Counterintuitive production of tumor-suppressive secretomes from Oct4- and c-Myc-overexpressing tumor cells and MSCs

dc.contributor.authorLi, Kexin
dc.contributor.authorSun, Xun
dc.contributor.authorZha, Rongrong
dc.contributor.authorLiu, Shengzhi
dc.contributor.authorFeng, Yan
dc.contributor.authorSano, Tomonori
dc.contributor.authorAryal, Uma K.
dc.contributor.authorSudo, Akihiro
dc.contributor.authorLi, Bai-Yan
dc.contributor.authorYokota, Hiroki
dc.contributor.departmentAnatomy, Cell Biology and Physiology, School of Medicine
dc.date.accessioned2023-06-14T15:40:02Z
dc.date.available2023-06-14T15:40:02Z
dc.date.issued2022-03-28
dc.description.abstractBackground: Advanced breast cancer frequently metastasizes to bone, but inhibiting tumor progression in chemotherapy may occasionally enhance tumorigenesis. Here, we employed a counterintuitive approach of overexpressing Yamanaka factors (Oct4, c-Myc, Sox2, and Klf4) and examined a conditioned medium (CM)-based treatment option with induced tumor-suppressing cells (iTSCs). Methods:In vitro proliferation and migration assays were conducted using tumor cell lines derived from breast cancer, as well as prostate and pancreatic cancers, and osteosarcoma. The tumor-suppressing capability of iTSC-derived CM was evaluated using freshly isolated breast cancer tissues and a mouse model of mammary tumors and tumor-induced osteolysis. The regulatory mechanism was evaluated using Western blotting, immunoprecipitation, pull-down, gene overexpression, and RNA interference based on mass spectrometry-based proteomics data. Results: The overexpression of Oct4 and c-Myc in tumor cells and MSCs, but not Sox2 or Klf4, generated anti-tumor CM, which suppressed the progression of mammary tumors and tumor-induced bone loss. Notably, CM downregulated histone demethylase, and PDL-1, a blocker of T-cell-based immune responses. Whole-genome proteomics predicted enolase 1 (Eno1), Hsp90ab1, Eef2, and vinculin as extracellular tumor suppressors. Specifically, CD44 was co-immunoprecipitated with Eno1 and the silencing of CD44 suppressed Eno1's anti-tumor action. The overexpression of Oct4 and c-Myc also generated secretomes that inhibited the development of bone-resorbing osteoclasts. Conclusions: In analogous to cell competition in which Myc-overexpressing cells in Drosophila and mouse embryos remove neighboring cells with a lower level of Myc, this study presented the possibility of eliminating tumor cells by the secretory proteomes derived from Myc/Oc4-overexpressing iTSCs.en_US
dc.eprint.versionFinal published versionen_US
dc.identifier.citationLi K, Sun X, Zha R, et al. Counterintuitive production of tumor-suppressive secretomes from Oct4- and c-Myc-overexpressing tumor cells and MSCs. Theranostics. 2022;12(7):3084-3103. Published 2022 Mar 28. doi:10.7150/thno.70549en_US
dc.identifier.urihttps://hdl.handle.net/1805/33757
dc.language.isoen_USen_US
dc.publisherIvyspring Internationalen_US
dc.relation.isversionof10.7150/thno.70549en_US
dc.relation.journalTheranosticsen_US
dc.rightsAttribution 4.0 International*
dc.rights.urihttp://creativecommons.org/licenses/by/4.0/*
dc.sourcePMCen_US
dc.subjectBreast canceren_US
dc.subjectCell competitionen_US
dc.subjectTumorigenesisen_US
dc.subjectInduced tumor-suppressing cellsen_US
dc.titleCounterintuitive production of tumor-suppressive secretomes from Oct4- and c-Myc-overexpressing tumor cells and MSCsen_US
dc.typeArticleen_US
Files
Original bundle
Now showing 1 - 1 of 1
Loading...
Thumbnail Image
Name:
thnov12p3084.pdf
Size:
3.29 MB
Format:
Adobe Portable Document Format
Description:
License bundle
Now showing 1 - 1 of 1
No Thumbnail Available
Name:
license.txt
Size:
1.99 KB
Format:
Item-specific license agreed upon to submission
Description: