期刊论文详细信息
Molecular Cancer
Hypoxia inducible prolyl hydroxylase PHD3 maintains carcinoma cell growth by decreasing the stability of p27
Research
Petra Miikkulainen1  Heidi Högel1  Panu M. Jaakkola2  Lucia Bino3 
[1] Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Tykistökatu 6B, 20520, Turku, Finland;Department of Medical Biochemistry, Faculty of Medicine, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland;Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Tykistökatu 6B, 20520, Turku, Finland;Department of Medical Biochemistry, Faculty of Medicine, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland;Department of Oncology and Radiotherapy, Turku University Hospital, Hämeentie 11, 20520, Turku, Finland;Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Tykistökatu 6B, 20520, Turku, Finland;Present address: Institute of Biophysics, The Academy of Sciences of the Czech Republic, Brno, Czech Republic;
关键词: CDKN1B;    Cell cycle;    EGLN;    HIF;    Hypoxia;    VHL;   
DOI  :  10.1186/s12943-015-0410-5
 received in 2014-09-19, accepted in 2015-07-09,  发布年份 2015
来源: Springer
PDF
【 摘 要 】

BackgroundHypoxia can halt cell cycle progression of several cell types at the G1/S interface. The arrest needs to be overcome by cancer cells. We have previously shown that the hypoxia-inducible cellular oxygen sensor PHD3/EGLN3 enhances hypoxic cell cycle entry at the G1/S boundary.MethodsWe used PHD3 knockdown by siRNA and shRNA in HeLa and 786–0 renal cancer cells. Flow cytometry with cell synchronization was used to study cell growth at different cell cycle phases. Total and phosphospecific antibodies together with cycloheximide chase were used to study p27/CDKN1B expression and fractionations for subcellular protein localization.ResultsHere we show that PHD3 enhances cell cycle by decreasing the expression of the CDK inhibitor p27/CDKN1B. PHD3 reduction led to increased p27 expression under hypoxia or VHL mutation. p27 was both required and sufficient for the PHD3 knockdown induced cell cycle block. PHD3 knockdown did not affect p27 transcription and the effect was HIF-independent. In contrast, PHD3 depletion increased the p27 half-life from G0 to S-phase. PHD3 depletion led to an increase in p27 phosphorylation at serine 10 without affecting threonine phosphorylation. Intact serine 10 was required for normal hypoxic and PHD3-mediated degradation of p27.ConclusionsThe data demonstrates that PHD3 can drive cell cycle entry at the G1/S transition through decreasing the half-life of p27 that occurs by attenuating p27S10 phosphorylation.

【 授权许可】

Unknown   
© Högel et al. 2015. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

【 预 览 】
附件列表
Files Size Format View
RO202311105457812ZK.pdf 2539KB PDF download
【 参考文献 】
  • [1]
  • [2]
  • [3]
  • [4]
  • [5]
  • [6]
  • [7]
  • [8]
  • [9]
  • [10]
  • [11]
  • [12]
  • [13]
  • [14]
  • [15]
  • [16]
  • [17]
  • [18]
  • [19]
  • [20]
  • [21]
  • [22]
  • [23]
  • [24]
  • [25]
  • [26]
  • [27]
  • [28]
  • [29]
  • [30]
  • [31]
  • [32]
  • [33]
  • [34]
  • [35]
  • [36]
  • [37]
  • [38]
  • [39]
  • [40]
  • [41]
  • [42]
  • [43]
  • [44]
  • [45]
  • [46]
  • [47]
  • [48]
  • [49]
  • [50]
  • [51]
  • [52]
  • [53]
  • [54]
  • [55]
  • [56]
  • [57]
  • [58]
  • [59]
  • [60]
  • [61]
  • [62]
  • [63]
  • [64]
  • [65]
  文献评价指标  
  下载次数:0次 浏览次数:0次