HMGB1-mediated autophagy regulates sodium/iodide symporter protein degradation in thyroid cancer cells

J Exp Clin Cancer Res. 2019 Jul 22;38(1):325. doi: 10.1186/s13046-019-1328-3.

Abstract

Background: Sodium/iodide symporter (NIS)-mediated iodide uptake plays an important physiological role in regulating thyroid gland function, as well as in diagnosing and treating Graves' disease and thyroid cancer. High-mobility group box 1 (HMGB1), a highly conserved nuclear protein, is a positive regulator of autophagy conferring resistance to chemotherapy, radiotherapy and immunotherapy in cancer cells. Here the authors intended to identify the role of HMGB1 in Hank's balanced salt solution (HBSS)-induced autophagy, explore NIS protein degradation through a autophagy-lysosome pathway in thyroid cancer cells and elucidate the possible molecular mechanisms.

Methods: Immunohistochemical staining and reverse transcription-polymerase chain reaction (RT-PCR) were performed for detecting the expression of HMGB1 in different tissues. HMGB1 was knocked down by lentiviral transfection in FTC-133/TPC-1 cells. Autophagic markers LC3-II, p62, Beclin1 and autophagosomal formation were employed for evaluating HMGB1-mediated autophagy in HBSS-treated cells by Western blot, immunofluorescence and electron microscopy. Western blot, quantitative RT-PCR and gamma counter analysis were performed for detecting NIS expression and iodide uptake in HMGB1-knockdown cells after different treatments. The reactive oxygen species (ROS) level, ROS-mediated LC3-II expression and HMGB1 cytosolic translocation were detected by fluorospectrophotometer, flow cytometry, Western blot and immunofluorescence. HMGB1-mediated AMPK, mTOR and p70S6K phosphorylation (p-AMPK, p-mTOR & p-p70S6K) were detected by Western blot. Furthermore, a nude murine model with transplanted tumor was employed for examining the effect of HMGB1-mediated autophagy on imaging and biodistribution of 99mTcO4-. NIS, Beclin1, p-AMPK and p-mTOR were detected by immunohistochemical staining and Western blot in transplanted tumor samples.

Results: HMGB1 was a critical regulator of autophagy-mediated NIS degradation in HBSS-treated FTC-133/TPC-1 cells. And HMGB1 up-regulation was rather prevalent in thyroid cancer tissues and closely correlated with worse overall lymph node metastasis and clinical stage. HMGB1-knockdown dramatically suppressed autophagy, NIS degradation and boosted iodide uptake in HBSS-treated cells. Moreover, HBSS enhanced ROS-sustained autophagy and promoted the cytosolic translocation of HMGB1. A knockdown of HMGB1 suppressed LC3-II conversion and NIS degradation via an AMPK/mTOR-dependent signal pathway through a regulation of ROS generation, rather than ATP. Furthermore, these data were further supported by our in vivo experiment of xenografts formed by HMGB1 knockdown cells reverting the uptake of 99mTcO4- as compared with control shRNA-transfected cells in hunger group.

Conclusions: Acting as a critical regulator of autophagy-mediated NIS degradation via ROS/AMPK/mTOR pathway, HMGB1is a potential intervention target of radioiodine therapy in thyroid cancer.

Keywords: AMPK; Autophagy; HMGB1; NIS; mTOR.

MeSH terms

  • Animals
  • Autophagosomes / drug effects
  • Autophagy / genetics*
  • Autophagy / radiation effects
  • Cell Line, Tumor
  • Gene Expression Regulation, Neoplastic / radiation effects
  • Gene Knockdown Techniques
  • HMGB1 Protein / genetics*
  • Heterografts
  • Humans
  • Iodine Radioisotopes / metabolism
  • Iodine Radioisotopes / pharmacology*
  • Mice
  • Microtubule-Associated Proteins / genetics
  • Proteolysis / radiation effects
  • Reactive Oxygen Species / metabolism
  • Symporters / chemistry
  • Symporters / genetics*
  • Thyroid Gland / radiation effects
  • Thyroid Neoplasms / genetics
  • Thyroid Neoplasms / pathology
  • Thyroid Neoplasms / radiotherapy*

Substances

  • HMGB1 Protein
  • HMGB1 protein, human
  • Iodine Radioisotopes
  • MAP1LC3B protein, human
  • Microtubule-Associated Proteins
  • Reactive Oxygen Species
  • Symporters
  • sodium-iodide symporter