Transcriptional activators YAP/TAZ and AXL orchestrate dedifferentiation, cell fate, and metastasis in human osteosarcoma

Salah-Eddine Lamhamedi-Cherradi, Sana Mohiuddin, Dhruva K Mishra, Sandhya Krishnan, Alejandra Ruiz Velasco, Amelia M Vetter, Kristi Pence, David McCall, Danh D Truong, Branko Cuglievan, Brian A Menegaz, Budi Utama, Najat C Daw, Eric R Molina, Rafal J Zielinski, John A Livingston, Richard Gorlick, Antonios G Mikos, Min P Kim, Joseph A Ludwig

Research output: Contribution to journalArticlepeer-review

Abstract

Osteosarcoma (OS) is a molecularly heterogeneous, aggressive, poorly differentiated pediatric bone cancer that frequently spreads to the lung. Relatively little is known about phenotypic and epigenetic changes that promote lung metastases. To identify key drivers of metastasis, we studied human CCH-OS-D OS cells within a previously described rat acellular lung (ACL) model that preserves the native lung architecture, extracellular matrix, and capillary network. This system identified a subset of cells-termed derived circulating tumor cells (dCTCs)-that can migrate, intravasate, and spread within a bioreactor-perfused capillary network. Remarkably, dCTCs highly expressed epithelial-to-mesenchymal transition (EMT)-associated transcription factors (EMT-TFs), such as ZEB1, TWIST, and SOX9, which suggests that they undergo cellular reprogramming toward a less differentiated state by coopting the same epigenetic machinery used by carcinomas. Since YAP/TAZ and AXL tightly regulate the fate and plasticity of normal mesenchymal cells in response to microenvironmental cues, we explored whether these proteins contributed to OS metastatic potential using an isogenic pair of human OS cell lines that differ in AXL expression. We show that AXL inhibition significantly reduced the number of MG63.2 pulmonary metastases in murine models. Collectively, we present a laboratory-based method to detect and characterize a pure population of dCTCs, which provides a unique opportunity to study how OS cell fate and differentiation contributes to metastatic potential. Though the important step of clinical validation remains, our identification of AXL, ZEB1, and TWIST upregulation raises the tantalizing prospect that EMT-TF-directed therapies might expand the arsenal of therapies used to combat advanced-stage OS.

Original languageEnglish (US)
Pages (from-to)1325-1338
Number of pages14
JournalCancer gene therapy
Volume28
Issue number12
DOIs
StatePublished - Dec 2021

Keywords

  • Animals
  • Cell Dedifferentiation
  • Disease Models, Animal
  • Humans
  • Mice
  • Neoplasm Metastasis
  • Osteosarcoma/genetics
  • Proto-Oncogene Proteins/metabolism
  • Receptor Protein-Tyrosine Kinases/metabolism
  • YAP-Signaling Proteins/metabolism
  • Axl Receptor Tyrosine Kinase

Fingerprint

Dive into the research topics of 'Transcriptional activators YAP/TAZ and AXL orchestrate dedifferentiation, cell fate, and metastasis in human osteosarcoma'. Together they form a unique fingerprint.

Cite this