Decoding CD271 in melanoma: Comparison
Please note this is a comparison between Version 3 by Karina Chen and Version 2 by Torben Redmer.

The evolution of melanoma, the most aggressive type of skin cancer, is triggered by

driver mutations that are acquired in the coding regions of particularly BRAF (rat fibrosarcoma

serine/threonine kinase, isoform B) or NRAS (neuroblastoma-type ras sarcoma virus) in melanocytes.

Although driver mutations strongly determine tumor progression, additional factors are likely

required and prerequisite for melanoma formation. Melanocytes are formed during vertebrate

development in a well-controlled di erentiation process of multipotent neural crest stem cells

(NCSCs). However, mechanisms determining the properties of melanocytes and melanoma cells

are still not well understood. The nerve growth factor receptor CD271 is likewise expressed in

melanocytes, melanoma cells and NCSCs and programs the maintenance of a stem-like and migratory

phenotype via a comprehensive network of associated genes. Moreover, CD271 regulates phenotype

switching, a process that enables the rapid and reversible conversion of proliferative into invasive or

non-stem-like states into stem-like states by yet largely unknown mechanisms. Here, we summarize

current findings about CD271-associated mechanisms in melanoma cells and illustrate the role of

CD271 for melanoma cell migration and metastasis, phenotype-switching, resistance to therapeutic

interventions, and the maintenance of an NCSC-like state.

  • CD271
  • melanoma
  • neural crest stem cells
  • migration
  • metastasis
Please wait, diff process is still running!

References

  1. Balch, C.M.; Gershenwald, J.E.; Soong, S.J.; Thompson, J.F.; Atkins, M.B.; Byrd, D.R.; Buzaid, A.C.; Cochran, A.J.; Coit, D.G.; Ding, S.; et al. Final version of 2009 AJCC melanoma staging and classification. J. Clin. Oncol. 2009, 27, 6199. [Google Scholar] [CrossRef] [PubMed]
  2. Obenauf, A.C.; Massague, J. Surviving at a distance: Organ-specific metastasis. Trends Cancer 2015, 1, 76–91. [Google Scholar] [CrossRef] [PubMed]
  3. Shibue, T.; Weinberg, R.A. Metastatic colonization: Settlement, adaptation and propagation of tumor cells in a foreign tissue environment. Semin. Cancer Biol. 2011, 21, 99–106. [Google Scholar] [CrossRef] [PubMed]
  4. Obenauf, A.C.; Zou, Y.; Ji, A.L.; Vanharanta, S.; Shu, W.; Shi, H.; Kong, X.; Bosenberg, M.C.; Wiesner, T.; Rosen, N.; et al. Therapy-induced tumour secretomes promote resistance and tumour progression. Nature 2015, 520, 368–372. [Google Scholar] [CrossRef] [PubMed]
  5. Plaks, V.; Kong, N.; Werb, Z. The cancer stem cell niche: How essential is the niche in regulating stemness of tumor cells? Cell Stem Cell 2015, 16, 225–238. [Google Scholar] [CrossRef] [PubMed]
  6. Herrmann, J.L.; Menter, D.G.; Hamada, J.; Marchetti, D.; Nakajima, M.; Nicolson, G.L. Mediation of ngf-stimulated extracellular matrix invasion by the human melanoma low-affinity p75 neurotrophin receptor: Melanoma p75 functions independently of trka. Mol. Biol. Cell 1993, 4, 1205–1216. [Google Scholar] [CrossRef]
  7. Marchetti, D.; Menter, D.; Jin, L.; Nakajima, M.; Nicolson, G.L. Nerve growth factor effects on human and mouse melanoma cell invasion and heparanase production. Int. J. Cancer 1993, 55, 692–699. [Google Scholar] [CrossRef]
  8. Boiko, A.D.; Razorenova, O.V.; van de Rijn, M.; Swetter, S.M.; Johnson, D.L.; Ly, D.P.; Butler, P.D.; Yang, G.P.; Joshua, B.; Kaplan, M.J.; et al. Human melanoma-initiating cells express neural crest nerve growth factor receptor cd271. Nature 2010, 466, 133–137. [Google Scholar] [CrossRef]
  9. Quintana, E.; Shackleton, M.; Foster, H.R.; Fullen, D.R.; Sabel, M.S.; Johnson, T.M.; Morrison, S.J. Phenotypic heterogeneity among tumorigenic melanoma cells from patients that is reversible and not hierarchically organized. Cancer Cell 2010, 18, 510–523. [Google Scholar] [CrossRef]
  10. Vandamme, N.; Berx, G. From neural crest cells to melanocytes: Cellular plasticity during development and beyond. Cell Mol. Life Sci. 2019, 76, 1919–1934. [Google Scholar] [CrossRef]
  11. Moscatelli, I.; Pierantozzi, E.; Camaioni, A.; Siracusa, G.; Campagnolo, L. P75 neurotrophin receptor is involved in proliferation of undifferentiated mouse embryonic stem cells. Exp. Cell Res. 2009, 315, 3220–3232. [Google Scholar] [CrossRef] [PubMed]
  12. Morrison, S.J.; White, P.M.; Zock, C.; Anderson, D.J. Prospective identification, isolation by flow cytometry, and in vivo self-renewal of multipotent mammalian neural crest stem cells. Cell 1999, 96, 737–749. [Google Scholar] [CrossRef]
  13. Betters, E.; Liu, Y.; Kjaeldgaard, A.; Sundstrom, E.; Garcia-Castro, M.I. Analysis of early human neural crest development. Dev. Biol. 2010, 344, 578–592. [Google Scholar] [CrossRef] [PubMed]
  14. Wislet, S.; Vandervelden, G.; Rogister, B. From neural crest development to cancer and vice versa: How p75(ntr) and (pro)neurotrophins could act on cell migration and invasion? Front. Mol. Neurosci. 2018, 11, 244. [Google Scholar] [CrossRef]
  15. Lee, G.; Kim, H.; Elkabetz, Y.; al Shamy, G.; Panagiotakos, G.; Barberi, T.; Tabar, V.; Studer, L. Isolation and directed differentiation of neural crest stem cells derived from human embryonic stem cells. Nat. Biotechnol. 2007, 25, 1468–1475. [Google Scholar] [CrossRef]
  16. Tomellini, E.; Lagadec, C.; Polakowska, R.; le Bourhis, X. Role of p75 neurotrophin receptor in stem cell biology: More than just a marker. Cell Mol. Life Sci. 2014, 71, 2467–2481. [Google Scholar] [CrossRef]
  17. Mundell, A.N.; Labosky, P.A. Neural crest stem cell multipotency requires foxd3 to maintain neural potential and repress mesenchymal fates. Development 2011, 138, 641–652. [Google Scholar] [CrossRef]
  18. Teng, L.; Mundell, N.A.; Frist, A.Y.; Wang, Q.; Labosky, P.A. Requirement for foxd3 in the maintenance of neural crest progenitors. Development 2008, 135, 1615–1624. [Google Scholar] [CrossRef]
  19. Douarin, L.M.N.; Dupin, E. Multipotentiality of the neural crest. Curr. Opin. Genet. Dev. 2003, 13, 529–536. [Google Scholar] [CrossRef]
  20. Mort, R.L.; Jackson, I.J.; Patton, E.E. The melanocyte lineage in development and disease. Development 2015, 142, 620–632. [Google Scholar] [CrossRef]
  21. Britsch, S.; Goerich, D.E.; Riethmacher, D.; Peirano, R.I.; Rossner, M.; Nave, K.A.; Birchmeier, C.; Wegner, M. The transcription factor sox10 is a key regulator of peripheral glial development. Genes Dev. 2001, 15, 66–78. [Google Scholar] [CrossRef] [PubMed]
  22. Shakhova, O.; Zingg, D.; Schaefer, S.M.; Hari, L.; Civenni, G.; Blunschi, J.; Claudinot, S.; Okoniewski, M.; Beermann, F.; Mihic-Probst, D.; et al. Sox10 promotes the formation and maintenance of giant congenital naevi and melanoma. Nat. Cell Biol. 2012, 14, 882–890. [Google Scholar] [CrossRef] [PubMed]
  23. Barrallo-Gimeno, A.; Nieto, M.A. The snail genes as inducers of cell movement and survival: Implications in development and cancer. Development 2005, 132, 3151–3161. [Google Scholar] [CrossRef] [PubMed]
  24. White, M.R.; Zon, L.I. Melanocytes in development, regeneration, and cancer. Cell Stem Cell 2008, 3, 242–252. [Google Scholar] [CrossRef]
  25. Blake, J.A.; Ziman, M.R. Pax genes: Regulators of lineage specification and progenitor cell maintenance. Development 2014, 141, 737–751. [Google Scholar] [CrossRef] [PubMed]
  26. Pincelli, C. P75 neurotrophin receptor in the skin: Beyond its neurotrophic function. Front. Med. (Lausanne) 2017, 4, 22. [Google Scholar] [CrossRef]
  27. Truzzi, F.; Saltari, A.; Palazzo, E.; Lotti, R.; Petrachi, T.; Dallaglio, K.; Gemelli, C.; Grisendi, G.; Dominici, M.; Pincelli, C.; et al. Cd271 mediates stem cells to early progeny transition in human epidermis. J. Investig. Dermatol. 2015, 135, 786–795. [Google Scholar] [CrossRef]
  28. Peacocke, M.; Yaar, M.; Mansur, C.P.; Chao, M.V.; Gilchrest, B.A. Induction of nerve growth factor receptors on cultured human melanocytes. Proc. Natl. Acad. Sci. USA 1988, 85, 5282–5286. [Google Scholar] [CrossRef]
  29. Yaar, M.; Grossman, K.; Eller, M.; Gilchrest, B.A. Evidence for nerve growth factor-mediated paracrine effects in human epidermis. J. Cell Biol. 1991, 115, 821–828. [Google Scholar] [CrossRef]
  30. Tron, V.A.; Coughlin, M.D.; Jang, D.E.; Stanisz, J.; Sauder, D.N. Expression and modulation of nerve growth factor in murine keratinocytes (pam 212). J. Clin. Investig. 1990, 85, 1085–1089. [Google Scholar] [CrossRef]
  31. Menzies, A.M.; Haydu, L.E.; Visintin, L.; Carlino, M.S.; Howle, J.R.; Thompson, J.F.; Kefford, R.F.; Scolyer, R.A.; Long, G.V. Distinguishing clinicopathologic features of patients with v600e and v600k braf-mutant metastatic melanoma. Clin. Cancer Res. 2012, 18, 3242–3249. [Google Scholar] [CrossRef] [PubMed]
  32. Pollock, P.M.; Harper, U.L.; Hansen, K.S.; Yudt, L.M.; Stark, M.; Robbins, C.M.; Moses, T.Y.; Hostetter, G.; Wagner, U.; Kakareka, J.; et al. High frequency of braf mutations in nevi. Nat. Genet. 2003, 33, 19–20. [Google Scholar] [CrossRef] [PubMed]
  33. Cheung, M.; Sharma, A.; Madhunapantula, S.V.; Robertson, G.P. Akt3 and mutant v600e b-raf cooperate to promote early melanoma development. Cancer Res. 2008, 68, 3429–3439. [Google Scholar] [CrossRef] [PubMed]
  34. Davies, H.; Bignell, G.R.; Cox, C.; Stephens, P.; Edkins, S.; Clegg, S.; Teague, J.; Woffendin, H.; Garnett, M.J.; Bottomley, W.; et al. Mutations of the braf gene in human cancer. Nature 2002, 417, 949–954. [Google Scholar] [CrossRef] [PubMed]
  35. Shain, A.H.; Bastian, B.C. From melanocytes to melanomas. Nat. Rev. Cancer 2016, 16, 345–358. [Google Scholar] [CrossRef]
  36. Shain, A.H.; Bastian, B.C. Author correction: From melanocytes to melanomas. Nat. Rev. Cancer 2020, 20, 355. [Google Scholar] [CrossRef]
  37. Dankort, D.; Curley, D.P.; Nelson, A.N.; Karnezis, W.E.; Damsky, J.; You, R.A.D.; Bosenberg, M. Braf(v600e) cooperates with pten loss to induce metastatic melanoma. Nat. Genet. 2009, 41, 544–552. [Google Scholar] [CrossRef]
  38. Perez-Guijarro, E.; Day, C.P.; Merlino, G.; Zaidi, M.R. Genetically engineered mouse models of melanoma. Cancer 2017, 123, 2089–2103. [Google Scholar] [CrossRef]
  39. Stahl, J.M.; Sharma, A.; Cheung, M.; Zimmerman, M.; Cheng, J.Q.; Bosenberg, M.W.; Kester, M.; Sandirasegarane, L.; Robertson, G.P. Deregulated akt3 activity promotes development of malignant melanoma. Cancer Res. 2004, 64, 7002–7010. [Google Scholar] [CrossRef]
  40. Redmer, T.; Welte, Y.; Behrens, D.; Fichtner, I.; Przybilla, D.; Wruck, W.; Yaspo, M.L.; Lehrach, H.; Schafer, R.; Regenbrecht, C.R. The nerve growth factor receptor cd271 is crucial to maintain tumorigenicity and stem-like properties of melanoma cells. PLoS ONE 2014, 9, e92596. [Google Scholar] [CrossRef]
  41. Adameyko, I.; Lallemend, F.; Furlan, A.; Zinin, N.; Aranda, S.; Kitambi, S.S.; Blanchart, A.; Favaro, R.; Nicolis, S.; Lubke, M.; et al. Sox2 and mitf cross-regulatory interactions consolidate progenitor and melanocyte lineages in the cranial neural crest. Development 2012, 139, 397–410. [Google Scholar] [CrossRef] [PubMed]
  42. Hartman, M.L.; Czyz, M. Mitf in melanoma: Mechanisms behind its expression and activity. Cell Mol. Life Sci. 2015, 72, 1249–1260. [Google Scholar] [CrossRef] [PubMed]
  43. Redmer, T.; Walz, I.; Klinger, B.; Khouja, S.; Welte, Y.; Schafer, R.; Regenbrecht, C. The role of the cancer stem cell marker cd271 in DNA damage response and drug resistance of melanoma cells. Oncogenesis 2017, 6, e291. [Google Scholar] [CrossRef] [PubMed]
  44. Lehraiki, A.; Cerezo, M.; Rouaud, F.; Abbe, P.; Allegra, M.; Kluza, J.; Marchetti, P.; Imbert, V.; Cheli, Y.; Bertolotto, C.; et al. Increased cd271 expression by the nf-kb pathway promotes melanoma cell survival and drives acquired resistance to braf inhibitor vemurafenib. Cell Discov. 2015, 1, 15030. [Google Scholar] [CrossRef] [PubMed]
  45. Zubrilov, I.; Sagi-Assif, O.; Izraely, S.; Meshel, T.; Ben-Menahem, S.; Ginat, R.; Pasmanik-Chor, M.; Nahmias, C.; Couraud, P.O.; Hoon, D.S.; et al. Vemurafenib resistance selects for highly malignant brain and lung-metastasizing melanoma cells. Cancer Lett. 2015, 361, 86–96. [Google Scholar] [CrossRef]
  46. Baeza-Raja, B.; Eckel-Mahan, K.; Zhang, L.; Vagena, E.; Tsigelny, I.F.; Sassone-Corsi, P.; Ptacek, L.J.; Akassoglou, K. P75 neurotrophin receptor is a clock gene that regulates oscillatory components of circadian and metabolic networks. J. Neurosci. 2013, 33, 10221–10234. [Google Scholar] [CrossRef]
  47. Boyle, S.E.; Fedele, C.G.; Corbin, V.; Wybacz, E.; Szeto, P.; Lewin, J.; Young, R.J.; Wong, A.; Fuller, R.; Spillane, J.; et al. Cd271 expression on patient melanoma cells is unstable and unlinked to tumorigenicity. Cancer Res. 2016, 76, 3965–3977. [Google Scholar] [CrossRef]
  48. Cheli, Y.; Bonnazi, V.F.; Jacquel, A.; Allegra, M.; de Donatis, G.M.; Bahadoran, P.; Bertolotto, C.; Ballotti, R. Cd271 is an imperfect marker for melanoma initiating cells. Oncotarget 2014, 5, 5272–5283. [Google Scholar] [CrossRef]
  49. Filipp, F.V.; Li, C.; Boiko, A.D. Cd271 is a molecular switch with divergent roles in melanoma and melanocyte development. Sci. Rep. 2019, 9, 7696. [Google Scholar] [CrossRef]
  50. Radke, J.; Rossner, F.; Redmer, T. Cd271 determines migratory properties of melanoma cells. Sci. Rep. 2017, 7, 9834. [Google Scholar] [CrossRef]
  51. Civenni, G.; Walter, A.; Kobert, N.; Mihic-Probst, D.; Zipser, M.; Belloni, B.; Seifert, B.; Moch, H.; Dummer, R.; van den Broek, M.; et al. Human cd271-positive melanoma stem cells associated with metastasis establish tumor heterogeneity and long-term growth. Cancer Res. 2011, 71, 3098–3109. [Google Scholar] [CrossRef] [PubMed]
  52. Mandalos, N.; Rhinn, M.; Granchi, Z.; Karampelas, I.; Mitsiadis, T.; Economides, A.N.; Dolle, P.; Remboutsika, E. Sox2 acts as a rheostat of epithelial to mesenchymal transition during neural crest development. Front. Physiol. 2014, 5, 345. [Google Scholar] [CrossRef] [PubMed]
  53. Restivo, G.; Diener, J.; Cheng, P.F.; Kiowski, G.; Bonalli, M.; Biedermann, T.; Reichmann, E.; Levesque, M.P.; Dummer, R.; Sommer, L. Publisher correction: The low affinity neurotrophin receptor cd271 regulates phenotype switching in melanoma. Nat. Commun. 2018, 9, 314. [Google Scholar] [CrossRef] [PubMed]
  54. Ackermann, J.; Frutschi, M.; Kaloulis, K.; McKee, T.; Trumpp, A.; Beermann, F. Metastasizing melanoma formation caused by expression of activated n-rasq61k on an ink4a-deficient background. Cancer Res. 2005, 65, 4005–4011. [Google Scholar] [CrossRef] [PubMed]
  55. Sandru, A.; Voinea, S.; Panaitescu, E.; Blidaru, A. Survival rates of patients with metastatic malignant melanoma. J. Med. Life 2014, 7, 572–576. [Google Scholar]
  56. Redmer, T. Deciphering mechanisms of brain metastasis in melanoma-the gist of the matter. Mol. Cancer 2018, 17, 106. [Google Scholar] [CrossRef]
  57. Bailey, C.M.; Morrison, J.A.; Kulesa, P.M. Melanoma revives an embryonic migration program to promote plasticity and invasion. Pigment Cell Melanoma Res. 2012, 25, 573–583. [Google Scholar] [CrossRef]
  58. Larribere, L.; Utikal, J. Stem cell-derived models of neural crest are essential to understand melanoma progression and therapy resistance. Front. Mol. Neurosci. 2019, 12, 111. [Google Scholar] [CrossRef]
  59. Marchetti, D.; McCutcheon, I.; Ross, M.; Nicolson, G. Inverse expression of neurotrophins and neurotrophin receptors at the invasion front of human-melanoma brain metastases. Int J. Oncol. 1995, 7, 87–94. [Google Scholar] [CrossRef]
  60. Truzzi, F.; Marconi, A.; Lotti, R.; Dallaglio, K.; French, L.E.; Hempstead, B.L.; Pincelli, C. Neurotrophins and their receptors stimulate melanoma cell proliferation and migration. J. Investig. Dermatol. 2008, 128, 2031–2040. [Google Scholar] [CrossRef]
  61. Liu, X.; Gao, Y.; Lu, Y.; Zhang, J.; Li, L.; Yin, F. Upregulation of nek2 is associated with drug resistance in ovarian cancer. Oncol Rep. 2014, 31, 745–754. [Google Scholar] [CrossRef] [PubMed]
  62. Xu, H.; Zeng, L.; Guan, Y.; Feng, X.; Zhu, Y.; Lu, Y.; Shi, C.; Chen, S.; Xia, J.; Guo, J.; et al. High nek2 confers to poor prognosis and contributes to cisplatin-based chemotherapy resistance in nasopharyngeal carcinoma. J. Cell Biochem. 2019, 120, 3547–3558. [Google Scholar] [CrossRef] [PubMed]
  63. Chang, Y.Y.; Yen, C.J.; Chan, S.H.; Chou, Y.W.; Lee, Y.P.; Bao, C.Y.; Huang, C.J.; Huang, W. Nek2 promotes hepatoma metastasis and serves as biomarker for high recurrence risk after hepatic resection. Ann. Hepatol. 2018, 17, 843–856. [Google Scholar] [CrossRef]
  64. Huang, J.; Sun, S.G.; Hou, S. Aberrant NEK2 expression might be an independent predictor for poor recurrence-free survival and overall survival of skin cutaneous melanoma. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 3694–3702. [Google Scholar] [CrossRef]
  65. Wen, S.; Liu, Y.; Yang, M.; Yang, K.; Huang, J.; Feng, D. Increased nek2 in hepatocellular carcinoma promotes cancer progression and drug resistance by promoting pp1/akt and wnt activation. Oncol. Rep. 2016, 36, 2193–2199. [Google Scholar] [CrossRef]
  66. Zhang, Y.; Wang, W.; Wang, Y.; Huang, X.; Zhang, Z.; Chen, B.; Xie, W.; Li, S.; Shen, S.; Peng, B. Nek2 promotes hepatocellular carcinoma migration and invasion through modulation of the epithelial-mesenchymal transition. Oncol. Rep. 2018, 39, 1023–1033. [Google Scholar] [CrossRef]
  67. Sankaran, D.; Pakala, S.B.; Nair, V.S.; Sirigiri, D.N.; Cyanam, D.; Ha, N.H.; Li, D.Q.; Santhoshkumar, T.R.; Pillai, M.R.; Kumar, R. Mechanism of mta1 protein overexpression-linked invasion: Mta1 regulation of hyaluronan-mediated motility receptor (hmmr) expression and function. J. Biol. Chem. 2012, 287, 5483–5491. [Google Scholar] [CrossRef] [PubMed]
  68. Tilghman, J.; Wu, H.; Sang, Y.; Shi, X.; Guerrero-Cazares, H.; Quinones-Hinojosa, A.; Eberhart, C.G.; Laterra, J.; Ying, M. Hmmr maintains the stemness and tumorigenicity of glioblastoma stem-like cells. Cancer Res. 2014, 74, 3168–3179. [Google Scholar] [CrossRef]
  69. Kauffmann, A.; Rosselli, F.; Lazar, V.; Winnepenninckx, V.; Mansuet-Lupo, A.; Dessen, P.; van den Oord, J.J.; Spatz, A.; Sarasin, A. High expression of DNA repair pathways is associated with metastasis in melanoma patients. Oncogene 2008, 27, 565–573. [Google Scholar] [CrossRef]
  70. Lu, H.; Liu, S.; Zhang, G.; Kwong, L.N.; Zhu, Y.; Miller, J.P.; Hu, Y.; Zhong, W.; Zeng, J.; Wu, L.; et al. Oncogenic braf-mediated melanoma cell invasion. Cell Rep. 2016, 15, 2012–2024. [Google Scholar] [CrossRef]
  71. Ravindran Menon, D.; Das, S.; Krepler, C.; Vultur, A.; Rinner, B.; Schauer, S.; Kashofer, K.; Wagner, K.; Zhang, G.; Rad, E.B.; et al. A stress-induced early innate response causes multidrug tolerance in melanoma. Oncogene 2015, 34, 4448–4459. [Google Scholar] [CrossRef] [PubMed]
  72. Tong, B.; Pantazopoulou, V.; Johansson, E.; Pietras, A. The p75 neurotrophin receptor enhances hif-dependent signaling in glioma. Exp. Cell Res. 2018, 371, 122–129. [Google Scholar] [CrossRef] [PubMed]
  73. Calvo-Anguiano, G.; Lugo-Trampe, J.J.; Camacho, A.; Said-Fernandez, S.; Mercado-Hernandez, R.; Zomosa-Signoret, V.; Rojas-Martinez, A.; Ortiz-Lopez, R. Comparison of specific expression profile in two in vitro hypoxia models. Exp. Ther. Med. 2018, 15, 4777–4784. [Google Scholar] [CrossRef] [PubMed]
  74. Dusedau, H.P.; Kleveman, J.; Figueiredo, C.A.; Biswas, A.; Steffen, J.; Kliche, S.; Haak, S.; Zagrebelsky, M.; Korte, M.; Dunay, I.R. P75(ntr) regulates brain mononuclear cell function and neuronal structure in toxoplasma infection-induced neuroinflammation. Glia 2019, 67, 193–211. [Google Scholar] [CrossRef]
  75. Cragnolini, A.B.; Huang, Y.; Gokina, P.; Friedman, W.J. Nerve growth factor attenuates proliferation of astrocytes via the p75 neurotrophin receptor. Glia 2009, 57, 1386–1392. [Google Scholar] [CrossRef]
  76. Schachtrup, C.; Ryu, J.K.; Mammadzada, K.; Khan, A.S.; Carlton, P.M.; Perez, A.; Christian, F.; le Moan, N.; Vagena, E.; Baeza-Raja, B.; et al. Nuclear pore complex remodeling by p75(ntr) cleavage controls tgf-beta signaling and astrocyte functions. Nat. Neurosci. 2015, 18, 1077–1080. [Google Scholar] [CrossRef]
  77. Landsberg, J.; Kohlmeyer, J.; Renn, M.; Bald, T.; Rogava, M.; Cron, M.; Fatho, M.; Lennerz, V.; Wolfel, T.; Holzel, M.; et al. Melanomas resist t-cell therapy through inflammation-induced reversible dedifferentiation. Nature 2012, 490, 412–416. [Google Scholar] [CrossRef]
  78. Guida, M.; Tommasi, S.; Strippoli, S.; Natalicchio, M.I.; de Summa, S.; Pinto, R.; Cramarossa, A.; Albano, A.; Pisconti, S.; Aieta, M.; et al. The search for a melanoma-tailored chemotherapy in the new era of personalized therapy: A phase ii study of chemo-modulating temozolomide followed by fotemustine and a cooperative study of goim (gruppo oncologico italia meridionale). BMC Cancer 2018, 18, 552. [Google Scholar] [CrossRef]
  79. Ngo, M.; Han, A.; Lakatos, A.; Sahoo, D.; Hachey, S.J.; Weiskopf, K.; Beck, A.H.; Weissman, I.L.; Boiko, A.D. Antibody therapy targeting cd47 and cd271 effectively suppresses melanoma metastasis in patient-derived xenografts. Cell Rep. 2016, 16, 1701–1716. [Google Scholar] [CrossRef]
  80. Tirosh, I.; Izar, B.; Prakadan, S.M.; Wadsworth, M.H.; Treacy, D.; Trombetta, J.J.; Rotem, A.; Rodman, C.; Lian, C.; Murphy, G.; et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell rna-seq. Science 2016, 352, 189–196. [Google Scholar] [CrossRef]
  81. Hoek, K.S.; Eichhoff, O.M.; Schlegel, N.C.; Dobbeling, 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] [PubMed]
  82. Muller, J.; Krijgsman, O.; Tsoi, J.; Robert, L.; Hugo, W.; Song, C.; Kong, X.; Possik, P.A.; Cornelissen-Steijger, P.D.; Foppen , M.H.; et al. Low mitf/axl ratio predicts early resistance to multiple targeted drugs in melanoma. Nat. Commun. 2014, 5, 5712. [Google Scholar] [CrossRef] [PubMed]
  83. Chen, G.; Chakravarti, N.; Aardalen, K.; Lazar, A.J.; Tetzlaff, M.T.; Wubbenhorst, B.; Kim, S.B.; Kopetz, S.; Ledoux, A.A.; Gopal, Y.N.; et al. Molecular profiling of patient-matched brain and extracranial melanoma metastases implicates the pi3k pathway as a therapeutic target. Clin. Cancer Res. 2014, 20, 5537–5546. [Google Scholar] [CrossRef]
  84. Rankin, E.B.; Giaccia, A.J. Hypoxic control of metastasis. Science 2016, 352, 175–180. [Google Scholar] [CrossRef] [PubMed]
  85. Widmer, D.S.; Hoek, K.S.; Cheng, P.F.; Eichhoff, O.M.; Biedermann, T.; Raaijmakers, M.I.G.; Hemmi, S.; Dummer, R.; Levesque, M.P. Hypoxia contributes to melanoma heterogeneity by triggering hif1alpha-dependent phenotype switching. J. Investig. Dermatol. 2013, 133, 2436–2443. [Google Scholar] [CrossRef] [PubMed]
  86. Rappa, G.; Fodstad, O.; Lorico, A. The stem cell-associated antigen cd133 (prominin-1) is a molecular therapeutic target for metastatic melanoma. Stem Cells 2008, 26, 3008–3017. [Google Scholar] [CrossRef]
  87. Guo, R.; Fierro-Fine, A.; Goddard, L.; Russell, M.; Chen, J.; Liu, C.Z.; Fung, K.M.; Hassell, L.A. Increased expression of melanoma stem cell marker cd271 in metastatic melanoma to the brain. Int. J. Clin. Exp. Pathol. 2014, 7, 8947–8951. [Google Scholar]
  88. Podlesniy, P.; Kichev, A.; Pedraza, C.; Saurat, J.; Encinas, M.; Perez, B.; Ferrer, I.; Espinet, C. Pro-ngf from alzheimer’s disease and normal human brain displays distinctive abilities to induce processing and nuclear translocation of intracellular domain of p75ntr and apoptosis. Am. J. Pathol. 2006, 169, 119–131. [Google Scholar] [CrossRef]
  89. Bilderback, T.R.; Grigsby, R.J.; Dobrowsky, R.T. Association of p75(ntr) with caveolin and localization of neurotrophin-induced sphingomyelin hydrolysis to caveolae. J. Biol. Chem. 1997, 272, 10922–10927. [Google Scholar] [CrossRef]
  90. Ceni, C.; Kommaddi, R.P.; Thomas, R.; Vereker, E.; Liu, X.; McPherson, P.S.; Ritter, B.; Barker, P.A. The p75ntr intracellular domain generated by neurotrophin-induced receptor cleavage potentiates trk signaling. J. Cell Sci. 2010, 123, 2299–2307. [Google Scholar] [CrossRef]
  91. van Strien, M.E.; Sluijs, J.A.; Reynolds, B.A.; Steindler, D.A.; Aronica, E.; Hol, E.M. Isolation of neural progenitor cells from the human adult subventricular zone based on expression of the cell surface marker cd271. Stem Cells Transl. Med. 2014, 3, 470–480. [Google Scholar] [CrossRef] [PubMed]
  92. Young, K.M.; Merson, T.D.; Sotthibundhu, A.; Coulson, E.J.; Bartlett, P.F. P75 neurotrophin receptor expression defines a population of bdnf-responsive neurogenic precursor cells. J. Neurosci. 2007, 27, 5146–5155. [Google Scholar] [CrossRef] [PubMed]
  93. Meeker, R.B.; Williams, K.S. The p75 neurotrophin receptor: At the crossroad of neural repair and death. Neural. Regen. Res. 2015, 10, 721–725. [Google Scholar] [CrossRef] [PubMed]
  94. Sofroniew, M.V. Astrogliosis. Cold Spring Harb. Perspect. Biol. 2014, 7, a020420. [Google Scholar] [CrossRef]
  95. Kisiswa, L.; Fernandez-Suarez, D.; Sergaki, M.C.; Ibanez, C.F. Rip2 gates traf6 interaction with death receptor p75(ntr) to regulate cerebellar granule neuron survival. Cell Rep. 2018, 24, 1013–1024. [Google Scholar] [CrossRef]
  96. Meeker, R.; Williams, K. Dynamic nature of the p75 neurotrophin receptor in response to injury and disease. J. Neuroimmune Pharmacol. 2014, 9, 615–628. [Google Scholar] [CrossRef]
  97. Lee, R.; Kermani, P.; Teng, K.K.; Hempstead, B.L. Regulation of cell survival by secreted proneurotrophins. Science 2001, 294, 1945–1948. [Google Scholar] [CrossRef]
  98. Nykjaer, A.; Willnow, T.E. Sortilin: A receptor to regulate neuronal viability and function. Trends Neurosci. 2012, 35, 261–270. [Google Scholar] [CrossRef]
  99. Fujita, Y.; Yamashita, T. Axon growth inhibition by rhoa/rock in the central nervous system. Front. Neurosci. 2014, 8, 338. [Google Scholar] [CrossRef]
  100. Clucas, J.; Valderrama, F. Erm proteins in cancer progression. J. Cell Sci. 2014, 127, 267–275. [Google Scholar] [CrossRef] [PubMed]
  101. Roux, P.P.; Barker, P.A. Neurotrophin signaling through the p75 neurotrophin receptor. Prog. Neurobiol. 2002, 67, 203–233. [Google Scholar] [CrossRef]
  102. Maguire, L.H.; Thomas, A.R.; Goldstein, A.M. Tumors of the neural crest: Common themes in development and cancer. Dev. Dyn. 2015, 244, 311–322. [Google Scholar] [CrossRef] [PubMed]
  103. Shonukan, O.; Bagayogo, I.; McCrea, P.; Chao, M.; Hempstead, B. Neurotrophin-induced melanoma cell migration is mediated through the actin-bundling protein fascin. Oncogene 2003, 22, 3616–3623. [Google Scholar] [CrossRef]
  104. Marchetti, D.; McQuillan, D.J.; Spohn, W.C.; Carson, D.D.; Nicolson, G.L. Neurotrophin stimulation of human melanoma cell invasion: Selected enhancement of heparanase activity and heparanase degradation of specific heparan sulfate subpopulations. Cancer Res. 1996, 56, 2856–2863. [Google Scholar] [CrossRef]
  105. Walch, T.E.; Albino, A.P.; Marchetti, D. Correlation of overexpression of the low-affinity p75 neurotrophin receptor with augmented invasion and heparanase production in human malignant melanoma cells. Int J. Cancer 1999, 82, 112–120. [Google Scholar] [CrossRef]
  106. Yamada, K.; Nomura, N.; Yamano, A.; Yamada, Y.; Wakamatsu, N. Identification and characterization of splicing variants of plekha5 (plekha5) during brain development. Gene 2012, 492, 270–275. [Google Scholar] [CrossRef]
  107. Bao, X.; Shi, J.; Xie, F.; Liu, Z.; Yu, J.; Chen, W.; Zhang, Z.; Xu, Q. Proteolytic release of the p75(ntr) intracellular domain by adam10 promotes metastasis and resistance to anoikis. Cancer Res. 2018, 78, 2262–2276. [Google Scholar] [CrossRef]
  108. Verbeke, S.; Tomellini, E.; Dhamani, F.; Meignan, S.; Adriaenssens, E.; Xuefen le, B. Extracellular cleavage of the p75 neurotrophin receptor is implicated in its pro-survival effect in breast cancer cells. FEBS Lett. 2013, 587, 2591–2596. [Google Scholar] [CrossRef]
  109. Becker, K.; Cana, A.; Baumgartner, W.; Spitzbarth, I. P75 neurotrophin receptor: A double-edged sword in pathology and regeneration of the central nervous system. Vet. Pathol. 2018, 55, 786–801. [Google Scholar] [CrossRef]
  110. Johnston, A.L.; Lun, X.; Rahn, J.J.; Liacini, A.; Wang, L.; Hamilton, M.G.; Parney, I.F.; Hempstead, B.L.; Robbins, S.M.; Forsyth, P.A.; et al. The p75 neurotrophin receptor is a central regulator of glioma invasion. PLoS Biol. 2007, 5, e212. [Google Scholar] [CrossRef] [PubMed]
  111. Wrensch, M.; Minn, Y.; Chew, T.; Bondy, M.; Berger, M.S. Epidemiology of primary brain tumors: Current concepts and review of the literature. Neuro. Oncol. 2002, 4, 278–299. [Google Scholar] [CrossRef] [PubMed]
  112. Ahn, B.Y.; Saldanha-Gama, R.F.; Rahn, J.J.; Hao, X.; Zhang, J.; Dang, N.H.; Alshehri, M.; Robbins, S.M.; Senger, D.L. Glioma invasion mediated by the p75 neurotrophin receptor (p75(ntr)/cd271) requires regulated interaction with pdlim1. Oncogene 2016, 35, 1411–1422. [Google Scholar] [CrossRef] [PubMed]
  113. Higuchi, H.; Yamashita, T.; Yoshikawa, H.; Tohyama, M. Pka phosphorylates the p75 receptor and regulates its localization to lipid rafts. EMBO J. 2003, 22, 1790–1800. [Google Scholar] [CrossRef]
  114. Chen, C.; Shin, J.H.; Eggold, J.T.; Chung, M.K.; Zhang, L.H.; Lee, J.; Sunwoo, J.B. Esm1 mediates ngfr-induced invasion and metastasis in murine oral squamous cell carcinoma. Oncotarget 2016, 7, 70738–70749. [Google Scholar] [CrossRef]
  115. Chung, M.K.; Jung, Y.H.; Lee, J.K.; Cho, S.Y.; Murillo-Sauca, O.; Uppaluri, R.; Shin, J.H.; Sunwoo, J.B. Cd271 confers an invasive and metastatic phenotype of head and neck squamous cell carcinoma through the upregulation of slug. Clin. Cancer Res. 2018, 24, 674–683. [Google Scholar] [CrossRef]
  116. Mochizuki, M.; Tamai, K.; Imai, T.; Sugawara, S.; Ogama, N.; Nakamura, M.; Matsuura, K.; Yamaguchi, K.; Satoh, K.; Sato, I.; et al. Cd271 regulates the proliferation and motility of hypopharyngeal cancer cells. Sci. Rep. 2016, 6, 30707. [Google Scholar] [CrossRef]
  117. Mochizuki, M.; Nakamura, M.; Sibuya, R.; Okazaki, T.; Abe, J.; Nakagawa, T.; Takahashi, S.; Yamazaki, T.; Imai, T.; Takano, A.; et al. Cd271 is a negative prognostic factor and essential for cell proliferation in lung squamous cell carcinoma. Lab. Investig. 2019, 99, 1349–1362. [Google Scholar] [CrossRef]
  118. Morita, S.; Mochizuki, M.; Wada, K.; Shibuya, R.; Nakamura, M.; Yamaguchi, K.; Yamazaki, T.; Imai, T.; Asada, Y.; Matsuura, K.; et al. Humanized anti-cd271 monoclonal antibody exerts an anti-tumor effect by depleting cancer stem cells. Cancer Lett. 2019, 461, 144–152. [Google Scholar] [CrossRef]
  119. Krygier, S.; Djakiew, D. Neurotrophin receptor p75(ntr) suppresses growth and nerve growth factor-mediated metastasis of human prostate cancer cells. Int. J. Cancer 2002, 98, 1–7. [Google Scholar] [CrossRef]
  120. Rosenberg, S.A.; Yang, J.C.; Sherry, R.M.; Kammula, U.S.; Hughes, M.S.; Phan, G.Q.; Citrin, D.E.; Restifo, N.P.; Robbins, P.F.; Wunderlich, J.R.; et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using t-cell transfer immunotherapy. Clin. Cancer Res. 2011, 17, 4550–4557. [Google Scholar] [CrossRef] [PubMed]
  121. Rosenberg, S.A.; Yang, J.C.; Sherry, R.M.; Kammula, U.S.; Hughes, M.S.; Phan, G.Q.; Citrin, D.E.; Restifo, N.P.; Robbins, P.F.; Wunderlich, J.R.; et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using t-cell transfer immunotherapy. Clin. Cancer Res. 2011, 17, 4550–4557. [Google Scholar] [CrossRef] [PubMed]
More
Video Production Service