Nat Immunol |NaCl增强CD8+ T细胞效应在肿瘤免疫治疗中的作用(Humanitas研究医院)

文摘   2024-10-02 19:43   上海  

Paper Reading

01

NaCl enhances CD8+ T cell effector functions in cancer immunotherapy

Caterina Scirgolea, et al.

Nat Immunol,2024

 After antigen recognition, CD8+ T cells undergo a series of differentiation steps, transitioning from an initial state to an effector and memory cell state, allowing them to recognize and eliminate foreign antigens for long-term persistence. However, in the tumor microenvironment (TME), these T cells typically face sustained antigen exposure and inhibitory signaling, leading to an adaptive state of low response known as "exhaustion" or "dysfunction", progressively losing effector function, decreasing proliferation, and expressing multiple inhibitory receptors such as PD-1 and CTLA-4. They demonstrated that high-salt treatment promotes the differentiation of human CD8+ naïve T cells into effector memory T cells, enhances IFN-γproduction, degranulation, and cytotoxicity, while maintaining a gene network associated with stem cell-like plasticity, which is dependent on SGK1. In mice, a high-salt diet (HSD) inhibited the growth of MC38 tumors, which depended on CD8+ T cells, and HSD increased the infiltration of CD8+ T cells and natural killer cells in tumors, altered the transcriptome of CD8+ T cells, and adjusted the activity of related transcription factors and signaling pathways, showing similar characteristics to anti-PD-1 immunotherapy. In addition, HSD-induced CD8+ T cells were similar to cells following tumor antigen recognition and CD8+ T cells in immunotherapy responders, with characteristic genes associated with anti-PD-1 therapy response, and elevated plasma sodium levels were associated with longer progression-free survival in cancer patients. Transcriptome analysis and metabolomic analysis showed that NaCl treatment resulted in altered expression of metabolic genes and nutrient transporters in CD8+ T cells, in which glutamine metabolism plays a key role, and that NaCl enhanced glutamine depletion and promoted glutamine-dependent epigenetic remodeling, including increased 5-hydroxymethylcytosine levels, decreased H3K27me3 levels, and altered chromatin accessibility. In vitro short-term treatment of CD8+ T cells with high concentrations of NaCl can lead to enhanced effector function and tumor control after adoptive metastasis, as evidenced by transient upregulation of inhibitory receptors, re-expression of stem cell-like memory markers, sustained secretion of large amounts of IFN-γ, and better control of tumor growth.

DOI: 10.1038/s41590-024-01923-9


02

PGE2 inhibits TIL expansion by disrupting IL-2 signalling and mitochondrial function


Matteo Morotti, et al.
Nature,2024


    Expansion of antigen-experienced CD8+ T cells is critical for the success of tumor-infiltrating lymphocyte (TIL)-adoptive cell therapy (ACT) in patients with cancer1. Interleukin-2 (IL-2) acts as a key regulator of CD8+ cytotoxic T lymphocyte functions by promoting expansion and cytotoxic capability. They demonstrated that PGE2 produced by cyclooxygenase (COX) activity in tumor cells does not impair the priming of naïve CD8+ T cells in tumor-draining lymph nodes. In contrast, PGE2 exerts a local role in the tumor microenvironment, selectively targeting TCF1+CD8+ TIL and restricting its expansion and differentiation into effector T cells. This inhibition is mediated by signaling of the PGE2 receptors EP2 and EP4 (EP2/EP4) expressed on CD8+ T cells. Using single-cell RNA sequencing and TCR sequencing, the authors provide a comprehensive overview of the CD8+ TIL population and its developmental trajectory within tumors. They revealed that TCF1+CD8+ TIL could not be clonally amplified and differentiated into effector populations expressing TIM-3 and CXCR6 in the presence of PGE2. Notably, gene knockout of EP2/EP4 signaling in CD8+ T cells rescued this defect, leading to robust amplification and effector differentiation of TCF1+ TIL, ultimately leading to tumor elimination in multiple mouse cancer models. Mechanistically, the authors found that PGE2EP2/EP4 signaling inhibits TCF1+ TIL responsiveness to IL-2, a key cytokine for T cell expansion and effector differentiation. PGE2 exposure impairs IL-2 receptor expression and downstream signaling pathways, including STAT5 phosphorylation and mTORC1 activation, in TCF1+CD8+ T cells. Restoration of IL-2 responsiveness by inhibition of EP2/EP4 signaling is sufficient to rescue the expansion and effector differentiation of TCF1+ TIL within tumors without compromising its stem-cell-like properties. The study further demonstrated that EP2/EP4-deficient tumor antigen-specific CD8+ T cells undergo clonal expansion and differentiation within the tumor, resulting in the generation of potent effector populations that mediate tumor elimination. This protective anticancer response is abrogated upon blockade of IL-2 receptor signaling, highlighting the critical role of IL-2 reactivity in driving the amplification and effector differentiation of TCF1+ TIL. Overall, this work reveals a previously unrecognized mechanism by which tumor-derived PGE2 limits the anticancer potential of TCF1+CD8+ TIL by inhibiting IL-2 responsiveness. By identifying the PGE2EP2/EP4 axis as a molecular target, this study provides a theoretical basis for improving T-cell immunotherapy in patients with tumors with high PGE2 expression.

DOI: 10.1038/s41586-024-07352-w







相关阅读:
1. Nat Immunol | PKM2缺乏激活磷酸戊糖途径并产生 TCF1+ CD8前体细胞从而改善免疫疗法 (威尔康奈尔医学院)

2. Nature | 单细胞CAR-T图谱揭示8年白血病缓解中的Type-2功能(耶鲁大学)

END


关注我们获取更多免疫学文献资讯

Editor & Reviewer: Shiyang Song


李斌课题组
文献阅读共享
 最新文章