4.7 Article

Mitoribosomal defects aggravate liver cancer via aberrant glycolytic flux and T cell exhaustion

期刊

出版社

BMJ PUBLISHING GROUP
DOI: 10.1136/jitc-2021-004337

关键词

liver neoplasms; lymphocytes; tumor-infiltrating; gene expression profiling; inflammation

资金

  1. Basic Science Research Program under the National Research Foundation of Korea (NRF) - Ministry of Science, ICT, and Future Planning, Korea [NRF-2021R1A2C4001829]
  2. NRF [NRF-2021R1C1C1006336]
  3. Korea Research Institute of Bioscience and Biotechnology Research Initiative Program [KGM9992211]
  4. Korea Health Technology R&D Project through the Korea Health Industry Development Institute - Ministry of Health & Welfare, Republic of Korea [HR20C0025]
  5. Korea Research Fellowship - Ministry of Science and ICT and NRF [2019H1D3A1A01071043]
  6. 'GIST Research Institute IIBR' grants - GIST [2021M3A9C4000991]
  7. Bio & Medical Technology Development Program [2021M3A9G8022959]
  8. Ministry of Science and ICT through the National Research Foundation
  9. National Research Council of Science & Technology (NST), Republic of Korea [KGM9992211] Funding Source: Korea Institute of Science & Technology Information (KISTI), National Science & Technology Information Service (NTIS)
  10. National Research Foundation of Korea [2019H1D3A1A01071043] Funding Source: Korea Institute of Science & Technology Information (KISTI), National Science & Technology Information Service (NTIS)

向作者/读者索取更多资源

Mitochondrial ribosomal protein dysfunction is associated with hepatocellular carcinoma progression and leads to an immunometabolic microenvironment favorable for cancer progression. Impaired mitoribosomal function promotes glucose partitioning toward glycolytic flux, lactate synthesis, and T cell exhaustion. This study provides insights into the critical role of mitoribosomes in regulating the immunometabolic characteristics of liver cancer.
Background Mitochondria are involved in cancer energy metabolism, although the mechanisms underlying the involvement of mitoribosomal dysfunction in hepatocellular carcinoma (HCC) remain poorly understood. Here, we investigated the effects of mitoribosomal impairment-mediated alterations on the immunometabolic characteristics of liver cancer. Methods We used a mouse model of HCC, liver tissues from patients with HCC, and datasets from The Cancer Genome Atlas (TCGA) to elucidate the relationship between mitoribosomal proteins (MRPs) and HCC. In a mouse model, we selectively disrupted expression of the mitochondrial ribosomal protein CR6-interacting factor 1 (CRIF1) in hepatocytes to determine the impact of hepatocyte-specific impairment of mitoribosomal function on liver cancer progression. The metabolism and immunophenotype of liver cancer was assessed by glucose flux assays and flow cytometry, respectively. Results Single-cell RNA-seq analysis of tumor tissue and TCGA HCC transcriptome analysis identified mitochondrial defects associated with high-MRP expression and poor survival outcomes. In the mouse model, hepatocyte-specific disruption of the mitochondrial ribosomal protein CRIF1 revealed the impact of mitoribosomal dysfunction on liver cancer progression. Crif1 deficiency promoted programmed cell death protein 1 expression by immune cells in the hepatic tumor microenvironment. A [U-C-13(6)]-glucose tracer demonstrated enhanced glucose entry into the tricarboxylic acid cycle and lactate production in mice with mitoribosomal defects during cancer progression. Mice with hepatic mitoribosomal defects also exhibited enhanced progression of liver cancer accompanied by highly exhausted tumor-infiltrating T cells. Crif1 deficiency induced an environment unfavorable to T cells, leading to exhaustion of T cells via elevation of reactive oxygen species and lactate production. Conclusions Hepatic mitoribosomal defects promote glucose partitioning toward glycolytic flux and lactate synthesis, leading to T cell exhaustion and cancer progression. Overall, the results suggest a distinct role for mitoribosomes in regulating the immunometabolic microenvironment during HCC progression.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据