Open Access

Proteomic analysis of radioiodine‑refractory differentiated thyroid cancer identifies CHI3L1 upregulation in association with dysfunction of the sodium‑iodine symporter

  • Authors:
    • Yunjie Li
    • Fengqiong Hu
    • Jie Deng
    • Xin Huang
    • Chunyan Zhou
    • Mengxue Wu
    • Dong Duan
  • View Affiliations

  • Published online on: December 7, 2022     https://doi.org/10.3892/ol.2022.13622
  • Article Number: 36
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Radioiodine refractory differentiated thyroid cancer (RR‑DTC) is the main factor adversely affecting the overall survival rate of patients with thyroid cancer. The aim of the present study was to investigate the underlying molecular mechanism of pathogenesis of RR‑DTC and to explore novel therapeutic targets for clinical treatment. A proteomic analysis was performed using the tumor tissues of patients with RR‑DTC. A total of 6 metastatic lymph nodes were collected during lymph node dissection, 3 from patients with RR‑DTC and 3 from patients with papillary thyroid cancer. The expression of chitinase‑3‑like 1 (CHI3L1) and sodium‑iodine symporter (NIS) in the tumor tissue was detected by immunohistochemistry (IHC). Western blotting was used to detect the expression of CHI3L1, phosphorylated (p)‑MEK and p‑ERK1/2 in PTC‑K1 cells transfected with CHI3L1 overexpression vector. The proteomic analysis identified 665 differentially expressed proteins (DEPs), including 327 upregulated and 338 downregulated proteins in the RR‑DTC group, which were enriched in 59 signaling pathways by Kyoto Encyclopedia of Genes and Genomes database analysis. In particular, CHI3L1 was demonstrated to be significantly upregulated in RR‑DTC as evidenced by quantitative proteomic analysis and IHC. Western blotting suggested that the overexpression of CHI3L1 activated the MEK/ERK1/2 signaling pathway, which may lead to NIS dysfunction. In conclusion, the present study suggests that CHI3L1 is a potential molecular target for the radiotherapy of patients with RR‑DTC.
View Figures
View References

Related Articles

Journal Cover

January-2023
Volume 25 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li Y, Hu F, Deng J, Huang X, Zhou C, Wu M and Duan D: Proteomic analysis of radioiodine‑refractory differentiated thyroid cancer identifies CHI3L1 upregulation in association with dysfunction of the sodium‑iodine symporter. Oncol Lett 25: 36, 2023
APA
Li, Y., Hu, F., Deng, J., Huang, X., Zhou, C., Wu, M., & Duan, D. (2023). Proteomic analysis of radioiodine‑refractory differentiated thyroid cancer identifies CHI3L1 upregulation in association with dysfunction of the sodium‑iodine symporter. Oncology Letters, 25, 36. https://doi.org/10.3892/ol.2022.13622
MLA
Li, Y., Hu, F., Deng, J., Huang, X., Zhou, C., Wu, M., Duan, D."Proteomic analysis of radioiodine‑refractory differentiated thyroid cancer identifies CHI3L1 upregulation in association with dysfunction of the sodium‑iodine symporter". Oncology Letters 25.1 (2023): 36.
Chicago
Li, Y., Hu, F., Deng, J., Huang, X., Zhou, C., Wu, M., Duan, D."Proteomic analysis of radioiodine‑refractory differentiated thyroid cancer identifies CHI3L1 upregulation in association with dysfunction of the sodium‑iodine symporter". Oncology Letters 25, no. 1 (2023): 36. https://doi.org/10.3892/ol.2022.13622