American Association for Cancer Research
Browse
00085472can131597-sup-114409_4_supp_0_n7btdw.pdf (159 kB)

Data Supplement from Crosstalk between Glioma-Initiating Cells and Endothelial Cells Drives Tumor Progression

Download (159 kB)
journal contribution
posted on 2023-03-30, 22:30 authored by Hye-Min Jeon, Sung-Hak Kim, Xun Jin, Jong Bae Park, Se Hoon Kim, Kaushal Joshi, Ichiro Nakano, Hyunggee Kim

Supplementary Figure S3. ID4 regulates microRNA-dependent Jagged1 expression.

History

ARTICLE ABSTRACT

Glioma-initiating cells (GIC), which reside within the perivascular microenvironment to maintain self-renewal capacity, are responsible for glioblastoma initiation, progression, and recurrence. However, the molecular mechanisms controlling crosstalk between GICs and endothelial cells are poorly understood. Here, we report that, in both GICs and endothelial cells, platelet-derived growth factor (PDGF)–driven activation of nitric oxide (NO) synthase increases NO-dependent inhibitor of differentiation 4 (ID4) expression, which in turn promotes JAGGED1–NOTCH activity through suppression of miR129 that specifically represses JAGGED1 suppression. This signaling axis promotes tumor progression along with increased GIC self-renewal and growth of tumor vasculature in the xenograft tumors, which is dramatically suppressed by NOTCH inhibitor. ID4 levels correlate positively with NOS2 (NO synthase-2), HES1, and HEY1 and negatively with miR129 in primary GICs. Thus, targeting the PDGF–NOS–ID4–miR129 axis and NOTCH activity in the perivascular microenvironment might serve as an efficacious therapeutic modality for glioblastoma. Cancer Res; 74(16); 4482–92. ©2014 AACR.

Usage metrics

    Cancer Research

    Licence

    Exports

    RefWorks
    BibTeX
    Ref. manager
    Endnote
    DataCite
    NLM
    DC