PLoS Computational Biology (Apr 2023)

A combined experimental-computational approach uncovers a role for the Golgi matrix protein Giantin in breast cancer progression.

  • Salim Ghannoum,
  • Damiano Fantini,
  • Muhammad Zahoor,
  • Veronika Reiterer,
  • Santosh Phuyal,
  • Waldir Leoncio Netto,
  • Øystein Sørensen,
  • Arvind Iyer,
  • Debarka Sengupta,
  • Lina Prasmickaite,
  • Gunhild Mari Mælandsmo,
  • Alvaro Köhn-Luque,
  • Hesso Farhan

DOI
https://doi.org/10.1371/journal.pcbi.1010995
Journal volume & issue
Vol. 19, no. 4
p. e1010995

Abstract

Read online

Our understanding of how speed and persistence of cell migration affects the growth rate and size of tumors remains incomplete. To address this, we developed a mathematical model wherein cells migrate in two-dimensional space, divide, die or intravasate into the vasculature. Exploring a wide range of speed and persistence combinations, we find that tumor growth positively correlates with increasing speed and higher persistence. As a biologically relevant example, we focused on Golgi fragmentation, a phenomenon often linked to alterations of cell migration. Golgi fragmentation was induced by depletion of Giantin, a Golgi matrix protein, the downregulation of which correlates with poor patient survival. Applying the experimentally obtained migration and invasion traits of Giantin depleted breast cancer cells to our mathematical model, we predict that loss of Giantin increases the number of intravasating cells. This prediction was validated, by showing that circulating tumor cells express significantly less Giantin than primary tumor cells. Altogether, our computational model identifies cell migration traits that regulate tumor progression and uncovers a role of Giantin in breast cancer progression.