Skip to main content

Advertisement

Log in

Glycoproteomic analysis reveals the effects of bisecting GlcNAc in intrahepatic cholangiocarcinoma

  • Original Article
  • Published:
Glycoconjugate Journal Aims and scope Submit manuscript

Abstract

Intrahepatic cholangiocarcinoma (ICC) is the second major subtype of primary liver cancer and has caused more and more attention with increasing incidence and mortality worldwide. Our previous study found that bisecting N-glycans are commonly increased in ICC, while the effects and potential functions of bisecting GlcNAc in ICC are still largely unclear. In this study, we further confirmed that the structures of bisecting GlcNAc were significantly up-regulated in ICC compared with paracancer tissues by glycoproteomic data and lectin histochemistry. The expression of its glycosyltransferase MGAT3 was also up-regulated in ICC tissues at both mRNA and protein levels, and expression of MGAT3 is negatively correlated with overall survival explored by bioinformatic analyses and published datasets from 255 patients. Next, the silencing of MGAT3 could inhibit the growth and invasion of ICC cells, and overexpressing of MGAT3 only promoted ICC cell invasion. Further glycoproteomic analysis showed that the commonly glycoproteins modified by bisecting GlcNAc after MGAT3-overexpression in two ICC cell lines were mainly involved in cell movement-related biological processes, such as cell adhesion, integrin-related and ECM-receptor interaction. This study sheds light on the potential effects of bisecting GlcNAc in ICC cells and suggests that MGAT3 might be used as a potential target in the therapy of ICC.

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

Similar content being viewed by others

Data Availability

The mass spectrometry data have been deposited to the ProteomeXchange Consortium (http://proteomecentral.proteomexchange.org) via the PRIDE partner repository with the dataset identifier PXD035321.

References

  1. Aljiffry, M., et al.: Evidence-based approach to cholangiocarcinoma: a systematic review of the current literature. J. Am. Coll. Surg. 208(1), 134–147 (2009)

    Article  Google Scholar 

  2. Gupta, A., Dixon, E.: Epidemiology and risk factors: intrahepatic cholangiocarcinoma. Hepatobiliary Surg. Nutr. 6(2), 101–104 (2017)

    Article  Google Scholar 

  3. de Jong, M.C., et al.: Intrahepatic Cholangiocarcinoma: An International Multi-Institutional Analysis of Prognostic Factors and Lymph Node Assessment. J. Clin. Oncol. 29(23), 3140–3145 (2011)

    Article  Google Scholar 

  4. Ohtsubo, K., Marth, J.D.: Glycosylation in cellular mechanisms of health and disease. Cell. 126(5), 855–867 (2006)

    Article  CAS  Google Scholar 

  5. Liu, H.M., et al.: Progress in research into the role of abnormal glycosylation modification in tumor immunity. Immunol. Lett. 229, 8–17 (2021)

    Article  CAS  Google Scholar 

  6. Li, J., et al.: Precision N-glycoproteomics reveals elevated LacdiNAc as a novel signature of intrahepatic cholangiocarcinoma. Mol. Oncol. 16(11), 2135–2152 (2022)

    Article  CAS  Google Scholar 

  7. Shen, J.C., et al.: StrucGP: de novo structural sequencing of site-specific N-glycan on glycoproteins using a modularization strategy. Nat. Methods. 18(8), 921– (2021)

    Article  CAS  Google Scholar 

  8. Nishikawa, A., et al.: Purification, cDNA cloning, and expression of UDP-N-acetylglucosamine: beta-D-mannoside beta-1,4 N-acetylglucosaminyltransferase III from rat kidney. J. Biol. Chem. 267(25), 18199–18204 (1992)

    Article  CAS  Google Scholar 

  9. Yoshimura, M., et al.: Bisecting N-acetylglucosamine on K562 cells suppresses natural killer cytotoxicity and promotes spleen colonization. Cancer Res. 56(2), 412–418 (1996)

    CAS  Google Scholar 

  10. Allam, H., et al.: The glycosyltransferase GnT-III activates Notch signaling and drives stem cell expansion to promote the growth and invasion of ovarian cancer. J. Biol. Chem. 292(39), 16351–16359 (2017)

    Article  CAS  Google Scholar 

  11. Sato, Y., et al.: Overexpression of N-acetylglucosaminyltransferase III enhances the epidermal growth factor-induced phosphorylation of ERK in HeLaS3 cells by up-regulation of the internalization rate of the receptors. J. Biol. Chem. 276(15), 11956–11962 (2001)

    Article  CAS  Google Scholar 

  12. Yoshimura, M., Ihara, Y., Taniguchi, N.: CHANGES OF BETA-1,4-N-ACETYLGLUCOSAMINYLTRANSFERASE-III (GNT-III) IN PATIENTS WITH LEUKEMIA. Glycoconj. J. 12(3), 234–240 (1995)

    Article  CAS  Google Scholar 

  13. Sheng, Y., et al.: Remodeling of glycoconjugates on CD44 enhances cell adhesion to hyaluronate, tumor growth and metastasis in B16 melanoma cells expressing beta1,4-N-acetylglucosaminyltransferase III. Int. J. Cancer. 73(6), 850–858 (1997)

    Article  CAS  Google Scholar 

  14. Bhaumik, M., et al.: Progression of hepatic neoplasms is severely retarded in mice lacking the, bisecting N-acetylglucosamine on N-glycans: Evidence for a glycoprotein factor that facilitates hepatic tumor progression. Cancer Res. 58(13), 2881–2887 (1998)

    CAS  Google Scholar 

  15. Li, J., et al.: MGAT3-mediated glycosylation of tetraspanin CD82 at asparagine 157 suppresses ovarian cancer metastasis by inhibiting the integrin signaling pathway. Theranostics. 10(14), 6467–6482 (2020)

    Article  CAS  Google Scholar 

  16. Song, Y.H., et al.: The Bisecting GlcNAc on N-Glycans Inhibits Growth Factor Signaling and Retards Mammary Tumor Progression. Cancer Res. 70(8), 3361–3371 (2010)

    Article  CAS  Google Scholar 

  17. Akama, R., et al.: N-acetylglucosaminyltransferase III expression is regulated by cell-cell adhesion via the E-cadherin-catenin-actin complex. Proteomics. 8(16), 3221–3228 (2008)

    Article  CAS  Google Scholar 

  18. Dong, L.Q., et al.: Proteogenomic characterization identifies clinically relevant subgroups of intrahepatic cholangiocarcinoma. Cancer Cell. 40(1), 70– (2022)

    Article  CAS  Google Scholar 

  19. Li, J., et al.: Precision N-glycoproteomics reveals elevated LacdiNAc as a novel signature of intrahepatic cholangiocarcinoma. Mol Oncol (2021)

  20. Weinstein, J.N., et al.: The Cancer Genome Atlas Pan-Cancer analysis project. Nat. Genet. 45(10), 1113–1120 (2013)

    Article  Google Scholar 

  21. Li, P., et al.: Comparative Proteomic Analysis of Polarized Human THP-1 and Mouse RAW264.7 Macrophages. Front. Immunol. 12, 700009 (2021)

    Article  CAS  Google Scholar 

  22. Shen, J., et al.: StrucGP: de novo structural sequencing of site-specific N-glycan on glycoproteins using a modularization strategy. Nat. Methods. 18(8), 921–929 (2021)

    Article  CAS  Google Scholar 

  23. Szklarczyk, D., et al.: STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47(D1), D607–D613 (2019)

    Article  CAS  Google Scholar 

  24. Dennis, G., et al.: DAVID: Database for annotation, visualization, and integrated discovery.Genome Biology, 4(9). (2003)

  25. Khan, S.A., et al.: Global trends in mortality from intrahepatic and extrahepatic cholangiocarcinoma. J. Hepatol. 71(6), 1261–1262 (2019)

    Article  Google Scholar 

  26. Li, J., et al.: Precision N-glycoproteomics reveals elevated LacdiNAc as a novel signature of intrahepatic cholangiocarcinoma.Molecular Oncology

  27. Dang, L.Y., et al.: Recognition of Bisecting N-Glycans on Intact Glycopeptides by Two Characteristic Ions in Tandem Mass Spectra. Anal. Chem. 91(9), 5478–5482 (2019)

    Article  CAS  Google Scholar 

  28. Yuan, W., et al.: Quantitative analysis of immunoglobulin subclasses and subclass specific glycosylation by LC-MS-MRM in liver disease. J. Proteom. 116, 24–33 (2015)

    Article  CAS  Google Scholar 

  29. Chen, Z.S., Gu, J.: Immunoglobulin G expression in carcinomas and cancer cell lines. Faseb J. 21(11), 2931–2938 (2007)

    Article  Google Scholar 

  30. Zauner, G., et al.: Glycoproteomic Analysis of Antibodies. Mol. Cell. Proteom. 12(4), 856–865 (2013)

    Article  CAS  Google Scholar 

  31. Pietraszek, K., et al.: Lumican: A new inhibitor of matrix metalloproteinase-14 activity. FEBS Lett. 588(23), 4319–4324 (2014)

    Article  CAS  Google Scholar 

  32. Bartsch, F., et al.: Influence of Lymphangio (L), Vascular (V), and Perineural (Pn) Invasion on Recurrence and Survival of Resected Intrahepatic Cholangiocarcinoma.Journal of Clinical Medicine, 10(11). (2021)

  33. Gu, J., et al.: A Mutual Regulation between Cell-Cell Adhesion and N-Glycosylation: Implication of the Bisecting GlcNAc for Biological Functions. J. Proteome Res. 8(2), 431–435 (2009)

    Article  CAS  Google Scholar 

  34. Strumane, K., Berx, G., Van Roy, F.: Cadherins in Cancer, in Cell Adhesion, J. Behrens and W.J. Nelson, Editors. Springer Berlin Heidelberg: Berlin, Heidelberg. p. 69–103. (2004)

  35. Niu, H.Y., Qu, A.N., Guan, C.Y.: Suppression of MGAT3 expression and the epithelial-mesenchymal transition of lung cancer cells by miR-188-5p. Biomedical J. 44(6), 678–685 (2021)

    Article  CAS  Google Scholar 

  36. Yoshimura, M., et al.: Aberrant glycosylation of E-cadherin enhances cell-cell binding to suppress metastasis. J. Biol. Chem. 271(23), 13811–13815 (1996)

    Article  CAS  Google Scholar 

  37. Deng, Q.Y., et al.: Expression of N-Acetylglucosaminyltransferase III Promotes Trophoblast Invasion and Migration in Early Human Placenta. Reproductive Sci. 26(10), 1373–1381 (2019)

    Article  CAS  Google Scholar 

  38. Yoshimura, M., Taniguchi, N.: Suppression of lung metastasis of B16 mouse melanoma cells by introduction of N-acetylglucosaminyltransferase III gene. Nihon rinsho. Japanese J. Clin. Med. 53(7), 1786–1790 (1995)

    CAS  Google Scholar 

  39. Takahashi, M., et al.: Core fucose and bisecting GlcNAc, the direct modifiers of the N-glycan core: their functions and target proteins. Carbohydr. Res. 344(12), 1387–1390 (2009)

    Article  CAS  Google Scholar 

  40. Czubryt, M.P.: Inhibition of collagen synthesis. (2016)

Download references

Acknowledgements

This work was supported by the National Key Research and Development Program of China (No. 2019YFA0905200), National Natural Science Foundation of China (No. 81773180, 91853123, 32101192, 21705127 and 81800655), and China Postdoctoral Science Foundation (No. 2019TQ0260 and 2019M663798).

Author information

Authors and Affiliations

Authors

Contributions

W.D. and S.S. designed the experiments; L.H. collected the tissue samples; W.D. and C.L. performed experiments with help from P.L., J.L, J.L.; W.D., C.L., Z.C and J.L., J.S., analyzed data; W.D., J.L., P.L., L.D., M.X., Z.Y., and S.S. wrote the paper with help from all authors.

Corresponding author

Correspondence to Shisheng Sun.

Ethics declarations

Competing interests

The authors have no relevant financial or non-financial interests to disclose.

Ethical statements

All human studies have been approved by the appropriate ethics committee and have therefore been performed in accordance with the ethical standards laid down in the 1964 Declaration of Helsinki and its later amendments. This study does not involve experiments of animal models.

Ethical approval

Tissue samples were prospectively collected from patients undergoing ICC resection at the First Affiliated Hospital of Xi’an Jiaotong University, China. The study was approved by Human Ethics Committee at the First Affiliated Hospital of Xi’an Jiaotong University, and written informed consents were obtained from all participants.

Consent to participate

All persons gave their informed consent prior to their inclusion in the study.

Consent to publish

Additional informed consent was obtained from all individual participants for whom identifying information is included in this article.

Additional information

Publisher’s Note

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

Electronic supplementary material

Rights and permissions

Springer Nature or its licensor 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

Dan, W., Li, C., Li, J. et al. Glycoproteomic analysis reveals the effects of bisecting GlcNAc in intrahepatic cholangiocarcinoma. Glycoconj J 39, 737–745 (2022). https://doi.org/10.1007/s10719-022-10085-5

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10719-022-10085-5

Keywords

Navigation