Experimental and Molecular Medicine (Sep 2024)

Emerin mislocalization during chromatin bridge resolution can drive prostate cancer cell invasiveness in a collagen-rich microenvironment

  • Marta Popęda,
  • Kamil Kowalski,
  • Tomasz Wenta,
  • Galina V. Beznoussenko,
  • Michał Rychłowski,
  • Alexander Mironov,
  • Zeno Lavagnino,
  • Sara Barozzi,
  • Julia Richert,
  • Rebecca Bertolio,
  • Kamil Myszczyński,
  • Jolanta Szade,
  • Michał Bieńkowski,
  • Kevin Miszewski,
  • Marcin Matuszewski,
  • Anna J. Żaczek,
  • Luca Braga,
  • Giannino Del Sal,
  • Natalia Bednarz-Knoll,
  • Paolo Maiuri,
  • Paulina Nastały

DOI
https://doi.org/10.1038/s12276-024-01308-w
Journal volume & issue
Vol. 56, no. 9
pp. 2016 – 2032

Abstract

Read online

Abstract Micronuclei (MN) can form through many mechanisms, including the breakage of aberrant cytokinetic chromatin bridges. The frequent observation of MN in tumors suggests that they might not merely be passive elements but could instead play active roles in tumor progression. Here, we propose a mechanism through which the presence of micronuclei could induce specific phenotypic and functional changes in cells and increase the invasive potential of cancer cells. Through the integration of diverse in vitro imaging and molecular techniques supported by clinical samples from patients with prostate cancer (PCa) defined as high-risk by the D’Amico classification, we demonstrate that the resolution of chromosome bridges can result in the accumulation of Emerin and the formation of Emerin-rich MN. These structures are negative for Lamin A/C and positive for the Lamin-B receptor and Sec61β. MN can act as a protein sinks and result in the pauperization of Emerin from the nuclear envelope. The Emerin mislocalization phenotype is associated with a molecular signature that is correlated with a poor prognosis in PCa patients and is enriched in metastatic samples. Emerin mislocalization corresponds with increases in the migratory and invasive potential of tumor cells, especially in a collagen-rich microenvironment. Our study demonstrates that the mislocalization of Emerin to MN results in increased cell invasiveness, thereby worsening patient prognosis.