Your browser doesn't support javascript.
loading
Death-Associated Protein Kinase 1 Inhibits Progression of Thyroid Cancer by Regulating Stem Cell Markers.
You, Mi-Hyeon; Lee, Woo Kyung; Jin, Meihua; Song, Dong Eun; Cheng, Sheue-Yann; Kim, Tae Yong; Kim, Won Bae; Jeon, Min Ji; Kim, Won Gu.
Affiliation
  • You MH; Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea.
  • Lee WK; Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
  • Jin M; Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea.
  • Song DE; Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea.
  • Cheng SY; Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
  • Kim TY; Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea.
  • Kim WB; Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea.
  • Jeon MJ; Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea.
  • Kim WG; Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea.
Cells ; 10(11)2021 11 03.
Article de En | MEDLINE | ID: mdl-34831219
ABSTRACT
The activation of metastatic reprogramming is vital for cancer metastasis, but little is known about its mechanism. This study investigated the potential role of death-associated protein kinase 1 (DAPK1) in thyroid cancer progression. We generated knockdown (KD) DAPK1 using siRNA or shRNA in 8505C and KTC-1 cell lines, which we transiently or stably overexpressed in MDA-T32 and BCPAP cell lines. DAPK1 KD in 8505C and KTC-1 cells significantly increased cell proliferation and colony formation compared with controls. We observed significant inhibition of cancer cell invasion in cells overexpressing DAPK1, but the opposite effect in KD cells. Tumorsphere formation significantly increased after inhibition of DAPK1 expression in 8505C cells and was significantly suppressed in DAPK1-overexpressing MDA-T32 and BCPAP cells. DAPK1 overexpression inhibited mRNA and protein levels of stem markers (OCT4, Sox2, KLF4, and Nanog). Furthermore, the expression of these markers increased after KD of DAPK1 in 8505C cells. Mechanistic studies suggest that DAPK1 may modulate the expression of stem cell markers through the inhibition of ß-catenin pathways. These findings were consistent with the public data and our thyroid tissue analysis, which showed higher DAPK1 expression was associated with advanced-stage papillary thyroid cancer with a higher stemness index and lower disease-free survival.
Sujet(s)
Mots clés

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Sujet principal: Cellules souches tumorales / Tumeurs de la thyroïde / Marqueurs biologiques tumoraux / Évolution de la maladie / Death-associated protein kinases Type d'étude: Risk_factors_studies Limites: Humans Langue: En Journal: Cells Année: 2021 Type de document: Article

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Sujet principal: Cellules souches tumorales / Tumeurs de la thyroïde / Marqueurs biologiques tumoraux / Évolution de la maladie / Death-associated protein kinases Type d'étude: Risk_factors_studies Limites: Humans Langue: En Journal: Cells Année: 2021 Type de document: Article