Journal for ImmunoTherapy of Cancer (Jun 2023)

G-MDSC-derived exosomes mediate the differentiation of M-MDSC into M2 macrophages promoting colitis-to-cancer transition

  • Zhe Zhang,
  • Yungang Wang,
  • Yanxia Ding,
  • Shengjun Wang,
  • Hongli Liu,
  • Dezhi Bian,
  • Keke Shao

DOI
https://doi.org/10.1136/jitc-2022-006166
Journal volume & issue
Vol. 11, no. 6

Abstract

Read online

Backgrounds In inflammatory bowel disease microenvironment, transdifferentiation of myeloid-derived suppressor cells (MDSCs) and M2 macrophage accumulation are crucial for the transition of colitis-to-cancer. New insights into the cross-talk and the underling mechanism between MDSCs and M2 macrophage during colitis-to-cancer transition are opening new avenues for colitis-associated cancer (CAC) prevention and treatment.Methods The role and underlying mechanism that granulocytic MDSCs (G-MDSCs) or exosomes (Exo) regulates the differentiation of monocytic MDSCs (M-MDSCs) into M2 macrophages were investigated using immunofluorescence, FACS, IB analysis, etc, and employing siRNA and antibodies. In vivo efficacy and mechanistic studies were conducted with dextran sulfate sodium-induced CAC mice, employed IL-6 Abs and STAT3 inhibitor.Results G-MDSCs promote the differentiation of M-MDSC into M2 macrophages through exosomal miR-93–5 p which downregulating STAT3 activity in M-MDSC. IL-6 is responsible for miR-93–5 p enrichment in G-MDSC exosomes (GM-Exo). Mechanistically, chronic inflammation-driven IL-6 promote the synthesis of miR-93–5 p in G-MDSC via IL-6R/JAK/STAT3 pathway. Early use of IL-6 Abs enhances the effect of STAT3 inhibitor against CAC.Conclusions IL-6-driven secretion of G-MDSC exosomal miR-93–5 p promotes the differentiation of M-MDSC into M2 macrophages and involves a STAT3 signaling mechanism that promote colitis-to-cancer transition. Combining STAT3 inhibitors with strategies that inhibit IL-6-mediated G-MDSC exosomal miR-93–5 p production is beneficial for the prevention and treatment of CAC.