Journal of Translational Medicine (Jan 2024)

Novel cellular systems unveil mucosal melanoma initiating cells and a role for PI3K/Akt/mTOR pathway in mucosal melanoma fitness

  • Matilde Monti,
  • Luisa Benerini Gatta,
  • Mattia Bugatti,
  • Irene Pezzali,
  • Sara Picinoli,
  • Marcello Manfredi,
  • Antonio Lavazza,
  • Virginia Vita Vanella,
  • Veronica De Giorgis,
  • Lucia Zanatta,
  • Francesco Missale,
  • Silvia Lonardi,
  • Benedetta Zanetti,
  • Giovanni Bozzoni,
  • Moris Cadei,
  • Andrea Abate,
  • Barbara Vergani,
  • Piera Balzarini,
  • Simonetta Battocchio,
  • Carla Facco,
  • Mario Turri-Zanoni,
  • Paolo Castelnuovo,
  • Piero Nicolai,
  • Ester Fonsatti,
  • Biagio Eugenio Leone,
  • Emilio Marengo,
  • Sandra Sigala,
  • Roberto Ronca,
  • Michela Perego,
  • Davide Lombardi,
  • William Vermi

DOI
https://doi.org/10.1186/s12967-023-04784-2
Journal volume & issue
Vol. 22, no. 1
pp. 1 – 24

Abstract

Read online

Abstract Background Mucosal Melanomas (MM) are highly aggressive neoplasms arising from mucosal melanocytes. Current treatments offer a limited survival benefit for patients with advanced MM; moreover, the lack of pre-clinical cellular systems has significantly limited the understanding of their immunobiology. Methods Five novel cell lines were obtained from patient-derived biopsies of MM arising in the sino-nasal mucosa and designated as SN-MM1-5. The morphology, ultrastructure and melanocytic identity of SN-MM cell lines were validated by transmission electron microscopy and immunohistochemistry. Moreover, in vivo tumorigenicity of SN-MM1-5 was tested by subcutaneous injection in NOD/SCID mice. Molecular characterization of SN-MM cell lines was performed by a mass-spectrometry proteomic approach, and their sensitivity to PI3K chemical inhibitor LY294002 was validated by Akt activation, measured by pAkt(Ser473) and pAkt(Thr308) in immunoblots, and MTS assay. Results This study reports the validation and functional characterization of five newly generated SN-MM cell lines. Compared to the normal counterpart, the proteomic profile of SN-MM is consistent with transformed melanocytes showing a heterogeneous degree of melanocytic differentiation and activation of cancer-related pathways. All SN-MM cell lines resulted tumorigenic in vivo and display recurrent structural variants according to aCGH analysis. Of relevance, the microscopic analysis of the corresponding xenotransplants allowed the identification of clusters of MITF-/CDH1-/CDH2 + /ZEB1 + /CD271 + cells, supporting the existence of melanoma-initiating cells also in MM, as confirmed in clinical samples. In vitro, SN-MM cell lines were sensitive to cisplatin, but not to temozolomide. Moreover, the proteomic analysis of SN-MM cell lines revealed that RICTOR, a subunit of mTORC2 complex, is the most significantly activated upstream regulator, suggesting a relevant role for the PI3K-Akt-mTOR pathway in these neoplasms. Consistently, phosphorylation of NDRG1 and Akt activation was observed in SN-MM, the latter being constitutive and sustained by PTEN loss in SN-MM2 and SN-MM3. The cell viability impairment induced by LY294002 confirmed a functional role for the PI3K-Akt-mTOR pathway in SN-MM cell lines. Conclusions Overall, these novel and unique cellular systems represent relevant experimental tools for a better understanding of the biology of these neoplasms and, as an extension, to MM from other sites.

Keywords