Skip to content
2000
Volume 28, Issue 11
  • ISSN: 1386-2073
  • E-ISSN: 1875-5402

Abstract

Objective

This study aimed to examine the associations of FTO expression with prognosis, tumor microenvironment (TME), immune cell infiltration, immune checkpoint genes, and relevant signaling pathways in GC. Furthermore, the relationship between FTO and TGF-β was studied in GC.

Methods

The mRNA expression and clinical survival data of GC samples were obtained from The Cancer Genome Atlas Stomach Adenocarcinoma (TCGA-STAD). TIMER2, TNM plot, and GEPIA database were used to analyze FTO expression. The associations of FTO with prognosis and clinicopathologic features were assessed using the Kaplan-Meier plotter and UALCAN database, respectively. The R software was employed to analyze its related signaling pathways and the associations with TME, immune cell infiltration, and immune checkpoint genes. GEPIA and ENCORI were used to examine the association of FTO with TGF-β expression. The SRAMP website was utilized to predict m6A modification of TGF-β. IHC, Western blot, and qPCR were used to analyze the expression levels of FTO and TGF-β in clinical gastric cancer tissue samples or gastric cancer cell lines. In addition, a m6A RNA methylation assay kit was used to determine m6A levels in gastric cancer cells.

Results

FTO mRNA and protein levels were significantly elevated in GC compared to normal gastric tissues. Kaplan-Meier survival analysis suggested that upregulated FTO was associated with a worse prognosis in GC. Upregulated FTO was markedly correlated with differentiation degree, lymph node metastasis, and clinical TNM stage. GO and KEGG pathway analyses revealed that FTO-associated molecules were enriched in neuroactive ligand-receptor interaction, calcium signaling, PI3k-Akt signaling, cAMP signaling pathways, and TGF-β signaling pathways, among others. The TME score was remarkably higher in the high-FTO group than in the low-FTO group. Furthermore, FTO expression had positive correlations with different types of immune cells and immune checkpoint genes. Moreover, FTO may regulate TGF-β in an m6A RNA modification manner in GC.

Conclusion

FTO may become an independent predictive prognostic biomarker correlating with TME, immune cell infiltration, and immune checkpoint genes in gastric cancer and might influence GC progression by regulating TGF-β expression.

Loading

Article metrics loading...

/content/journals/cchts/10.2174/0113862073299882240530051559
2024-06-14
2025-10-15
Loading full text...

Full text loading...

References

  1. ThriftA.P. WenkerT.N. El-SeragH.B. Global burden of gastric cancer: epidemiological trends, risk factors, screening and prevention.Nat. Rev. Clin. Oncol.202320533834910.1038/s41571‑023‑00747‑0 36959359
    [Google Scholar]
  2. CuiY. ZhangC. MaS. LiZ. WangW. LiY. MaY. FangJ. WangY. CaoW. GuanF. RNA m6A demethylase FTO-mediated epigenetic up-regulation of LINC00022 promotes tumorigenesis in esophageal squamous cell carcinoma.J. Exp. Clin. Cancer Res.202140129410.1186/s13046‑021‑02096‑1 34544449
    [Google Scholar]
  3. BianX. ShiD. XingK. ZhouH. LuL. YuD. WuW. AMD1 upregulates hepatocellular carcinoma cells stemness by FTO mediated mRNA demethylation.Clin. Transl. Med.2021113e35210.1002/ctm2.352 33783988
    [Google Scholar]
  4. TanZ. ShiS. XuJ. LiuX. LeiY. ZhangB. HuaJ. MengQ. WangW. YuX. LiangC. RNA N6-methyladenosine demethylase FTO promotes pancreatic cancer progression by inducing the autocrine activity of PDGFC in an m6A-YTHDF2-dependent manner.Oncogene202241202860287210.1038/s41388‑022‑02306‑w 35422475
    [Google Scholar]
  5. XuY. YeS. ZhangN. ZhengS. LiuH. ZhouK. WangL. CaoY. SunP. WangT. The FTO/miR‐181b‐3p/ARL5B signaling pathway regulates cell migration and invasion in breast cancer.Cancer Commun.2020401048450010.1002/cac2.12075 32805088
    [Google Scholar]
  6. GuoC. ChuH. GongZ. ZhangB. LiC. ChenJ. HuangL. HOXB13 promotes gastric cancer cell migration and invasion via IGF-1R upregulation and subsequent activation of PI3K/AKT/mTOR signaling pathway.Life Sci.202127811952210.1016/j.lfs.2021.119522 33894267
    [Google Scholar]
  7. YangZ. JiangX. ZhangZ. ZhaoZ. XingW. LiuY. JiangX. ZhaoH. HDAC3-dependent transcriptional repression of FOXA2 regulates FTO/m6A/MYC signaling to contribute to the development of gastric cancer.Cancer Gene Ther.2021281-214115510.1038/s41417‑020‑0193‑8 32655129
    [Google Scholar]
  8. ZhouY. WangQ. DengH. XuB. ZhouY. LiuJ. LiuY. ShiY. ZhengX. JiangJ. N6-methyladenosine demethylase FTO promotes growth and metastasis of gastric cancer via m6A modification of caveolin-1 and metabolic regulation of mitochondrial dynamics.Cell Death Dis.20221317210.1038/s41419‑022‑04503‑7 35064107
    [Google Scholar]
  9. ZhangY. GaoL. WangW. ZhangT. DongF. DingW.M. 6 A demethylase fat mass and obesity‐associated protein regulates cisplatin resistance of gastric cancer by modulating autophagy activation through ULK1.Cancer Sci.202211393085309610.1111/cas.15469 35730319
    [Google Scholar]
  10. JiaG. FuY. ZhaoX. DaiQ. ZhengG. YangY. YiC. LindahlT. PanT. YangY.G. HeC. N6-Methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO.Nat. Chem. Biol.201171288588710.1038/nchembio.687 22002720
    [Google Scholar]
  11. HuangJ. SunW. WangZ. LvC. ZhangT. ZhangD. DongW. ShaoL. HeL. JiX. ZhangP. ZhangH. FTO suppresses glycolysis and growth of papillary thyroid cancer via decreasing stability of APOE mRNA in an N6-methyladenosine-dependent manner.J. Exp. Clin. Cancer Res.20224114210.1186/s13046‑022‑02254‑z 35090515
    [Google Scholar]
  12. WangY. LiM. ZhangL. ChenY. ZhangS. m6A demethylase FTO induces NELL2 expression by inhibiting E2F1 m6A modification leading to metastasis of non-small cell lung cancer.Mol. Ther. Oncolytics20212136737610.1016/j.omto.2021.04.011 34169146
    [Google Scholar]
  13. LinZ. WanA.H. SunL. LiangH. NiuY. DengY. YanS. WangQ.P. BuX. ZhangX. HuK. WanG. HeW. N6-methyladenosine demethylase FTO enhances chemo-resistance in colorectal cancer through SIVA1-mediated apoptosis.Mol. Ther.202331251753410.1016/j.ymthe.2022.10.012 36307991
    [Google Scholar]
  14. PittJ.M. MarabelleA. EggermontA. SoriaJ.C. KroemerG. ZitvogelL. Targeting the tumor microenvironment: Removing obstruction to anticancer immune responses and immunotherapy.Ann. Oncol.20162781482149210.1093/annonc/mdw168 27069014
    [Google Scholar]
  15. WanW. AoX. ChenQ. YuY. AoL. XingW. GuoW. WuX. PuC. HuX. LiZ. YaoM. LuoD. XuX. METTL3/IGF2BP3 axis inhibits tumor immune surveillance by upregulating N6-methyladenosine modification of PD-L1 mRNA in breast cancer.Mol. Cancer20222116010.1186/s12943‑021‑01447‑y 35197058
    [Google Scholar]
  16. SuR. DongL. LiY. GaoM. HanL. WunderlichM. DengX. LiH. HuangY. GaoL. LiC. ZhaoZ. RobinsonS. TanB. QingY. QinX. PrinceE. XieJ. QinH. LiW. ShenC. SunJ. KulkarniP. WengH. HuangH. ChenZ. ZhangB. WuX. OlsenM.J. MüschenM. MarcucciG. SalgiaR. LiL. FathiA.T. LiZ. MulloyJ.C. WeiM. HorneD. ChenJ. Targeting FTO suppresses cancer stem cell maintenance and immune evasion.Cancer Cell20203817996.e1110.1016/j.ccell.2020.04.017 32531268
    [Google Scholar]
  17. LiX. ChenW. GaoY. SongJ. GuY. ZhangJ. ChengX. AiY. Fat mass and obesity‐associated protein regulates arecoline‐exposed oral cancer immune response through programmed cell death‐ligand 1.Cancer Sci.202211392962297310.1111/cas.15332 35289035
    [Google Scholar]
  18. YuH. LiuJ. BuX. MaZ. YaoY. LiJ. ZhangT. SongW. XiaoX. SunY. XiongW. ShiJ. DaiP. XiangB. DuanH. YanX. WuF. ZhangW.C. LinD. HuH. ZhangH. SlackF.J. HeH.H. FreemanG.J. WeiW. ZhangJ. Targeting METTL3 reprograms the tumor microenvironment to improve cancer immunotherapy.Cell Chem. Biol.2023S2451-9456(23)00291-X10.1016/j.chembiol.2023.09.001 37751743
    [Google Scholar]
  19. WangH. TangA. CuiY. GongH. LiH. LRPPRC facilitates tumor progression and immune evasion through upregulation of m6A modification of PD-L1 mRNA in hepatocellular carcinoma.Front. Immunol.202314114477410.3389/fimmu.2023.1144774 37063837
    [Google Scholar]
  20. WenJ. XueL. WeiY. LiangJ. JiaW. YongT. ChuL. LiH. HanS. LiaoJ. ChenZ. LiuY. LiuQ. DingZ. LiangH. GanL. ChenX. HuangZ. ZhangB. YTHDF2 is a therapeutic target for HCC by suppressing immune evasion and angiogenesis through ETV5/PD‐L1/VEGFA Axis.Adv. Sci.20241113230724210.1002/advs.202307242 38247171
    [Google Scholar]
  21. WangY. JinP. WangX. N6-methyladenosine regulator YTHDF1 represses the CD8 + T cell-mediated antitumor immunity and ferroptosis in prostate cancer via m6A/PD-L1 manner.Apoptosis2024291-214215310.1007/s10495‑023‑01885‑7 37698736
    [Google Scholar]
  22. PengY. ZhangZ. YangG. DaiZ. CaiX. LiuZ. YunQ. XuL. N6-methyladenosine reader protein IGF2BP1 suppresses CD8 + T cells-mediated tumor cytotoxicity and apoptosis in colon cancer.Apoptosis2024293-433134310.1007/s10495‑023‑01893‑7 37848671
    [Google Scholar]
  23. JiangT. XiaY. LiY. LuC. LinJ. ShenY. LvJ. XieL. GuC. XuZ. WangL. TRIM29 promotes antitumor immunity through enhancing IGF2BP1 ubiquitination and subsequent PD-L1 downregulation in gastric cancer.Cancer Lett.202458121651010.1016/j.canlet.2023.216510 38029830
    [Google Scholar]
  24. YouY. WenD. ZengL. LuJ. XiaoX. ChenY. SongH. LiuZ. ALKBH5/MAP3K8 axis regulates PD-L1+ macrophage infiltration and promotes hepatocellular carcinoma progression.Int. J. Biol. Sci.202218135001501810.7150/ijbs.70149 35982895
    [Google Scholar]
  25. QinS. LiuG. JinH. ChenX. HeJ. XiaoJ. QinY. MaoY. ZhaoL. The comprehensive expression and functional analysis of m6A modification “readers” in hepatocellular carcinoma.Aging202214156269629810.18632/aging.204217 35963644
    [Google Scholar]
  26. PengC. XiongF. PuX. HuZ. YangY. QiaoX. JiangY. HanM. WangD. LiX. m6A methylation modification and immune cell infiltration: implications for targeting the catalytic subunit m6A-METTL complex in gastrointestinal cancer immunotherapy.Front. Immunol.202314132603110.3389/fimmu.2023.1326031 38187373
    [Google Scholar]
  27. ZhangY. ChenY. ChenR. ZhouH. LinY. LiB. SongH. ZhouG. DongM. XuH. YTHDF3 as a prognostic predictive biomarker of thyroid cancer and its correlation with immune infiltration.BMC Cancer202323188210.1186/s12885‑023‑11361‑9 37726690
    [Google Scholar]
  28. WangZ. ShangJ. QiuY. ChengH. TaoM. XieE. PeiX. LiW. ZhangL. WuA. LiG. Suppression of the METTL3-m6A-integrin β1 axis by extracellular acidification impairs T cell infiltration and antitumor activity.Cell Rep.202443211379610.1016/j.celrep.2024.113796 38367240
    [Google Scholar]
  29. DerynckR. TurleyS.J. AkhurstR.J. TGFβ biology in cancer progression and immunotherapy.Nat. Rev. Clin. Oncol.202118193410.1038/s41571‑020‑0403‑1 32710082
    [Google Scholar]
  30. LiJ. ChenF. PengY. LvZ. LinX. ChenZ. WangH. N6-methyladenosine regulates the expression and secretion of TGFβ1 to affect the epithelial–mesenchymal transition of cancer cells.Cells20209229610.3390/cells9020296 31991845
    [Google Scholar]
  31. LiuY. DaM. Wilms tumor 1 associated protein promotes epithelial mesenchymal transition of gastric cancer cells by accelerating TGF-β and enhances chemoradiotherapy resistance.J. Cancer Res. Clin. Oncol.202314973977398810.1007/s00432‑022‑04320‑7 36030434
    [Google Scholar]
  32. SunZ. SuZ. ZhouZ. WangS. WangZ. TongX. LiC. WangY. ChenX. LeiZ. ZhangH.T. RNA demethylase ALKBH5 inhibits TGF‐β‐induced EMT by regulating TGF‐β/SMAD signaling in non‐small cell lung cancer.FASEB J.2022365e2228310.1096/fj.202200005RR 35344216
    [Google Scholar]
/content/journals/cchts/10.2174/0113862073299882240530051559
Loading
/content/journals/cchts/10.2174/0113862073299882240530051559
Loading

Data & Media loading...


  • Article Type:
    Research Article
Keyword(s): FTO; gastric cancer; immune infiltrate; prognosis; TGF-β
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error
Please enter a valid_number test