Acta Neuropathologica Communications (Sep 2023)

Translocator protein (18kDA) (TSPO) marks mesenchymal glioblastoma cell populations characterized by elevated numbers of tumor-associated macrophages

  • Lorraine Weidner,
  • Julia Lorenz,
  • Stefanie Quach,
  • Frank K. Braun,
  • Tanja Rothhammer-Hampl,
  • Laura-Marie Ammer,
  • Arabel Vollmann-Zwerenz,
  • Laura M. Bartos,
  • Franziska J. Dekorsy,
  • Adrien Holzgreve,
  • Sabrina V. Kirchleitner,
  • Niklas Thon,
  • Tobias Greve,
  • Viktoria Ruf,
  • Jochen Herms,
  • Stefanie Bader,
  • Vladimir M. Milenkovic,
  • Louisa von Baumgarten,
  • Ayse N. Menevse,
  • Abir Hussein,
  • Julian Sax,
  • Christian H. Wetzel,
  • Rainer Rupprecht,
  • Martin Proescholdt,
  • Nils O. Schmidt,
  • Philipp Beckhove,
  • Peter Hau,
  • Joerg-Christian Tonn,
  • Peter Bartenstein,
  • Matthias Brendel,
  • Nathalie L. Albert,
  • Markus J. Riemenschneider

DOI
https://doi.org/10.1186/s40478-023-01651-5
Journal volume & issue
Vol. 11, no. 1
pp. 1 – 21

Abstract

Read online

Abstract TSPO is a promising novel tracer target for positron-emission tomography (PET) imaging of brain tumors. However, due to the heterogeneity of cell populations that contribute to the TSPO-PET signal, imaging interpretation may be challenging. We therefore evaluated TSPO enrichment/expression in connection with its underlying histopathological and molecular features in gliomas. We analyzed TSPO expression and its regulatory mechanisms in large in silico datasets and by performing direct bisulfite sequencing of the TSPO promotor. In glioblastoma tissue samples of our TSPO-PET imaging study cohort, we dissected the association of TSPO tracer enrichment and protein labeling with the expression of cell lineage markers by immunohistochemistry and fluorescence multiplex stains. Furthermore, we identified relevant TSPO-associated signaling pathways by RNA sequencing. We found that TSPO expression is associated with prognostically unfavorable glioma phenotypes and that TSPO promotor hypermethylation is linked to IDH mutation. Careful histological analysis revealed that TSPO immunohistochemistry correlates with the TSPO-PET signal and that TSPO is expressed by diverse cell populations. While tumor core areas are the major contributor to the overall TSPO signal, TSPO signals in the tumor rim are mainly driven by CD68-positive microglia/macrophages. Molecularly, high TSPO expression marks prognostically unfavorable glioblastoma cell subpopulations characterized by an enrichment of mesenchymal gene sets and higher amounts of tumor-associated macrophages. In conclusion, our study improves the understanding of TSPO as an imaging marker in gliomas by unveiling IDH-dependent differences in TSPO expression/regulation, regional heterogeneity of the TSPO PET signal and functional implications of TSPO in terms of tumor immune cell interactions.

Keywords