• Media type: E-Article
  • Title: Extracellular vesicle-associated tyrosine kinase-like orphan receptors ROR1 and ROR2 promote breast cancer progression
  • Contributor: Irmer, Barnabas; Efing, Janes; Reitnauer, Lea Elisabeth; Angenendt, Allegra; Heinrichs, Saskia; Schubert, Antonia; Schulz, Matthias; Binder, Claudia; Tio, Joke; Hansen, Uwe; Geyer, Christiane; Gerwing, Mirjam; Bleckmann, Annalen; Menck, Kerstin
  • imprint: Springer Science and Business Media LLC, 2023
  • Published in: Cell Communication and Signaling, 21 (2023) 1
  • Language: English
  • DOI: 10.1186/s12964-023-01186-1
  • ISSN: 1478-811X
  • Keywords: Cell Biology ; Molecular Biology ; Biochemistry
  • Origination:
  • Footnote:
  • Description: <jats:title>Abstract</jats:title><jats:sec> <jats:title>Background</jats:title> <jats:p>Extracellular vesicles (EVs) harbor a plethora of different biomolecules, which they can transport across cells. In cancer, tumor-derived EVs thereby support the creation of a favorable tumor microenvironment. So far, EV uptake and cargo delivery into target cells have been regarded as the main mechanisms for the pro-tumoral function of EVs. To test this hypothesis, we investigated the fate of the oncogenic transmembrane Wnt tyrosine kinase-like orphan receptor 1 and 2 (ROR1, ROR2) delivered via distinct EV subpopulations to breast cancer cells and aimed to unravel their impact on tumor progression.</jats:p> </jats:sec><jats:sec> <jats:title>Methods</jats:title> <jats:p>EVs were isolated by differential ultracentrifugation from cell culture supernatant as well as plasma samples from healthy individuals (<jats:italic>n</jats:italic> = 27) and breast cancer patients (<jats:italic>n</jats:italic> = 41). EVs were thoroughly characterized by electron microscopy, nanoparticle tracking analysis, immunoblot, and flow cytometry. ROR transfer to target cells was observed using microscopy-based assays and biodistribution experiments were conducted in syngeneic mice. EV impact on cancer cell migration and invasion was tested in functional assays.</jats:p> </jats:sec><jats:sec> <jats:title>Results</jats:title> <jats:p>We observed that the supernatant of ROR-overexpressing cells was sufficient for transferring the receptors to ROR-negative cells. Analyzing the secretome of the ROR-overexpressing cells, we detected a high enrichment of ROR1/2 on large and small EVs, but not on large oncosomes. Interestingly, the majority of ROR-positive EVs remained attached to the target cell surface after 24 h of stimulation and was quickly removed by treatment with trypsin. Nonetheless, ROR-positive EVs increased migration and invasion of breast cancer cells, even after chemically inhibiting EV uptake, in dependence of RhoA downstream signaling. In vivo, ROR-depleted EVs tended to distribute less into organs prone for the formation of breast cancer metastases. ROR-positive EVs were also significantly elevated in the plasma of breast cancer patients and allowed to separate them from healthy controls.</jats:p> </jats:sec><jats:sec> <jats:title>Conclusions</jats:title> <jats:p>The oncogenic Wnt receptors ROR1/2 are transferred via EVs to the surface of ROR-negative cancer cells, in which they induce an aggressive phenotype supporting tumor progression.</jats:p> </jats:sec>
  • Access State: Open Access