1044 Proteasome mediated protein catabolism fuels antitumor immunity

Regular and Young Investigator Award Abstracts(2022)

引用 0|浏览1
暂无评分
摘要
BackgroundThe solid tumor microenvironment (TME) exposes CD8 T cells to metabolic stressors including nutrient and oxygen deprivation coupled with persistent antigen stimulation which work in concert to inhibit cell function.1-5 Importantly, successful immunotherapy is predicated on CD8 T cells overcoming these hurdles to maintain proliferation and protein secretion. While recent advances in single cell RNA and ATAC sequencing have allowed for identification of genetic and epigenetic traits associated with enhanced antitumor T cell function 6-7, little is known about the mechanisms responsible for controlling the translation of these instructions into effector functions. In this study, we sought to identify the mechanisms responsible for allowing sustained protein translation in the TME.MethodsProtein synthesis was monitored using a flow cytometry-based approach whereby the fluorescent analogue of methionine, L-homopropargylglycine (L-HPG), is incorporated into new forming polypeptide chains and quantified by flow cytometry through Click-IT chemistry.8-11 Optimal antitumor T cells were generated via cytokine conditioning with IL-15 and then subjected to a series of in vitro tumor-T cell coculture systems and in vivo tumor growth experiments. Proteasome activity was monitored using a fluorescent activity probe via flow cytometry and pharmacologic intervention using the proteasome inhibitor MG-132 paired with metabolomics. Proteasome stimulator Cyclosporine A was used to validate our findings.ResultsUsing both human and mouse tumors we found that protein translation is repressed in T cells by the solid TME through activation of the unfolded protein response element p-eIF2a due to competition for glucose. Reprogramming T cells away from glucose dependency alleviated p-eIF2a mediated translation attenuation allowing for sustained translation under TME stress. Using metabolic and pharmacological approaches, we discovered that optimal antitumor T cells mitigate p-eIF2a through enhanced proteasome activity, protecting from translation attenuation enabling sustained cytokine synthesis in solid tumors that resulted in enriched tumor control. Additionally, we found the ability to access protein degradation via the proteasome complex was associated with metabolic programs previously linked to optimal antitumor immunity such as glutathione metabolism and gluconeogenesis.12ConclusionsThese findings suggest that stress mediated attenuation of translation represents a cellular checkpoint which must be overcome for optimal tumor immunity. Our findings demonstrate protein degradation is a critical component of T cell tumor control and strategies that relieve the misfolded protein burden could be avenues to supplement current immunotherapy approaches.ReferencesWherry EJ. T cell exhaustion. Nat Immunol 2011;12(6):492–9. Bian Y, Li W, Kremer DM, et al. Cancer SLC43A2 alters T cell methionine metabolism and histone methylation. Nature 2020;585(7824):277–82. Chang CH, Qiu J, O’Sullivan D, et al. Metabolic Competition in the Tumor Microenvironment Is a Driver of Cancer Progression. Cell 2015;162(6):1229–41. Thevenot PT, Sierra RA, Raber PL, et al. The stress-response sensor chop regulates the function and accumulation of myeloid-derived suppressor cells in tumors. Immunity 2014;41(3):389–401. Brand A, Singer K, Koehl GE, et al. LDHA-Associated Lactic Acid Production Blunts Tumor Immunosurveillance by T and NK Cells. Cell Metab 2016;24(5):657–71. Miller BC, Sen DR, Al Abosy R, et al. Subsets of exhausted CD8(+) T cells differentially mediate tumor control and respond to checkpoint blockade. Nat Immunol 2019;20(3):326–36. Shin HM, Kim G, Kim S, et al. Chromatin accessibility of circulating CD8+ T cells predicts treatment response to PD-1 blockade in patients with gastric cancer. Nature Communications 2021;12(1):975. Araki K, Morita M, Bederman AG, et al. Translation is actively regulated during the differentiation of CD8(+) effector T cells. Nat Immunol 2017;18(9):1046–57. Graczyk D, White RJ, Ryan KM. Involvement of RNA Polymerase III in Immune Responses. Mol Cell Biol 2015;35(10):1848–59. Lai CP, Kim EY, Badr CE, et al. Visualization and tracking of tumour extracellular vesicle delivery and RNA translation using multiplexed reporters. Nat Commun 2015;6:7029. Best MD. Click chemistry and bioorthogonal reactions: unprecedented selectivity in the labeling of biological molecules. Biochemistry 2009;48(28):6571–84. Ma R, Ji T, Zhang H, et al. A Pck1-directed glycogen metabolic program regulates formation and maintenance of memory CD8(+) T cells. Nat Cell Biol 2018;20(1):21–27.Ethics ApprovalPatients undergoing surgical removal of tumors granted consent via MUSC Biorepository surgical consent forms. This work was determined by MUSC Institutional Review Board to be exempt under protocol Pro00050181. Tissue samples were de-identified. Studies were conducted in accordance with the Declaration of Helsinki, International Ethical Guidelines for Biomedical Research Involving Human Subjects (CIOMS), Belmont Report, or U.S. Common Rule. All animal experiments were approved by both the Medical University of South Carolina (MUSC) Institutional Animal Care and Use Committee and the University of North Carolina at Chapel hill (UNC) Division of Comparative Medicine. Mice were maintained by the Division of Laboratory Animal Resources at MUSC and Division of Comparative Medicine at UNC.
更多
查看译文
关键词
proteasome,protein catabolism fuels,immunity
AI 理解论文
溯源树
样例
生成溯源树,研究论文发展脉络
Chat Paper
正在生成论文摘要