transforming growth factor-beta signaling in cancer. J. Natl. Cancer Inst. 2000, 92, 1388–1402. [Google Scholar] [CrossRef] [Green Version]
- Krasagakis, K.; Garbe, C.; Schrier, P.I.; Orfanos, C.E. Paracrine and autocrine regulation of human melanocyte and melanoma cell growth by transforming growth factor beta in vitro. Anticancer Res. 1994, 14, 2565–2571. [Google Scholar]
- Rodeck, U.; Bossler, A.; Graeven, U.; Fox, F.E.; Nowell, P.C.; Knabbe, C.; Kari, C. Transforming growth factor beta production and responsiveness in normal human melanocytes and melanoma cells. Cancer Res. 1994, 54, 575–581. [Google Scholar] [PubMed]
- Javelaud, D.; Alexaki, V.I.; Mauviel, A. Transforming growth factor-beta in cutaneous melanoma. Pigment Cell Melanoma Res. 2008, 21, 123–132. [Google Scholar] [CrossRef] [PubMed]
- Van Belle, P.; Rodeck, U.; Nuamah, I.; Halpern, A.C.; Elder, D.E. Melanoma-associated expression of transforming growth factor-beta isoforms. Am. J. Pathol. 1996, 148, 1887–1894. [Google Scholar] [PubMed]
- Reed, J.A.; McNutt, N.S.; Prieto, V.G.; Albino, A.P. Expression of transforming growth factor-beta 2 in malignant melanoma correlates with the depth of tumor invasion. Implications for tumor progression. Am. J. Pathol. 1994, 145, 97–104. [Google Scholar] [PubMed]
- Polanska, U.M.; Orimo, A. Carcinoma-associated fibroblasts: Non-neoplastic tumour-promoting mesenchymal cells. J. Cell. Physiol. 2013, 228, 1651–1657. [Google Scholar] [CrossRef]
- Wipff, P.J.; Rifkin, D.B.; Meister, J.J.; Hinz, B. Myofibroblast contraction activates latent TGF-beta1 from the extracellular matrix. J. Cell Biol. 2007, 179, 1311–1323. [Google Scholar] [CrossRef] [Green Version]
- Pasco, S.; Brassart, B.; Ramont, L.; Maquart, F.X.; Monboisse, J.C. Control of melanoma cell invasion by type IV collagen. Cancer Detect. Prev. 2005, 29, 260–266. [Google Scholar] [CrossRef]
- Najafi, M.; Farhood, B.; Mortezaee, K. Extracellular matrix (ECM) stiffness and degradation as cancer drivers. J. Cell. Biochem. 2019, 120, 2782–2790. [Google Scholar] [CrossRef]
- Burchardt, E.R.; Hein, R.; Bosserhoff, A.K. Laminin, hyaluronan, tenascin-C and type VI collagen levels in sera from patients with malignant melanoma. Clin. Exp. Dermatol. 2003, 28, 515–520. [Google Scholar] [CrossRef]
- Khan, H.Y.; Orimo, A. Transforming growth factor-β: Guardian of catabolic metabolism in carcinoma-associated fibroblasts. Cell. Cycle 2012, 11, 4302–4303. [Google Scholar] [CrossRef] [Green Version]
- Li, Z.; Zhang, J.; Zhou, J.; Lu, L.; Wang, H.; Zhang, G.; Wan, G.; Cai, S.; Du, J. Nodal facilitates differentiation of fibroblasts to cancer-associated fibroblasts that support tumor growth in melanoma and colorectal cancer. Cells 2019, 8, 538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Topczewska, J.M.; Postovit, L.M.; Margaryan, N.V.; Sam, A.; Hess, A.R.; Wheaton, W.W.; Nickoloff, B.J.; Topczewski, J.; Hendrix, M.J. Embryonic and tumorigenic pathways converge via nodal signaling: Role in melanoma aggressiveness. Nat. Med. 2006, 12, 925–932. [Google Scholar] [CrossRef] [PubMed]
- Kodet, O.; Dvořánková, B.; Bendlová, B.; Sýkorová, V.; Krajsová, I.; Štork, J.; Kučera, J.; Szabo, P.; Strnad, H.; Kolář, M.; et al. Microenvironment-driven resistance to B-raf inhibition in a melanoma patient is accompanied by broad changes of gene methylation and expression in distal fibroblasts. Int. J. Mol. Med. 2018, 41, 2687–2703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pramong, N.; Gojaseni, P.; Suttipongkeat, S.; Kiattisunthorn, K.; Chittinandana, A. Diagnostic accuracy of fibroblast growth factor 23 for predicting acute kidney injury in patients with acute decompensated heart failure. Nephrology (Carlton) 2020, e13780. [Google Scholar] [CrossRef]
- Carlino, M.S.; Long, G.V.; Kefford, R.F.; Rizos, H. Targeting oncogenic BRAF and aberrant MAPK activation in the treatment of cutaneous melanoma. Crit. Rev. Oncol. Hematol. 2015, 96, 385–398. [Google Scholar] [CrossRef]
- Daniotti, M.; Oggionni, M.; Ranzani, T.; Vallacchi, V.; Campi, V.; Di Stasi, D.; Torre, G.D.; Perrone, F.; Luoni, C.; Suardi, S.; et al. BRAF alterations are associated with complex mutational profiles in malignant melanoma. Oncogene 2004, 23, 5968–5977. [Google Scholar] [CrossRef] [Green Version]
- Uribe, P.; Wistuba, I.I.; González, S. BRAF mutation: A frequent event in benign, atypical, and malignant melanocytic lesions of the skin. Am. J. Dermatopathol. 2003, 25, 365–370. [Google Scholar] [CrossRef]
- Vanni, I.; Tanda, E.T.; Spagnolo, F.; Andreotti, V.; Bruno, W.; Ghiorzo, P. The current state of molecular testing in the BRAF-mutated melanoma landscape. Front. Mol. Biosci. 2020, 7, 113. [Google Scholar] [CrossRef]
- Halaban, R.; Krauthammer, M. RASopathy gene mutations in melanoma. J. Investig. Dermatol. 2016, 136, 1755–1759. [Google Scholar] [CrossRef] [Green Version]
- Vanni, I.; Tanda, E.T.; Dalmasso, B.; Pastorino, L.; Andreotti, V.; Bruno, W.; Boutros, A.; Spagnolo, F.; Ghiorzo, P. Non-BRAF mutant melanoma: Molecular features and therapeutical implications. Front. Mol. Biosci. 2020, 7, 172. [Google Scholar] [CrossRef]
- Platz, A.; Egyhazi, S.; Ringborg, U.; Hansson, J. Human cutaneous melanoma; A review of NRAS and BRAF mutation frequencies in relation to histogenetic subclass and body site. Mol. Oncol. 2008, 1, 395–405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dantonio, P.M.; Klein, M.O.; Freire, M.R.V.B.; Araujo, C.N.; Chiacetti, A.C.; Correa, R.G. Exploring major signaling cascades in melanomagenesis: A rationale route for targetted skin cancer therapy. Biosci. Rep. 2018, 38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dhomen, N.; Marais, R. New insight into BRAF mutations in cancer. Curr. Opin. Genet. Dev. 2007, 17, 31–39. [Google Scholar] [CrossRef] [PubMed]
- Damsky, W.E.; Bosenberg, M. Melanocytic nevi and melanoma: Unraveling a complex relationship. Oncogene 2017, 36, 5771–5792. [Google Scholar] [CrossRef] [Green Version]
- Kuwata, T.; Kitagawa, M.; Kasuga, T. Proliferative activity of primary cutaneous melanocytic tumours. Virchows Arch. A Pathol. Anat. Histopathol. 1993, 423, 359–364. [Google Scholar] [CrossRef]
- Maldonado, J.L.; Timmerman, L.; Fridlyand, J.; Bastian, B.C. Mechanisms of cell-cycle arrest in spitz nevi with constitutive activation of the MAP-kinase pathway. Am. J. Pathol. 2004, 164, 1783–1787. [Google Scholar] [CrossRef] [Green Version]
- Michaloglou, C.; Vredeveld, L.C.; Soengas, M.S.; Denoyelle, C.; Kuilman, T.; van der Horst, C.M.; Majoor, D.M.; Shay, J.W.; Mooi, W.J.; Peeper, D.S. BRAFE600-associated senescence-like cell cycle arrest of human naevi. Nature 2005, 436, 720–724. [Google Scholar] [CrossRef]
- Gray-Schopfer, V.C.; Cheong, S.C.; Chong, H.; Chow, J.; Moss, T.; Abdel-Malek, Z.A.; Marais, R.; Wynford-Thomas, D.; Bennett, D.C. Cellular senescence in naevi and immortalisation in melanoma: A role for p16? Br. J. Cancer 2006, 95, 496–505. [Google Scholar] [CrossRef] [Green Version]
- Dhomen, N.; Reis-Filho, J.S.; da Rocha Dias, S.; Hayward, R.; Savage, K.; Delmas, V.; Larue, L.; Pritchard, C.; Marais, R. Oncogenic BRAF induces melanocyte senescence and melanoma in mice. Cancer Cell 2009, 15, 294–303. [Google Scholar] [CrossRef]
- Huang, J.M.; Chikeka, I.; Hornyak, T.J. Melanocytic nevi and the genetic and epigenetic control of oncogene-induced senescence. Dermatol. Clin. 2017, 35, 85–93. [Google Scholar] [CrossRef] [Green Version]
- Uribe, P.; Andrade, L.; Gonzalez, S. Lack of association between BRAF mutation and MAPK ERK activation in melanocytic nevi. J. Investig. Dermatol. 2006, 126, 161–166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Damsky, W.; Micevic, G.; Meeth, K.; Muthusamy, V.; Curley, D.P.; Santhanakrishnan, M.; Erdelyi, I.; Platt, J.T.; Huang, L.; Theodosakis, N.; et al. MTORC1 activation blocks BRAFV600E-induced growth arrest but is insufficient for melanoma formation. Cancer Cell 2015, 27, 41–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Proietti, I.; Skroza, N.; Michelini, S.; Mambrin, A.; Balduzzi, V.; Bernardini, N.; Marchesiello, A.; Tolino, E.; Volpe, S.; Maddalena, P.; et al. BRAF Inhibitors: Molecular targeting and immunomodulatory actions. Cancers 2020, 12, 1823. [Google Scholar] [CrossRef] [PubMed]
- Croce, L.; Coperchini, F.; Magri, F.; Chiovato, L.; Rotondi, M. The multifaceted anti-cancer effects of BRAF-inhibitors. Oncotarget 2019, 10, 6623–6640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Young, H.L.; Rowling, E.J.; Bugatti, M.; Giurisato, E.; Luheshi, N.; Arozarena, I.; Acosta, J.C.; Kamarashev, J.; Frederick, D.T.; Cooper, Z.A.; et al. An adaptive signaling network in melanoma inflammatory niches confers tolerance to MAPK signaling inhibition. J. Exp. Med. 2017, 214, 1691–1710. [Google Scholar] [CrossRef] [Green Version]
- Diazzi, S.; Tartare-Deckert, S.; Deckert, M. Bad neighborhood: Fibrotic stroma as a new player in melanoma resistance to targeted therapies. Cancers 2020, 12, 1364. [Google Scholar] [CrossRef]
- Atzori, M.G.; Ceci, C.; Ruffini, F.; Trapani, M.; Barbaccia, M.L.; Tentori, L.; D’Atri, S.; Lacal, P.M.; Graziani, G. Role of VEGFR-1 in melanoma acquired resistance to the BRAF inhibitor vemurafenib. J. Cell. Mol. Med. 2020, 24, 465–475. [Google Scholar] [CrossRef] [Green Version]
- Khalili, J.S.; Liu, S.; Rodríguez-Cruz, T.G.; Whittington, M.; Wardell, S.; Liu, C.; Zhang, M.; Cooper, Z.A.; Frederick, D.T.; Li, Y.; et al. Oncogenic BRAF(V600E) promotes stromal cell-mediated immunosuppression via induction of interleukin-1 in melanoma. Clin. Cancer Res. 2012, 18, 5329–5340. [Google Scholar] [CrossRef] [Green Version]
- Fedorenko, I.V.; Wargo, J.A.; Flaherty, K.T.; Messina, J.L.; Smalley, K.S.M. BRAF inhibition generates a host-tumor niche that mediates therapeutic escape. J. Investig. Dermatol. 2015, 135, 3115–3124. [Google Scholar] [CrossRef] [Green Version]
- Hirata, E.; Girotti, M.R.; Viros, A.; Hooper, S.; Spencer-Dene, B.; Matsuda, M.; Larkin, J.; Marais, R.; Sahai, E. Intravital imaging reveals how BRAF inhibition generates drug-tolerant microenvironments with high integrin β1/FAK signaling. Cancer Cell 2015, 27, 574–588. [Google Scholar] [CrossRef] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Bellei, B.; Migliano, E.; Picardo, M. A Framework of Major Tumor-Promoting Signal Transduction Pathways Implicated in Melanoma-Fibroblast Dialogue. Cancers 2020, 12, 3400. https://doi.org/10.3390/cancers12113400
Bellei B, Migliano E, Picardo M. A Framework of Major Tumor-Promoting Signal Transduction Pathways Implicated in Melanoma-Fibroblast Dialogue. Cancers. 2020; 12(11):3400. https://doi.org/10.3390/cancers12113400
Chicago/Turabian StyleBellei, Barbara, Emilia Migliano, and Mauro Picardo. 2020. "A Framework of Major Tumor-Promoting Signal Transduction Pathways Implicated in Melanoma-Fibroblast Dialogue" Cancers 12, no. 11: 3400. https://doi.org/10.3390/cancers12113400
APA StyleBellei, B., Migliano, E., & Picardo, M. (2020). A Framework of Major Tumor-Promoting Signal Transduction Pathways Implicated in Melanoma-Fibroblast Dialogue. Cancers, 12(11), 3400. https://doi.org/10.3390/cancers12113400