1. Academic Validation
  2. SHMT2 regulates esophageal cancer cell progression and immune Escape by mediating m6A modification of c-myc

SHMT2 regulates esophageal cancer cell progression and immune Escape by mediating m6A modification of c-myc

  • Cell Biosci. 2023 Nov 6;13(1):203. doi: 10.1186/s13578-023-01148-7.
Zhe Qiao 1 Yu Li 1 Yao Cheng 1 Shaomin Li 1 Shiyuan Liu 2
Affiliations

Affiliations

  • 1 Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West 5th Road, 710004, Xi'an, Shaanxi, China.
  • 2 Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West 5th Road, 710004, Xi'an, Shaanxi, China. liushiyuan@xjtu.edu.cn.
Abstract

Background: In recent years, the role of altered cellular metabolism in tumor progression has attracted widespread attention. Related metabolic enzymes have also been considered as potential Cancer therapeutic targets. Serine hydroxymethyltransferase 2 (SHMT2) has been reported to be upregulated in several cancers and associated with poor prognosis. However, there are few studies of SHMT2 in esophageal Cancer (EC), and the related functions and mechanisms also need to be further explored.

Methods: In this study, we first analyzed SHMT2 expression in EC by online database and clinical samples. Then, the biological functions of SHMT2 in EC were investigated by cell and animal experiments. The intracellular m6A methylation modification levels were also evaluated by MeRIP. Linked genes and mechanisms of SHMT2 were analyzed by bioinformatics and rescue experiments.

Results: We found that SHMT2 expression was abnormally upregulated in EC and associated with poor prognosis. Functionally, SHMT2 silencing suppressed c-Myc expression in an m6A-dependent manner, thereby blocking the proliferation, migration, invasion and immune escape abilities of EC cells. Mechanistically, SHMT2 encouraged the accumulation of methyl donor SAM through a one-carbon metabolic network, thereby regulating the m6A modification and stability of c-Myc mRNA in a METTL3/FTO/ALKBH5/IGF2BP2-dependent way. In vivo animal experiments also demonstrated that SHMT2 mediated MYC expression by m6A-methylation modification, thus boosting EC tumorigenesis.

Conclusion: In conclusion, our data illustrated that SHMT2 regulated malignant progression and immune escape of EC cell through c-Myc m6A modification. These revealed mechanisms related to SHMT2 in EC and maybe offer promise for the development of new therapeutic approaches.

Keywords

EC; One carbon metabolism; SHMT2; c-myc; m6A modification.

Figures
Products