4.7 Article

Tumorous expression of NAC1 restrains antitumor immunity through the LDHA-mediated immune evasion

期刊

出版社

BMJ PUBLISHING GROUP
DOI: 10.1136/jitc-2022-004856

关键词

-

资金

  1. National Institutes of Health [R01CA221867, R01AI121180, R21AI167793]
  2. Department of Defense [LC210150]

向作者/读者索取更多资源

This study investigates the role of nucleus accumbens-associated protein-1 (NAC1) in the tumor microenvironment and its impact on anti-tumor immunity. The results demonstrate that NAC1 negatively affects immune evasion by regulating lactate dehydrogenase A (LDHA) expression and lactic acid production. Targeting NAC1 therapeutically may be a potential strategy to enhance cancer immunotherapy.
Background T cell-mediated antitumor immunity has a vital role in cancer prevention and treatment; however, the immune-suppressive tumor microenvironment (TME) constitutes a significant contributor to immune evasion that weakens antitumor immunity. Here, we explore the relationship between nucleus accumbens-associated protein-1 (NAC1), a nuclear factor of the BTB (broadcomplex, Tramtrack, brit a brac)/POZ (Poxvirus, and Zinc finger) gene family, and the TME. Methods Adoptive cell transfer (ACT) of mouse or human tumor antigen (Ag)-specific CDR' cytotoxic T lymphocytes (CTLs) was tested in an immunocompetent or immunodeficient mouse model of melanoma with or without expression of NAC1. The effects of NAC1 expression on immune evasion in tumor cells were assessed in vitro and in vivo. CRISPR/Cas9, glycolysis analysis, retroviral transduction, quantitative real-time PCR, flow cytometric analysis, immunoblotting, database analyses were used to screen the downstream target and underlying mechanism of NAC1 in tumor cells. Results Tumorous expression of NAC1 negatively impacts the CTL-mediated antitumor immunity via lactate dehydrogenase A (LDHA)-mediated suppressive TME. NAC1 positively regulated the expression of LDHA at the transcriptional level, which led to higher accumulation of lactic acid in the TME. This inhibited the cytokine production and induced exhaustion and apoptosis of CTLs, impairing their cell-killing ability. In the immunocompetent and immunodeficient mice, NAC1 depleted melanoma tumors grew significantly slower and had an elevated infiltration of tumor Ag-specific CTLs following ACT, compared with the control groups. Conclusions Tumor expression of NAC1 contributes substantially to immune evasion through its regulatory role in LDHA expression and lactic acid production. Thus, therapeutic targeting of NAC1 warrants further exploration as a potential strategy to reinforce cancer immunotherapy, such as the ACT of CTLs.

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