Expression of cell-cycle regulators (cyclin D1, cyclin E, p27kip1, p57kip2) in papillary thyroid carcinoma

2010 ◽  
Vol 142 (3) ◽  
pp. 332-337 ◽  
Author(s):  
Sang Hyuk Lee ◽  
Jong Kyu Lee ◽  
Sung Min Jin ◽  
Kyung Chul Lee ◽  
Jin Hee Sohn ◽  
...  
2020 ◽  
Vol 10 (1) ◽  
Author(s):  
Mojgan Sanjari ◽  
Zeinab Kordestani ◽  
Moeinadin Safavi ◽  
Mahdieh Mashrouteh ◽  
Maryam FekriSoofiAbadi ◽  
...  

2015 ◽  
Vol 15 (2) ◽  
pp. 25
Author(s):  
Jae Yeon Seok ◽  
Dong Hae Chung ◽  
Yoo Seung Chung ◽  
Jung Won Ryu ◽  
Young Don Lee

2002 ◽  
Vol 87 (4) ◽  
pp. 1814-1818 ◽  
Author(s):  
Mark L. C. Khoo ◽  
Nigel J. P. Beasley ◽  
Shereen Ezzat ◽  
Jeremy L. Freeman ◽  
Sylvia L. Asa

2007 ◽  
Vol 1299 ◽  
pp. 263-270
Author(s):  
Alexander Yu. Abrosimov ◽  
Olesya A. Shkurko ◽  
Vladimir A. Saenko ◽  
Serik Meirmanov ◽  
Tatiana I. Rogounovitch ◽  
...  

2020 ◽  
Vol 15 (1) ◽  
pp. 561-571
Author(s):  
Yihui Lin ◽  
Jianjia Jiang

AbstractPapillary thyroid carcinoma (PTC) is a common malignancy worldwide. LncRNA LINC00704 (mitotically associated long non-coding RNA) was reported as a crucial regulator in PTC. However, the biological mechanism of LINC00704 action remains unclear in PTC. The mRNA levels of LINC00704, miR-204-5p, and high-mobility group box 1 (HMGB1) were measured by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) assay. HMGB1, proliferating cell nuclear antigen (PCNA), and cyclin D1 protein levels were detected using the Western blot assay. The binding relationship between miR-204-5p and LINC00704 or HMGB1 was predicted by LncBase Predicted v.2 or TargetScan, respectively, and then validated by dual luciferase reporter assay. Cell viability, cell cycle, cell migration and invasion, and migration ratio were assessed by MTT, flow cytometry, transwell cell migration and invasion, and wound-healing assays, respectively. Results suggested that LINC00704 and HMGB1 were elevated and miR-204-5p decreased in PTC tissues and cells. Furthermore, rescue experiments demonstrated that the miR-204-5p inhibitor alleviated the inhibitory effects of LINC00704 knockdown on cell proliferation, cell cycle, migration, and invasion. Meanwhile, miR-204-5p overexpression repressed proliferation, migration, and invasion by targeting HMGB1. Mechanical analysis discovered that LINC00704 could act as an miR-204-5p sponge to modulate HMGB1 expression. In conclusion, LINC00704 promoted PTC cell proliferation, cell cycle, migration, and invasion by the miR-204-5p/HMGB1 axis, providing a novel therapeutic target for PTC patients.


2018 ◽  
Vol 46 (2) ◽  
pp. 579-590 ◽  
Author(s):  
Xiaobin Li ◽  
Zongze Li ◽  
Yimin Song ◽  
Wenjing Liu ◽  
Ziwen Liu

Background/Aim: Mammalian target of rapamycin (mTOR) plays an important role in papillary thyroid carcinoma (PTC) cell progression. CZ415 is a novel, highly-efficient and specific mTOR kinase inhibitor. The current study tested the potential anti-tumor activity of CZ415 in human PTC cells. Methods: The established (TPC-1 cell line) and primary human PTC cells were treated with CZ415. Cell survival and growth were tested by Cell Counting Kit-8 assay and BrdU ELISA assay, respectively. Cell apoptosis was tested by caspase-3/-9 activity assay, Hoechst-33342 staining assay and single-stranded DNA ELISA assay. Cell cycle progression was tested by propidium iodide-FACS assay. The mTOR signaling was tested by Western blotting assay and co-immunoprecipitation assay. The mouse xenograft tumor model was applied to study the effect of CZ415 in vivo. Results: In cultured human PTC cells, treatment with CZ415 at nM concentrations significantly inhibited cell survival and growth. CZ415 induced apoptosis activation and cell cycle arrest in human PTC cells. CZ415 disrupted assembling of mTORC1 (mTOR-Raptor association) and mTORC2 (mTOR-Rictor-GβL association) in TPC-1 cells, which led to de-phosphorylation of the mTORC1 substrates (S6K1 and 4E-BP1) and the mTORC2 substrate AKT (Ser-473). Further studies show that the autophagy inhibitor 3-methyladenine (3-MA) or Beclin-1 shRNA aggravated CZ415-induced cytotoxicity against PTC cells. In vivo, CZ415 oral administration inhibited TPC-1 xenograft tumor growth in mice. Conclusion: Our results show that mTOR blockage by CZ415 inhibits PTC cell growth in vitro and in vivo.


2017 ◽  
Vol 37 (1) ◽  
pp. 76-81
Author(s):  
Nanis S. Holah ◽  
Mohammad I. Shaban ◽  
Shereen F. El-Goday

Sign in / Sign up

Export Citation Format

Share Document