Hypoxia-induced TMTC3 expression in esophageal squamous cell carcinoma potentiates tumor angiogenesis through Rho GTPase/STAT3/VEGFA pathway

J Exp Clin Cancer Res. 2023 Sep 26;42(1):249. doi: 10.1186/s13046-023-02821-y.

Abstract

Background: Hypoxia is one of most typical features in the tumor microenvironment of solid tumor and an inducer of endoplasmic reticulum (ER) stress, and HIF-1α functions as a key transcription factor regulator to promote tumor angiogenesis in the adaptive response to hypoxia. Increasing evidence has suggested that hypoxia plays an important regulatory role of ER homeostasis. We previously identified TMTC3 as an ER stress mediator under nutrient-deficiency condition in esophageal squamous cell carcinoma (ESCC), but the molecular mechanism in hypoxia is still unclear.

Methods: RNA sequencing data of TMTC3 knockdown cells and TCGA database were analyzed to determine the association of TMTC3 and hypoxia. Moreover, ChIP assay and dual-luciferase reporter assay were performed to detect the interaction of HIF-1α and TMTC3 promoter. In vitro and in vivo assays were used to investigate the function of TMTC3 in tumor angiogenesis. The molecular mechanism was determined using co-immunoprecipitation assays, immunofluorescence assays and western blot. The TMTC3 inhibitor was identified by high-throughput screening of FDA-approved drugs. The combination of TMTC3 inhibitor and cisplatin was conducted to confirm the efficiency in vitro and in vivo.

Results: The expression of TMTC3 was remarkably increased under hypoxia and regulated by HIF-1α. Knockdown of TMTC3 inhibited the capability of tumor angiogenesis and ROS production in ESCC. Mechanistically, TMTC3 promoted the production of GTP through interacting with IMPDH2 Bateman domain. The activity of Rho GTPase/STAT3, regulated by cellular GTP levels, decreased in TMTC3 knockdown cells, whereas reversed by IMPDH2 overexpression. Additionally, TMTC3 regulated the expression of VEGFA through Rho GTPase/STAT3 pathway. Allopurinol inhibited the expression of TMTC3 and further reduced the phosphorylation and activation of STAT3 signaling pathway in a dose-dependent manner in ESCC. Additionally, the combination of allopurinol and cisplatin significantly inhibited the cell viability in vitro and tumor growth in vivo, comparing with single drug treatment, respectively.

Conclusions: Collectively, our study clarified the molecular mechanism of TMTC3 in regulating tumor angiogenesis and highlighted the potential therapeutic combination of TMTC3 inhibitor and cisplatin, which proposed a promising strategy for the treatment of ESCC.

Keywords: Hypoxia; IMPDH2; Rho GTPase/STAT3; TMTC3; Tumor angiogenesis.

MeSH terms

  • Allopurinol
  • Carrier Proteins
  • Cisplatin / pharmacology
  • Esophageal Neoplasms* / genetics
  • Esophageal Squamous Cell Carcinoma* / genetics
  • Guanosine Triphosphate
  • Humans
  • Membrane Proteins
  • STAT3 Transcription Factor / genetics
  • Tumor Microenvironment
  • Vascular Endothelial Growth Factor A

Substances

  • Allopurinol
  • Cisplatin
  • Guanosine Triphosphate
  • VEGFA protein, human
  • Vascular Endothelial Growth Factor A
  • STAT3 protein, human
  • STAT3 Transcription Factor
  • TMTC3 protein, human
  • Carrier Proteins
  • Membrane Proteins