Your browser doesn't support javascript.
loading
Translocated HMGB3 is involved in papillary thyroid cancer progression by activating cytoplasmic TLR3 and transmembrane TREM1.
Zhao, Yang; Lv, Hong-Jun; Deng, Xue-Yang; Chen, Pu; Garstka, Malgorzata A; Shi, Bing-Yin; Fu, Jiao.
Affiliation
  • Zhao Y; Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China.
  • Lv HJ; Department of Endocrinology, Shaanxi Provincial People's Hospital, Xi'an, China.
  • Deng XY; Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China.
  • Chen P; Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China.
  • Garstka MA; Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China.
  • Shi BY; Core Research Laboratory; Department of Endocrinology; National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
  • Fu J; Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China.
Cell Cycle ; 22(23-24): 2584-2601, 2023.
Article in En | MEDLINE | ID: mdl-38197217
ABSTRACT
The family of high mobility group box (HMGB) proteins participates in various biological processes including immunity, inflammation, as well as cancer formation and progression. However, its role in thyroid cancer remains to be clarified. We performed quantitative RT-PCR (qRT-PCR), western blot, enzyme-linked immunosorbent, immunohistochemistry, and immunofluorescence assays to evaluate the expression level and subcellular location of HMGB3. The effects of HMGB3 knockdown on malignant biological behaviors of thyroid cancer were determined by cell proliferation assays, cell cycle and apoptosis assays, and transwell chamber migration and invasion assays. Differential expression genes (DEGs) altered by HMGB3 were analyzed using the Ingenuity Pathway Analysis (IPA) and TRRUST v2 database. HMGB3 correlated pathways predicted by bioinformatic analysis were then confirmed using western blot, co-immunoprecipitation, dual-luciferase reporter assay, and flow cytometry. We found that HMGB3 is overexpressed and its downregulation inhibits cell viability, promotes cell apoptosis and cell cycle arrest, and suppresses cell migration and invasion in thyroid cancer. In PTC, both tissue and serum levels of HMGB3 are elevated and are correlated with lymph node metastasis and advanced tumor stage. Mechanistically, we observed the translocation of HMGB3 in PTC, induced at least partially by hypoxia. Cytoplasmic HMGB3 activates nucleic-acid-mediated TLR3/NF-κB signaling and extracellular HMGB3 interacts with the transmembrane TREM1 receptor in PTC. This study demonstrates the oncogenic role of HMGB3 cytoplasmic and extracellular translocation in papillary thyroid cancers; we recommend its future use as a potential circulating biomarker and therapeutic target for PTC.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Thyroid Neoplasms / HMGB3 Protein / MicroRNAs Limits: Humans Language: En Journal: Cell Cycle Year: 2023 Document type: Article Affiliation country: China Country of publication: Estados Unidos

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Thyroid Neoplasms / HMGB3 Protein / MicroRNAs Limits: Humans Language: En Journal: Cell Cycle Year: 2023 Document type: Article Affiliation country: China Country of publication: Estados Unidos