Abstract

MicroRNA-155 (miR-155) regulates antitumor immune responses. However, its specific functions within distinct immune cell types have not been delineated in conditional KO mouse models. In this study, we investigated the role of miR-155 specifically within T cells during the immune response to syngeneic tumors. We found that miR-155 expression within T cells is required to limit syngeneic tumor growth and promote IFNγ production by T cells within the tumor microenvironment. Consequently, we found that miR-155 expression by T cells is necessary for proper tumor-associated macrophage expression of IFNγ-inducible genes. We also found that immune checkpoint-blocking (ICB) antibodies against programmed cell death protein 1/programmed death ligand 1 (PD-1/PD-L1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4) restored antitumor immunity in miR-155 T cell-conditional KO mice. We noted that these ICB antibodies rescued the levels of IFNγ-expressing T cells, expression of multiple activation and effector genes expressed by tumor-infiltrating CD8+ and CD4+ T cells, and tumor-associated macrophage activation. Moreover, the ICB approach partially restored expression of several derepressed miR-155 targets in tumor-infiltrating, miR-155-deficient CD8+ T cells, suggesting that miR-155 and ICB regulate overlapping pathways to promote antitumor immunity. Taken together, our findings highlight the multifaceted role of miR-155 in T cells, in which it promotes antitumor immunity. These results suggest that the augmentation of miR-155 expression could be used to improve anticancer immunotherapies.

Highlights

  • We found that immune checkpoint– blocking (ICB) antibodies against programmed cell death protein 1/programmed death ligand 1 (PD-1/PD-L1) and cytotoxic T lymphocyte–associated protein 4 (CTLA-4) restored antitumor immunity in miR-155 T cell– conditional KO mice

  • We noted a trending decrease in the percentage of CD8ϩ T cells and a significant decrease in CD8ϩIFN␥ϩ T cells (Fig. 1, E and F) within the tumor microenvironment when miR-155 was lacking from T cells

  • This study found that miR-155 plays an expanded role in modulating the expression of effector molecules such as granzyme B, perforin, and Tnf in CD8ϩ T cells within the tumor microenvironment and that it regulates the expression of several activation genes in both CD8ϩ and CD4ϩ T cells in this environment

Read more

Summary

Results

T cell–specific deletion of miR-155 reduces the levels of intratumor IFN␥-expressing T cells and promotes the growth of B16f10 tumors. Defective CD40 and MHCI expression on TAMs from miR-155 TCKO mice returned to WT levels following ICB (Fig. 3E) These data indicate that ICB can rescue miR-155 tumor immunity phenotypes, where it reduces tumor growth, restores the levels of IFN␥-producing T cells, and promotes the activation of TAMs in miR-155 TCKO mice. This reveals that defective T cell antitumor immunity in the absence of T cell– expressed miR-155 is a defect that can be reversed. This indicates that miR-155 and ICB regulate overlapping pathways as they promote antitumor immunity mediated by T cells

Discussion
Experimental procedures
Cell culture and tumor experiments
Intracellular staining and flow cytometry
RNA isolation and qPCR
ICB experiments
Luciferase assay
Statistical analysis
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call