TET 2 promotes anti‐tumor immunity by governing G‐ MDSC s and CD 8 + T‐cell numbers

Shuangqi Li,Jiuxing Feng,Feizhen Wu,Jiabin Cai,Xinyu Zhang,Haikun Wang,Irfete S Fetahu,Isabella Iwanicki,Dingailu Ma,Tao Hu,Hang Liu,Bingjie Wang,Guoming Shi,Li Tan,Yujiang Geno Shi
DOI: https://doi.org/10.15252/embr.201949425
IF: 9.071
2020-09-14
EMBO Reports
Abstract:<p>The host immune response is a fundamental mechanism for attenuating cancer progression. Here we report a role for the DNA demethylase and tumor suppressor TET2 in host anti‐tumor immunity. Deletion of Tet2 in mice elevates IL‐6 levels upon tumor challenge. Elevated IL‐6 stimulates immunosuppressive granulocytic myeloid‐derived suppressor cells (G‐MDSCs), which in turn reduce CD8<sup>+</sup> T cells upon tumor challenge. Consequently, systematic knockout of Tet2 in mice leads to accelerated syngeneic tumor growth, which is constrained by anti‐PD‐1 blockade. Removal of G‐MDSCs by the anti‐mouse Ly6g antibodies restores CD8<sup>+</sup> T‐cell numbers in Tet2<sup>−/−</sup> mice and reboots their anti‐tumor activity. Importantly, anti‐IL‐6 antibody treatment blocks the expansion of G‐MDSCs and inhibits syngeneic tumor growth. Collectively, these findings reveal a TET2‐mediated IL‐6/G‐MDSCs/CD8<sup>+</sup> T‐cell immune response cascade that safeguards host adaptive anti‐tumor immunity, offering a cell non‐autonomous mechanism of TET2 for tumor suppression.</p>
cell biology,biochemistry & molecular biology
What problem does this paper attempt to address?
The paper attempts to address the role of TET2 in the host's anti-tumor immune response, particularly its non-cell-autonomous effects (i.e., influencing other cell types rather than directly acting on tumor cells) on tumor growth. Specifically, the researchers focus on how TET2 affects tumor growth by regulating the number of immunosuppressive granulocytic myeloid-derived suppressor cells (G-MDSCs) and CD8+ T cells. ### Main Issues: 1. **Role of TET2 in anti-tumor immunity**: Researchers aim to understand how TET2 participates in the host's anti-tumor immune response. 2. **Impact of TET2 deficiency on tumor growth**: By knocking out the Tet2 gene in mice, they observe its effects on tumor growth rate and size. 3. **Immune changes caused by TET2 deficiency**: Investigate how TET2 deficiency affects the composition and function of immune cells, particularly the number and activity of G-MDSCs and CD8+ T cells. 4. **Role of IL-6 in TET2 deficiency**: Study whether elevated IL-6 levels are a key mechanism by which TET2 deficiency accelerates tumor growth. ### Research Methods: - **Animal Model**: Use C57BL/6 mice and establish a Tet2-deficient mouse model by knocking out the Tet2 gene. - **Tumor Model**: Inject Hepa1-6 liver cancer cells or Py8119 breast cancer cells into mice and observe tumor growth. - **Immune Analysis**: Detect the composition and function of immune cells in the tumor microenvironment using flow cytometry, immunohistochemistry, and other methods. - **Molecular Mechanism Study**: Analyze changes in gene expression and IL-6 levels after TET2 deficiency using mRNA-seq, ELISA, and other techniques. ### Main Findings: - **TET2 deficiency promotes tumor growth**: Tumor growth is significantly accelerated, and survival rate is reduced in Tet2 knockout mice. - **Immune cell changes**: In Tet2 knockout mice, the number of CD8+ T cells decreases, while the number of G-MDSCs increases. - **Role of IL-6**: IL-6 levels are significantly elevated in Tet2 knockout mice, which may cause excessive proliferation of G-MDSCs and reduction of CD8+ T cells. - **Effect of anti-PD-1 therapy**: Anti-PD-1 antibody treatment can partially restore the anti-tumor immune response in Tet2 knockout mice and inhibit tumor growth. ### Conclusion: TET2 affects the host's anti-tumor immune response by regulating IL-6 levels and the number of G-MDSCs. TET2 deficiency leads to excessive proliferation of G-MDSCs, which in turn inhibits the function of CD8+ T cells, thereby accelerating tumor growth. This finding reveals the non-cell-autonomous mechanism of TET2 in anti-tumor immunity and provides a theoretical basis for developing new anti-tumor strategies.