Mathematical Model of a Telomerase Transcriptional Regulatory Network Developed by Cell-Based Screening: Analysis of Inhibitor Effects and Telomerase Expression Mechanisms

作者:Bilsland Alan E*; Stevenson Katrina; Liu Yu; Hoare Stacey; Cairney Claire J; Roffey Jon; Keith W Nicol
来源:PLoS Computational Biology, 2014, 10(2): e1003448.
DOI:10.1371/journal.pcbi.1003448

摘要

Cancer cells depend on transcription of telomerase reverse transcriptase (TERT). Many transcription factors affect TERT, though regulation occurs in context of a broader network. Network effects on telomerase regulation have not been investigated, though deeper understanding of TERT transcription requires a systems view. However, control over individual interactions in complex networks is not easily achievable. Mathematical modelling provides an attractive approach for analysis of complex systems and some models may prove useful in systems pharmacology approaches to drug discovery. In this report, we used transfection screening to test interactions among 14 TERT regulatory transcription factors and their respective promoters in ovarian cancer cells. The results were used to generate a network model of TERT transcription and to implement a dynamic Boolean model whose steady states were analysed. Modelled effects of signal transduction inhibitors successfully predicted TERT repression by Src-family inhibitor SU6656 and lack of repression by ERK inhibitor FR180204, results confirmed by RT-QPCR analysis of endogenous TERT expression in treated cells. Modelled effects of GSK3 inhibitor 6-bromoindirubin-3 '-oxime (BIO) predicted unstable TERT repression dependent on noise and expression of JUN, corresponding with observations from a previous study. MYC expression is critical in TERT activation in the model, consistent with its well known function in endogenous TERT regulation. Loss of MYC caused complete TERT suppression in our model, substantially rescued only by co-suppression of AR. Interestingly expression was easily rescued under modelled Ets-factor gain of function, as occurs in TERT promoter mutation. RNAi targeting AR, JUN, MXD1, SP3, or TP53, showed that AR suppression does rescue endogenous TERT expression following MYC knockdown in these cells and SP3 or TP53 siRNA also cause partial recovery. The model therefore successfully predicted several aspects of TERT regulation including previously unknown mechanisms. An extrapolation suggests that a dominant stimulatory system may programme TERT for transcriptional stability. Author Summary Tumour cells acquire the ability to divide and multiply indefinitely whereas normal cells can undergo only a limited number of divisions. The switch to immortalisation of the tumour cell is dependent on maintaining the integrity of telomere DNA which forms chromosome ends and is achieved through activation of the telomerase enzyme by turning on synthesis of the TERT gene, which is usually silenced in normal cells. Suppressing telomerase is toxic to cancer cells and it is widely believed that understanding TERT regulation could lead to potential cancer therapies. Previous studies have identified many of the factors which individually contribute to activate or repress TERT levels in cancer cells. However, transcription factors do not behave in isolation in cells, but rather as a complex co-operative network displaying inter-regulation. Therefore, full understanding of TERT regulation will require a broader view of the transcriptional network. In this paper we take a computational modelling approach to study TERT regulation at the network level. We tested interactions between 14 TERT-regulatory factors in an ovarian cancer cell line using a screening approach and developed a model to analyse which network interventions were able to silence TERT.

  • 出版日期2014-2