Glioblastoma-instructed microglia transition to heterogeneous phenotypic states with phagocytic and dendritic cell-like features in patient tumors and patient-derived orthotopic xenografts

Publikation: Beitrag in FachzeitschriftForschungsartikelBeigetragen

Beitragende

  • Yahaya A Yabo - , University of Luxembourg (Autor:in)
  • Pilar M Moreno-Sanchez - , University of Luxembourg (Autor:in)
  • Yolanda Pires-Afonso - , Luxembourg Institute of Health (Autor:in)
  • Tony Kaoma - , Luxembourg Institute of Health (Autor:in)
  • Bakhtiyor Nosirov - , Luxembourg Institute of Health (Autor:in)
  • Andrea Scafidi - , Luxembourg Institute of Health (Autor:in)
  • Luca Ermini - , Luxembourg Institute of Health (Autor:in)
  • Anuja Lipsa - , Luxembourg Institute of Health (Autor:in)
  • Anaïs Oudin - , Luxembourg Institute of Health (Autor:in)
  • Dimitrios Kyriakis - , University of Luxembourg (Autor:in)
  • Kamil Grzyb - , University of Luxembourg (Autor:in)
  • Suresh K Poovathingal - , Vlaams Instituut voor Biotechnologie (Autor:in)
  • Aurélie Poli - , Luxembourg Institute of Health (Autor:in)
  • Arnaud Muller - , Luxembourg Institute of Health (Autor:in)
  • Reka Toth - , Luxembourg Institute of Health (Autor:in)
  • Barbara Klink - , Institut für Klinische Genetik, Deutsches Konsortium für Translationale Krebsforschung (Partner: DKTK, DKFZ), Nationales Centrum für Tumorerkrankungen (Partner: UKD, MFD, HZDR, DKFZ), Laboratoire National de Santé, Deutsches Krebsforschungszentrum (DKFZ) (Autor:in)
  • Guy Berchem - , Center Hospitalier de Luxembourg (Autor:in)
  • Christophe Berthold - , Center Hospitalier de Luxembourg (Autor:in)
  • Frank Hertel - , Center Hospitalier de Luxembourg (Autor:in)
  • Michel Mittelbronn - , Laboratoire National de Santé (Autor:in)
  • Dieter H Heiland - , Deutsches Konsortium für Translationale Krebsforschung (DKTK) - Freiburg (Autor:in)
  • Alexander Skupin - , University of California at San Diego (Autor:in)
  • Petr V Nazarov - , Luxembourg Institute of Health (Autor:in)
  • Simone P Niclou - , University of Luxembourg (Autor:in)
  • Alessandro Michelucci - , University of Luxembourg (Autor:in)
  • Anna Golebiewska - , Luxembourg Institute of Health (Autor:in)

Abstract

BACKGROUND: A major contributing factor to glioblastoma (GBM) development and progression is its ability to evade the immune system by creating an immune-suppressive environment, where GBM-associated myeloid cells, including resident microglia and peripheral monocyte-derived macrophages, play critical pro-tumoral roles. However, it is unclear whether recruited myeloid cells are phenotypically and functionally identical in GBM patients and whether this heterogeneity is recapitulated in patient-derived orthotopic xenografts (PDOXs). A thorough understanding of the GBM ecosystem and its recapitulation in preclinical models is currently missing, leading to inaccurate results and failures of clinical trials.

METHODS: Here, we report systematic characterization of the tumor microenvironment (TME) in GBM PDOXs and patient tumors at the single-cell and spatial levels. We applied single-cell RNA sequencing, spatial transcriptomics, multicolor flow cytometry, immunohistochemistry, and functional studies to examine the heterogeneous TME instructed by GBM cells. GBM PDOXs representing different tumor phenotypes were compared to glioma mouse GL261 syngeneic model and patient tumors.

RESULTS: We show that GBM tumor cells reciprocally interact with host cells to create a GBM patient-specific TME in PDOXs. We detected the most prominent transcriptomic adaptations in myeloid cells, with brain-resident microglia representing the main population in the cellular tumor, while peripheral-derived myeloid cells infiltrated the brain at sites of blood-brain barrier disruption. More specifically, we show that GBM-educated microglia undergo transition to diverse phenotypic states across distinct GBM landscapes and tumor niches. GBM-educated microglia subsets display phagocytic and dendritic cell-like gene expression programs. Additionally, we found novel microglial states expressing cell cycle programs, astrocytic or endothelial markers. Lastly, we show that temozolomide treatment leads to transcriptomic plasticity and altered crosstalk between GBM tumor cells and adjacent TME components.

CONCLUSIONS: Our data provide novel insights into the phenotypic adaptation of the heterogeneous TME instructed by GBM tumors. We show the key role of microglial phenotypic states in supporting GBM tumor growth and response to treatment. Our data place PDOXs as relevant models to assess the functionality of the TME and changes in the GBM ecosystem upon treatment.

Details

OriginalspracheEnglisch
Aufsatznummer51
FachzeitschriftGenome medicine
Jahrgang16
Ausgabenummer1
PublikationsstatusVeröffentlicht - 2 Apr. 2024
Peer-Review-StatusNein

Externe IDs

PubMedCentral PMC10988817
Scopus 85189167658

Schlagworte

Schlagwörter

  • Mice, Animals, Humans, Glioblastoma/genetics, Microglia/metabolism, Ecosystem, Heterografts, Brain Neoplasms/genetics, Phenotype, Disease Models, Animal, Dendritic Cells/metabolism, Tumor Microenvironment/genetics