Skip to main content

Advertisement

Log in

Identification of two potential immune-related biomarkers of Graves’ disease based on integrated bioinformatics analyses

  • Original Article
  • Published:
Endocrine Aims and scope Submit manuscript

Abstract

Background

Graves’ disease (GD) is an autoimmune disease, the incidence of which is increasing yearly. GD requires long-life therapy. Therefore, the potential immune-related biomarkers of GD need to be studied.

Method

In our study, differentially expressed genes (DEGs) were derived from the online Gene Expression Omnibus (GEO) microarray expression dataset GSE71956. Protein‒protein interaction (PPI) network analyses were used to identify hub genes, which were validated by qPCR. GSEA was used to screen potential pathways and related immune cells. Next, CIBERSORT analysis was used to further explore the immune subtype distribution pattern among hub genes. ROC curves were used to analyze the specificity and sensitivity of hub genes.

Result

44 DEGs were screened from the GEO dataset. Two hub genes, EEF1A1 and EIF4B, were obtained from the PPI network and validated by qPCR (p < 0.05). GSEA was conducted to identify potential pathways and immune cells related to these the two hub genes. Immune cell subtype analysis revealed that hub genes had extensive associations with many different types of immune cells, particularly resting memory CD4+ T cells. AUCs of ROC analysis were 0.687 and 0.733 for EEF1A1 and EIF4B, respectively.

Conclusion

Our study revealed two hub genes, EEF1A1 and EIF4B, that are associated with resting memory CD4+ T cells and potential immune-related molecular biomarkers and therapeutic targets of GD.

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

Similar content being viewed by others

References

  1. Y. He, et al. Association of HLA-B and HLA-DRB1 polymorphisms with antithyroid drug-induced agranulocytosis in a Han population from northern China. Sci. Rep. 7(1), 11950 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  2. S. De Leo, S.Y. Lee, L.E. Braverman,, Hyperthyroidism. Lancet 388(10047), 906–918 (2016)

    Article  PubMed  PubMed Central  Google Scholar 

  3. T.J. Smith, L. Hegedus, Graves’ disease. N. Engl. J. Med. 375(16), 1552–1565 (2016)

    Article  PubMed  Google Scholar 

  4. P.N. Taylor et al. Global epidemiology of hyperthyroidism and hypothyroidism. Nat. Rev. Endocrinol. 14(5), 301–316 (2018)

    Article  PubMed  Google Scholar 

  5. T.F. Davies et al. Graves’ disease. Nat. Rev. Dis. Prim. 6(1), 52 (2020)

    Article  PubMed  Google Scholar 

  6. H.T. Hao et al. Treatment of Graves’ ophthalmopathy with an in-house Phosphorus-32 source: Initial clinical observations. Exp. Ther. Med 14(4), 2795–2800 (2017)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. W. Gu, S. Miller, C.Y. Chiu, Clinical metagenomic next-generation sequencing for pathogen detection. Annu Rev. Pathol. 14, 319–338 (2019)

    Article  CAS  PubMed  Google Scholar 

  8. A. Antonelli et al. Graves’ disease: Clinical manifestations, immune pathogenesis (cytokines and chemokines) and therapy. Best. Pr. Res Clin. Endocrinol. Metab. 34(1), 101388 (2020)

    Article  CAS  Google Scholar 

  9. M. Limbach et al. Epigenetic profiling in CD4+ and CD8+ T cells from Graves’ disease patients reveals changes in genes associated with T cell receptor signaling. J. Autoimmun. 67, 46–56 (2016)

    Article  CAS  PubMed  Google Scholar 

  10. T.J. Smith, L. Hegedus, Graves’ disease. N. Engl. J. Med 376(2), 185 (2017)

    PubMed  Google Scholar 

  11. J.Q. Chen, P. Szodoray, M. Zeher, Toll-like receptor pathways in autoimmune diseases. Clin. Rev. Allergy Immunol. 50(1), 1–17 (2016)

    Article  CAS  PubMed  Google Scholar 

  12. K.A. Dvornikova et al. Polymorphism of toll-like receptor genes and autoimmune endocrine diseases. Autoimmun. Rev. 19(4), 102496 (2020)

    Article  CAS  PubMed  Google Scholar 

  13. S.Y. Han et al. High-mobility group box 1 is associated with the inflammatory pathogenesis of graves’ orbitopathy. Thyroid 29(6), 868–878 (2019)

    Article  CAS  PubMed  Google Scholar 

  14. S. Shah et al. Ras and Rap1: A tale of two GTPases. Semin Cancer Biol. 54, 29–39 (2019)

    Article  CAS  PubMed  Google Scholar 

  15. N. Minato, K. Kometani, M. Hattori, Regulation of immune responses and hematopoiesis by the Rap1 signal. Adv. Immunol. 93, 229–264 (2007)

    Article  CAS  PubMed  Google Scholar 

  16. J.J. Hamey, M.R. Wilkins, Methylation of elongation factor 1A: where, who, and why?. Trends Biochem Sci. 43(3), 211–223 (2018)

    Article  CAS  PubMed  Google Scholar 

  17. J. Pelletier, N. Sonenberg, The organizing principles of eukaryotic ribosome recruitment. Annu Rev. Biochem 88, 307–335 (2019)

    Article  CAS  PubMed  Google Scholar 

  18. H.J. Lee et al. Immunogenetics of autoimmune thyroid diseases: A comprehensive review. J. Autoimmun. 64, 82–90 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. P.S. Minhas et al. Macrophage de novo NAD(+) synthesis specifies immune function in aging and inflammation. Nat. Immunol. 20(1), 50–63 (2019)

    Article  CAS  PubMed  Google Scholar 

  20. E.L. Mills et al. Succinate dehydrogenase supports metabolic repurposing of mitochondria to drive inflammatory macrophages. Cell 167(2), 457–470 e13 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. S.A. Vardhana et al. Impaired mitochondrial oxidative phosphorylation limits the self-renewal of T cells exposed to persistent antigen. Nat. Immunol. 21(9), 1022–1033 (2020)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Y. Zhao et al. m(6)A-binding proteins: the emerging crucial performers in epigenetics. J. Hematol. Oncol. 13(1), 35 (2020)

    Article  PubMed  PubMed Central  Google Scholar 

  23. S. Fang et al. Mechanisms that underly T cell immunity in graves’ orbitopathy. Front Endocrinol. (Lausanne) 12, 648732 (2021)

    Article  Google Scholar 

  24. S. Crotty, Do memory CD4 T cells keep their cell-type programming: plasticity versus fate commitment? Complexities of interpretation due to the heterogeneity of memory CD4 T cells, including T follicular helper cells. Cold Spring Harb. Perspect. Biol. 10(3), a032102 (2018)

    Article  PubMed  PubMed Central  Google Scholar 

  25. S. Sengupta, R.F. Siliciano, Targeting the latent reservoir for HIV-1. Immunity 48(5), 872–895 (2018)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. S. Sureshchandra et al. Phenotypic and epigenetic adaptations of cord blood CD4+ T cells to maternal obesity. Front Immunol. 12, 617592 (2021)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Z. Wang et al. IP-10 promotes latent HIV infection in resting memory CD4(+) T cells via LIMK-cofilin pathway. Front Immunol. 12, 656663 (2021)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. N. Corcos et al. Oral Fc-coupled preproinsulin achieves systemic and thymic delivery through the neonatal Fc receptor and partially delays autoimmune diabetes. Front Immunol. 12, 616215 (2021)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. T. Liu et al. Human eukaryotic elongation factor 1A forms oligomers through specific cysteine residues. Acta Biochim Biophys. Sin. (Shanghai) 47(12), 1011–1017 (2015)

    Article  CAS  Google Scholar 

  30. Z. Cai et al. A three-gene signature and clinical outcome in pediatric acute myeloid leukemia. Clin. Transl. Oncol. 23(4), 866–873 (2021)

    Article  CAS  PubMed  Google Scholar 

  31. H.J. Ditzel et al. Cloning and expression of a novel human antibody-antigen pair associated with Felty’s syndrome. Proc. Natl Acad. Sci. USA 97(16), 9234–9239 (2000)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Q. Feng et al. Etodolac improves collagen induced rheumatoid arthritis in rats by inhibiting synovial inflammation, fibrosis and hyperplasia. Mol. Biomed. 2(1), 33 (2021)

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Throughout the writing of this dissertation, I have received a great deal of support and assistance. I would first like to thank my supervisor, H.X., whose expertise was invaluable in formulating the research questions and methodology. Your insightful feedback pushed me to sharpen my thinking and brought my work to a higher level. I would particularly like to acknowledge my colleagues J.W., H.Z., B.L., Y.C., F.Q., and J.L. for their wonderful collaboration and patient support.

Funding

This work has been in part supported by the National Natural Science Foundation of China (81773741 and 81973329) and Natural Science Foundation Project of Shanghai (19ZR1440800).

Author information

Authors and Affiliations

Authors

Contributions

Y.Z., X.X. and H.X. conceived of and designed the project. J.W., B.L. and F.Q. acquired the data, and Y.C., B.L. and H.Z. analyzed and interpreted the data. Y.Z. and J.W. wrote the paper.

Corresponding authors

Correspondence to Xin Xie or Huanbai Xu.

Ethics declarations

Conflict of interest

The authors declare no competing interests.

Ethical approval

This study was performed in line with the principles of the Declaration of Helsinki. Approval was granted by the Ethical Committee of Shanghai General Hospital (Date 2018-2-27/No 2018KY088).

Additional information

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

Supplementary Information

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

Zhang, Y., Wei, J., Zhou, H. et al. Identification of two potential immune-related biomarkers of Graves’ disease based on integrated bioinformatics analyses. Endocrine 78, 306–314 (2022). https://doi.org/10.1007/s12020-022-03156-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12020-022-03156-y

Keywords

Navigation