Skip to main content

Advertisement

Log in

Overexpressed FAM111B degrades GSDMA to promote esophageal cancer tumorigenesis and cisplatin resistance

  • Research
  • Published:
Cellular Oncology Aims and scope Submit manuscript

Abstract

Background

Chemotherapeutic agents such as cisplatin are commonly used in patients with clinically unresectable or recurrent esophageal cancer (ESCA). However, patients often develop resistance to cisplatin, which in turn leads to a poor prognosis. Studies have shown that FAM111B may be involved in the development of tumors as an oncogene or tumor suppressor gene. However, the pathological role and corresponding mechanism of FAM111B in ESCA are still unclear.

Methods

The GEPIA web tool, ENCORI Pan-Cancer Analysis Platform and UALCAN-TCGA database were used to study the expression of FAM111B in ESCA. CCK-8, angiogenesis, Transwell and xenograft assays were applied to explore the biological function of FAM111B in ESCA. Western blot, RT-qPCR, and RNA-seq analyses were applied to study the FAM111B/GSDMA axis in the progression of ESCA cells. CCK-8 and xenograft assays were used to study the role of the FAM111B/GSDMA axis in determining the sensitivity of ESCA to cisplatin.

Results

Our results demonstrated that FAM111B is highly expressed in ESCA tissues compared to normal tissues. We showed that FAM111B promotes the progression of ESCC cells by binding to GSDMA and that the trypsin protease domain is essential for the activity of FAM111B. Furthermore, we showed that the FAM111B/GSDMA axis regulates cisplatin sensitivity in ESCA.

Conclusions

Overall, we identified a novel FAM111B/GSDMA axis regulating ESCA tumorigenesis and chemosensitivity, at least in ESCC cells.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

Data availability

The databases used and/or analyzed during the current study are available from the corresponding authors (Xiaoxiong Xiao, E-mail: xiaoxx1988@csu.edu.cn) on reasonable request.

References

  1. H.C. Puhr, G.W. Prager, A. Ilhan-Mutlu, How we treat esophageal squamous cell carcinoma. ESMO Open 8(1), 100789 (2023)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. M. Sheikh et al., Current status and future prospects for esophageal cancer. Cancers (Basel) 15(3), 765 (2023)

  3. J. An et al., Natural products for esophageal cancer therapy: from traditional medicine to modern drug discovery. Int J Mol Sci 23(21), 13558 (2022)

  4. Y. Baba et al., Tumor immune microenvironment and immune checkpoint inhibitors in esophageal squamous cell carcinoma. Cancer Sci 111(9), 3132–3141 (2020)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Y. Jia et al., Long non-coding RNA NORAD/miR-224-3p/MTDH axis contributes to CDDP resistance of esophageal squamous cell carcinoma by promoting nuclear accumulation of β-catenin. Mol Cancer 20(1), 162 (2021)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. E. Perland, R. Fredriksson, Classification systems of secondary active transporters. Trends Pharmacol Sci 38(3), 305–315 (2017)

    Article  CAS  PubMed  Google Scholar 

  7. A.K. Holzer, S.B. Howell, The internalization and degradation of human copper transporter 1 following cisplatin exposure. Cancer Res 66(22), 10944–10952 (2006)

    Article  CAS  PubMed  Google Scholar 

  8. A. Yaneff et al., MRP4/ABCC4 As a new therapeutic target: meta-analysis to determine cAMP binding sites as a tool for drug design. Curr Med Chem 26(7), 1270–1307 (2019)

    Article  CAS  PubMed  Google Scholar 

  9. J. Zhou et al., The drug-resistance mechanisms of five platinum-based antitumor agents. Front Pharmacol 11, 343 (2020)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. S. Dilruba, G.V. Kalayda, Platinum-based drugs: past, present and future. Cancer Chemother Pharmacol 77(6), 1103–1124 (2016)

    Article  CAS  PubMed  Google Scholar 

  11. W.P. Roos, B. Kaina, DNA damage-induced cell death: from specific DNA lesions to the DNA damage response and apoptosis. Cancer Lett 332(2), 237–248 (2013)

    Article  CAS  PubMed  Google Scholar 

  12. X. Zhu et al., DNMT3B-mediated FAM111B methylation promotes papillary thyroid tumor glycolysis, growth and metastasis. Int J Biol Sci 18(11), 4372–4387 (2022)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. S. Mercier et al., Mutations in FAM111B cause hereditary fibrosing poikiloderma with tendon contracture, myopathy, and pulmonary fibrosis. Am J Hum Genet 93(6), 1100–1107 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. H. Sun et al., FAM111B, a direct target of p53, promotes the malignant process of lung adenocarcinoma. Onco Targets Ther 12, 2829–2842 (2019)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. S. Mercier et al., FAM111B mutation is associated with pancreatic cancer predisposition. Pancreas 48(5), e41–e42 (2019)

    Article  CAS  PubMed  Google Scholar 

  16. H. Wu, C. Liang, Pan-cancer analysis of the tumorigenic effect and prognostic diagnostic value of fam111b in human carcinomas. Int J Gen Med 16, 1845–1865 (2023)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. X. Xiao et al., Silencing of UAP1L1 inhibits proliferation and induces apoptosis in esophageal squamous cell carcinoma. Mol Carcinog 60(3), 179–187 (2021)

    Article  CAS  PubMed  Google Scholar 

  18. K. Kawasaki et al., FAM111B enhances proliferation of KRAS-driven lung adenocarcinoma by degrading p16. Cancer Sci 111(7), 2635–2646 (2020)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. S. Wenmaekers et al., A Potential Role for HUWE1 in Modulating Cisplatin Sensitivity. Cells 10(5), 1262 (2021)

  20. C. He et al., Elevated H3K27me3 levels sensitize osteosarcoma to cisplatin. Clin Epigenetics 11(1), 8 (2019)

    Article  PubMed  PubMed Central  Google Scholar 

  21. I. Lakshmanan et al., MUC16 Regulates TSPYL5 for Lung Cancer Cell Growth and Chemoresistance by Suppressing p53. Clin Cancer Res 23(14), 3906–3917 (2017)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. X. Gong et al., The structure and regulation of the E3 ubiquitin ligase HUWE1 and its biological functions in cancer. Invest New Drugs 38(2), 515–524 (2020)

    Article  MathSciNet  CAS  PubMed  Google Scholar 

  23. C. Guo et al., Integrated analysis of multi-omics alteration, immune profile, and pharmacological landscape of pyroptosis-derived lncRNA pairs in gastric cancer. Front Cell Dev Biol 10, 816153 (2022)

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  24. M. Boivin et al., CA125 (MUC16) tumor antigen selectively modulates the sensitivity of ovarian cancer cells to genotoxic drug-induced apoptosis. Gynecol Oncol 115(3), 407–413 (2009)

    Article  CAS  PubMed  Google Scholar 

  25. M. Liu et al., Predictive biomarkers of dicycloplatin resistance or susceptibility in prostate cancer. Front Genet 12, 669605 (2021)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. X. Lu, T. Guo, X. Zhang, Pyroptosis in Cancer: Friend or Foe? Cancers (Basel) 13(14), 3620 (2021)

  27. X. Ouyang et al., Pyroptosis, inflammasome, and gasdermins in tumor immunity. Innate Immun 29(1–2), 3–13 (2023)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. J. Shi et al., Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 526(7575), 660–665 (2015)

    Article  ADS  CAS  PubMed  Google Scholar 

  29. K.W. Chen et al., Extrinsic and intrinsic apoptosis activate pannexin-1 to drive NLRP3 inflammasome assembly. Embo J 38(10), e101638 (2019)

  30. Q. Wang et al., A bioorthogonal system reveals antitumour immune function of pyroptosis. Nature 579(7799), 421–426 (2020)

    Article  ADS  CAS  PubMed  Google Scholar 

  31. W. Deng et al., Streptococcal pyrogenic exotoxin B cleaves GSDMA and triggers pyroptosis. Nature 602(7897), 496–502 (2022)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  32. W. Zou, J.D. Wolchok, L. Chen, PD-L1 (B7–H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci Transl Med 8(328), 328rv4 (2016)

    Article  PubMed  PubMed Central  Google Scholar 

  33. A. Ribas, J.D. Wolchok, Cancer immunotherapy using checkpoint blockade. Science 359(6382), 1350–1355 (2018)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  34. M. Macchiaiolo et al., Expanding phenotype of FAM111B-related disease focusing on liver involvement: Literature review, report of a case with end-stage liver disease and proposal for a new acronym. Am J Med Genet A 188(10), 2920–2931 (2022)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. A. Seo et al., FAM111B mutation is associated with inherited exocrine pancreatic dysfunction. Pancreas 45(6), 858–862 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. A.L. Welter, Y.J. Machida, Functions and evolution of FAM111 serine proteases. Front Mol Biosci 9, 1081166 (2022)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. A. Arowolo, C. Rhoda, N. Khumalo, Mutations within the putative protease domain of the human FAM111B gene may predict disease severity and poor prognosis: A review of POIKTMP cases. Exp Dermatol 31(5), 648–654 (2022)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. S. Hoffmann et al., FAM111 protease activity undermines cellular fitness and is amplified by gain-of-function mutations in human disease. EMBO Rep 21(10), e50662 (2020)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work were supported by grants from the National Natural Science Youth Foundation of China (Grant No. 82203838 (Xiaoxiong Xiao)); Natural Science Foundation General Program of Hunan Province (Grant No. 2022JJ40830 (Xiaoxiong Xiao)); Natural Science Foundation General Program of Changsha City (Grant No. kq2014290 (Xiaoxiong Xiao)); National Multidisciplinary Cooperative Diagnosis and Treatment Capacity Building Project for Major Diseases (Lung Cancer, grant number: z027002 (Xiaoxiong Xiao)); National Natural Science Foundation of China (Grant No. 81772800, 82072945 (Pian Liu)).

Author information

Authors and Affiliations

Authors

Contributions

Haiqin Wang. supervised the experiments, and was responsible for bioinformatics and statistical analysis. Haohui Wang. carried out the in vitro and in vivo experiments, and wrote the original draft. Jiajing Chen. collaborated to collect specimens and record clinical information. Pian Liu. conducted the major revision work, and provided critical reviews. Xiaoxiong Xiao. generated the study design, provided the clinical specimens and revised the manuscript.

Corresponding authors

Correspondence to Pian Liu or Xiaoxiong Xiao.

Ethics declarations

Ethics approval and consent to participate

The study was conducted in accordance with the principles of the Declaration of Helsinki principles. It was approved by the Animal Use and Care Committees at Xiangya hospital, Central South University.

Consent for publication

All subjects have written informed consent.

Conflict of interest

The authors have declared that no conflict of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Haiqin Wang and Haohui Wang contributed equally.

Supplementary Information

Below is the link to the electronic supplementary material.

13402_2023_871_MOESM1_ESM.tif

Supplementary file1 Supplementary figure 1 A and B, The univariate and multivariate Cox regression analyses through the GEO dataset GSE53622. p values as indicated. C and D, Lymph node metastasis of KYSE410 cells with FAM111B knockdown. E, The expression of SNAI1 in RNA-seq of FAM111B. Fand G, KYSE410 and TE-1 cells were infected with shControl, shFAM111B #1, or shFAM111B #2 for 72 h. Cells were collected for RT-qPCR analysis (F), and Western blot analysis (G). H, The excised subcutaneous tumors of nude mice were subjected to IHC staining of SNAI1. Data presents as mean ± SD with six replicates. ***, p < 0.001 (TIF 35268 KB)

13402_2023_871_MOESM2_ESM.tif

Supplementary file2 Supplementary figure 2 A and B, KYSE410 cells were transfected with indicated shRNAs for 72 h. Cells were collected and subcutaneously injected into the nude mice. The tumor growth curve was shown in panel B. Data presents as mean ± SD with six replicates. ***, p < 0.001. C and D, KYSE410 and TE-1 cells were transfected with indicated shRNAs for 72 h and treated with cisplatin, cells were collected for Western blot analysis to measure the expression of cleaved caspase 3. ***, p < 0.001. E and F, The excised subcutaneous tumors of nude mice were subjected to IHC staining of cleaved caspase 3. Data presents as mean ± SD with six replicates. ***, p < 0.001 (TIF 47227 KB)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, H., Wang, H., Chen, J. et al. Overexpressed FAM111B degrades GSDMA to promote esophageal cancer tumorigenesis and cisplatin resistance. Cell Oncol. 47, 343–359 (2024). https://doi.org/10.1007/s13402-023-00871-0

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13402-023-00871-0

Keywords

Navigation