4.6 Article

Meclofenamate causes loss of cellular tethering and decoupling of functional networks in glioblastoma

Journal

NEURO-ONCOLOGY
Volume 23, Issue 11, Pages 1885-1897

Publisher

OXFORD UNIV PRESS INC
DOI: 10.1093/neuonc/noab092

Keywords

glioblastoma; intercellular network architecture; meclofenamate; tumor microtubes

Funding

  1. Bonfor
  2. Familie Mehdorn Stiftung
  3. Mildred-Scheel School of Oncology (MSSO) Cologne-Bonn - German Cancer Aid [70113307]
  4. Else Kroner-Fresenius-Foundation
  5. BMBF (Bundesministerium fur Bildung und Forschung) [031L0260B]
  6. The Norwegian Cancer Society [190170]

Ask authors/readers for more resources

This study explored the relationship between network architecture and transcriptional profile in glioblastoma cells, finding that meclofenamate (MFA) can effectively disrupt syncytial network structures. MFA treatment led to reduced intercellular cytosolic traffic and breakdown of long membrane protrusions, ultimately affecting the communication networks and response to chemotherapy in glioblastoma cells. These findings suggest a potential therapeutic approach targeting tumor microtubes (TMs) with MFA in clinical settings.
Background. Glioblastoma cells assemble to a syncytial communicating network based on tumor microtubes (TMs) as ultra-long membrane protrusions. The relationship between network architecture and transcriptional profile remains poorly investigated. Drugs that interfere with this syncytial connectivity such as meclofenamate (MFA) may be highly attractive for glioblastoma therapy. Methods. In a human neocortical slice model using glioblastoma cell populations of different transcriptional signatures, three-dimensional tumor networks were reconstructed, and TM-based intercellular connectivity was mapped on the basis of two-photon imaging data. MFA was used to modulate morphological and functional connectivity; downstream effects of MFA treatment were investigated by RNA sequencing and fluorescence-activated cell sorting (FACS) analysis. Results. TM-based network morphology strongly differed between the transcriptional cellular subtypes of glioblastoma and was dependent on axon guidance molecule expression. MFA revealed both a functional and morphological demolishment of glioblastoma network architectures which was reflected by a reduction of TM-mediated intercellular cytosolic traffic as well as a breakdown of TM length. RNA sequencing confirmed a downregulation of NCAM and axon guidance molecule signaling upon MFA treatment. Loss of glioblastoma communicating networks was accompanied by a failure in the upregulation of genes that are required for DNA repair in response to temozolomide (TMZ) treatment and culminated in profound treatment response to TMZ-mediated toxicity. Conclusion. The capacity of TM formation reflects transcriptional cellular heterogeneity. MFA effectively demolishes functional and morphological TM-based syncytial network architectures. These findings might pave the way to a clinical implementation of MFA as a TM-targeted therapeutic approach.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available