Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Kirsty Flower is active.

Publication


Featured researches published by Kirsty Flower.


Journal of Virology | 2012

Dynamic Chromatin Environment of Key Lytic Cycle Regulatory Regions of the Epstein-Barr Virus Genome

Sharada Ramasubramanyan; Kay Osborn; Kirsty Flower; Alison J. Sinclair

ABSTRACT The ability of Epstein-Barr virus (EBV) to establish latency allows it to evade the immune system and to persist for the lifetime of its host; one distinguishing characteristic is the lack of transcription of the majority of viral genes. Entry into the lytic cycle is coordinated by the viral transcription factor, Zta (BZLF1, ZEBRA, and EB1), and downstream effectors, while viral genome replication requires the concerted action of Zta and six other viral proteins at the origins of lytic replication. We explored the chromatin context at key EBV lytic cycle promoters (BZLF1, BRLF1, BMRF1, and BALF5) and the origins of lytic replication during latency and lytic replication. We show that a repressive heterochromatin-like environment (trimethylation of histone H3 at lysine 9 [H3K9me3] and lysine 27 [H3K27me3]), which blocks the interaction of some transcription factors with DNA, encompasses the key early lytic regulatory regions. Epigenetic silencing of the EBV genome is also imposed by DNA methylation during latency. The chromatin environment changes during the lytic cycle with activation of histones H3, H4, and H2AX occurring at both the origins of replication and at the key lytic regulatory elements. We propose that Zta is able to reverse the effects of latency-associated repressive chromatin at EBV early lytic promoters by interacting with Zta response elements within the H3K9me3-associated chromatin and demonstrate that these interactions occur in vivo. Since the interaction of Zta with DNA is not inhibited by DNA methylation, it is clear that Zta uses two routes to overcome epigenetic silencing of its genome.


PLOS ONE | 2011

Epigenetic Control of Viral Life-Cycle by a DNA-Methylation Dependent Transcription Factor

Kirsty Flower; David Thomas; James M. Heather; Sharada Ramasubramanyan; Susan Jones; Alison J. Sinclair

Epstein-Barr virus (EBV) encoded transcription factor Zta (BZLF1, ZEBRA, EB1) is the prototype of a class of transcription factor (including C/EBPalpha) that interact with CpG-containing DNA response elements in a methylation-dependent manner. The EBV genome undergoes a biphasic methylation cycle; it is extensively methylated during viral latency but is reset to an unmethylated state following viral lytic replication. Zta is expressed transiently following infection and again during the switch between latency and lytic replication. The requirement for CpG-methylation at critical Zta response elements (ZREs) has been proposed to regulate EBV replication, specifically it could aid the activation of viral lytic gene expression from silenced promoters on the methylated genome during latency in addition to preventing full lytic reactivation from the non-methylated EBV genome immediately following infection. We developed a computational approach to predict the location of ZREs which we experimentally assessed using in vitro and in vivo DNA association assays. A remarkably different binding motif is apparent for the CpG and non-CpG ZREs. Computational prediction of the location of these binding motifs in EBV revealed that the majority of lytic cycle genes have at least one and many have multiple copies of methylation-dependent CpG ZREs within their promoters. This suggests that the abundance of Zta protein coupled with the methylation status of the EBV genome act together to co-ordinate the expression of lytic cycle genes at the majority of EBV promoters.


Journal of General Virology | 2009

The Epstein-Barr virus lytic cycle activator Zta interacts with methylated ZRE in the promoter of host target gene egr1.

James M. Heather; Kirsty Flower; Samine Isaac; Alison J. Sinclair

Activation of the host gene egr1 is essential for the lytic replication of Epstein–Barr virus (EBV). egr1 is activated by Zta (BZLF1, ZEBRA). Zta interacts directly with DNA through a series of closely related Zta-response elements (ZREs). Here we dissect the mechanism used by Zta to interact with the egr1 promoter and identify a weak interaction with egr1ZRE that is dependent on the distal part of egr1ZRE. Furthermore, we demonstrate that the ability of Zta to interact with egr1ZRE is enhanced at least tenfold by methylation. The ability of Zta to transactivate a reporter construct driven by the egr1 promoter can be enhanced by methylation. As the ability of Zta to interact with a methylated ZRE in the EBV genome correlates with its ability to activate the expression of the endogenous viral gene BRLF1, this suggests that Zta may also have the capability to overturn epigenetic control of egr1.


Journal of Virology | 2012

Genome-Wide Analyses of Zta Binding to the Epstein-Barr Virus Genome Reveals Interactions in both Early and Late Lytic Cycles and an Epigenetic Switch Leading to an Altered Binding Profile

Sharada Ramasubramanyan; Aditi Kanhere; Kay Osborn; Kirsty Flower; Richard G. Jenner; Alison J. Sinclair

ABSTRACT The Epstein-Barr virus (EBV) genome sustains substantial epigenetic modification involving chromatin remodelling and DNA methylation during lytic replication. Zta (ZEBRA, BZLF1), a key regulator of the EBV lytic cycle, is a transcription and replication factor, binding to Zta response elements (ZREs) in target promoters and EBV lytic origins of replication. In vitro, Zta binding is modulated by DNA methylation; a subset of CpG-containing Zta binding sites (CpG ZREs) is bound only in a DNA methylation-dependent manner. The question of how the dynamic epigenetic environment impacts Zta interaction during the EBV lytic cycle is unknown. To address this, we used chromatin immunoprecipitation coupled with next-generation sequencing (ChIP-Seq) to identify Zta binding sites across the EBV genome before and after viral DNA replication. Replication did not alter the association of Zta across many regions of the EBV genome, but a striking reduction in Zta binding occurred at some loci that contain CpG ZREs. Separating Zta-bound DNA into methylated and nonmethylated fractions, we found that promoters that contain CpG ZREs were enriched in the methylated fraction but that Zta binding to promoters lacking CpG ZREs was not reduced. We hypothesize that the loss of DNA methylation on the EBV genome during the lytic cycle causes the reduced binding to CpG ZREs; this may act as a lytic cycle epigenetic switch. However, the epigenetic changes associated with the replicated EBV genome do not affect the interaction of Zta with many loci that are rich in non-CpG ZREs; this leads to sustained binding at these regions.


Carcinogenesis | 2016

Stilbenoids remodel the DNA methylation patterns in breast cancer cells and inhibit oncogenic NOTCH signaling through epigenetic regulation of MAML2 transcriptional activity

Katarzyna Lubecka; Lucinda Kurzava; Kirsty Flower; Hannah Buvala; Hao Zhang; Dorothy Teegarden; Ignacio G. Camarillo; Matthew Suderman; Shihuan Kuang; Ourania M. Andrisani; James M. Flanagan; Barbara Stefanska

Summary We established the role for epigenetics in regulation of NOTCH signaling in breast cancer. This may constitute a common mechanism of activation of oncogenic signals. Our study provides support for epigenetic-targeting strategies in anticancer approaches.


Journal of Virology | 2007

Atypical bZIP Domain of Viral Transcription Factor Contributes to Stability of Dimer Formation and Transcriptional Function

Celine Schelcher; Salama Al Mehairi; Elizabeth Verrall; Questa Hope; Kirsty Flower; Beth Bromley; Derek N. Woolfson; Michelle J. West; Alison J. Sinclair

ABSTRACT The Epstein-Barr virus transcription factor Zta (encoded by BZLF1) is a bZIP protein containing an α-helical coiled-coil homodimerization motif (zipper). The Zta zipper forms less-stable dimers than other bZIP proteins, and an adjacent region (CT) interacts with the zipper to form a novel structure that is proposed to strengthen the dimer. Here we question the role of the CT region for Zta function. Cross-linking experiments demonstrate that the entire CT region lies adjacent to the zipper. Detailed analyses of Zta truncation mutations identify an involvement of the proximal CT region (221 to 230) in dimer formation with a further contribution from the distal region (236 to 243). Biophysical analyses reveal that residues 221 to 230 enhance the stability of the coiled coil. The ability of the Zta truncation mutants to interact with three Zta-binding sites also requires the proximal CT region. Fine mapping of DNA-binding requirements highlighted the contribution of these amino acids for Zta function. Thus, the proximal part of the CT region is required to aid the dimerization of Zta and thereby its DNA-binding ability. In contrast, although the distal part of the CT region aids dimerization, it promotes only a modest increase in DNA binding. To probe this further, we defined the contribution from the CT region for Zta to transactivate a promoter embedded within the viral genome. From this we conclude that the proximal part of the CT region is absolutely required, whereas the distal part is dispensable.


PLOS ONE | 2010

Evaluation of a Prediction Protocol to Identify Potential Targets of Epigenetic Reprogramming by the Cancer Associated Epstein Barr Virus

Kirsty Flower; Elizabeth Hellen; Melanie J. Newport; Susan Jones; Alison J. Sinclair

Background Epstein Barr virus (EBV) infects the majority of the human population, causing fatal diseases in a small proportion in conjunction with environmental factors. Following primary infection, EBV remains latent in the memory B cell population for life. Recurrent reactivation of the virus occurs, probably due to activation of the memory B-lymphocytes, resulting in viral replication and re-infection of B-lymphocytes. Methylation of the viral DNA at CpG motifs leads to silencing of viral gene expression during latency. Zta, the key viral protein that mediates the latency/reactivation balance, interacts with methylated DNA. Zta is a transcription factor for both viral and host genes. A sub-set of its DNA binding sites (ZREs) contains a CpG motif, which is recognised in its methylated form. Detailed analysis of the promoter of the viral gene BRLF1 revealed that interaction with a methylated CpG ZRE (RpZRE3) is key to overturning the epigenetic silencing of the gene. Methodology and Principal Findings Here we question whether we can use this information to identify which host genes contain promoters with similar response elements. A computational search of human gene promoters identified 274 targets containing the 7-nucleotide RpZRE3 core element. DNA binding analysis of Zta with 17 of these targets revealed that the flanking context of the core element does not have a profound effect on the ability of Zta to interact with the methylated sites. A second juxtaposed ZRE was observed for one promoter. Zta was able to interact with this site, although co-occupancy with the RpZRE3 core element was not observed. Conclusions/Significance This research demonstrates 274 human promoters have the potential to be regulated by Zta to overturn epigenetic silencing of gene expression during viral reactivation from latency.


Current Cancer Drug Targets | 2017

Is there a Role for Epigenetic Enhancement of Immunomodulatory Approaches to Cancer Treatment

Kirsty Flower; Sadaf Ghaem-Maghami; Robert Brown

The efficacy of cancer immunotherapy relies on the ability of the host immune system to recognise the cancer as non-self and eliminate it from the body. Whilst this is an extremely fertile area of medical research, with positive clinical trials showing durable responses, attention must be paid to the subset of patients that do not respond to these treatments. Immune surveillance and immunoediting by the host could itself select for immune-evasive tumour cells during tumour development leading to immunotherapy resistance. One such mechanism of non-efficacy or resistance is the epigenetic silencing of a specific gene required in the immunotherapy response pathway. Epigenetics is the study of the control of expression patterns in a cell via mechanisms not involving a change in DNA sequence. All tumour types show aberrant epigenetic regulation of genes involved in all the hallmarks of cancer, including immunomodulation. Inhibition of key enzymes involved in maintenance of epigenetic states is another important area of research for new treatment strategies for cancer. Could epigenetic therapies be used to successfully enhance the action of immunomodulatory agents in cancer, and are they acting in the way we imagine? An understanding of the effects of epigenetic therapies on immunological pathways in both the tumour and host cells, especially the tumour microenvironment, will be essential to further develop such combination approaches.


Clinical Cancer Research | 2017

Clinical use of programmed cell death-1 (PD-1) and its ligand (PD-L1) expression as discriminatory and predictive markers in ovarian cancer

Jayanta Chatterjee; Wei Dai; Nor Haslinda Abd Aziz; Pei Yun Teo; John Wahba; David L. Phelps; Christian J. Maine; Lynsey M. Whilding; Roberto Dina; Giorgia Trevisan; Kirsty Flower; Andrew J. T. George; Sadaf Ghaem-Maghami

Purpose: We aimed to establish whether programmed cell death-1 (PD-1) and programmed cell death ligand 1 (PD-L1) expression, in ovarian cancer tumor tissue and blood, could be used as biomarkers for discrimination of tumor histology and prognosis of ovarian cancer. Experimental Design: Immune cells were separated from blood, ascites, and tumor tissue obtained from women with suspected ovarian cancer and studied for the differential expression of possible immune biomarkers using flow cytometry. PD-L1 expression on tumor-associated inflammatory cells was assessed by immunohistochemistry and tissue microarray. Plasma soluble PD-L1 was measured using sandwich ELISA. The relationships among immune markers were explored using hierarchical cluster analyses. Results: Biomarkers from the discovery cohort that associated with PD-L1+ cells were found. PD-L1+ CD14+ cells and PD-L1+ CD11c+ cells in the monocyte gate showed a distinct expression pattern when comparing benign tumors and epithelial ovarian cancers (EOCs)—confirmed in the validation cohort. Receiver operating characteristic curves showed PD-L1+ and PD-L1+ CD14+ cells in the monocyte gate performed better than the well-established tumor marker CA-125 alone. Plasma soluble PD-L1 was elevated in patients with EOC compared with healthy women and patients with benign ovarian tumors. Low total PD-1+ expression on lymphocytes was associated with improved survival. Conclusions: Differential expression of immunological markers relating to the PD-1/PD-L1 pathway in blood can be used as potential diagnostic and prognostic markers in EOC. These data have implications for the development and trial of anti–PD-1/PD-L1 therapy in ovarian cancer. Clin Cancer Res; 23(13); 3453–60. ©2016 AACR.


Cancer Research | 2016

Abstract 3154: Cell-free circulating tumor DNA methylation in high-grade serous ovarian cancer

Kayleigh R. Davis; Kirsty Flower; Jane Borley; Charlotte Wilhelm-Benartzi; Robert Brown

Background Ovarian cancer is often diagnosed late leading to poor patient survival. Over two thirds of women with ovarian cancer are diagnosed with Stages III/IV where the relative 5year survival is 90% for Stage I. New strategies for early detection and patient stratification are urgently required, as well as non-invasive ways of monitoring tumour evolution. Circulating tumour DNA (ctDNA) can be detected in plasma from ovarian cancer patients. Genome-wide methylation analysis of tissue has identified tumour specific changes in DNA methylation which could be present in ctDNA. However, lysis of peripheral blood mononuclear cells (PBMCs) can release DNA into the plasma confounding detection of tumour DNA. If specific methylation changes that differentiate tumour DNA from PBMC DNA are identified, this would allow DNA methylation in ctDNA to be more accurately quantified. Methods In this study, publically available genome-wide array-based DNA methylation datasets (Cancer Genome Atlas Research, 2011) (Fridley et al., 2014) were interrogated to identify regions that are highly methylated in high-grade serous ovarian carcinoma, (HGSOC), and hypomethylated in PBMCs and vice versa. The methylation status of these loci was then confirmed in an independent dataset of HGSOC and benign tissue. Sodium heparin blood samples were collected from healthy volunteers and patients suspected of having HGSOC at Hammersmith Hospital, London. Blood samples were separated by low speed followed by high speed centrifugation, DNA extracted and analysed using bisulfite pyrosequencing. Results Ten loci were identified from the publically available data that had >80% methylation in HGSOC (n = 342) as well as 80% methylation in PBMCs. Seven of these have been independently validated as differentially methylated by bisulphite pyrosequencing in HGSOC versus PBMCs from the Hammersmith patient cohort. Methylation can be detected using pyrosequencing using DNA amounts less than 10ng. For three loci analysed so far, we have shown significant differences in methylation between DNA isolated from plasma of ovarian cancer patients compared to healthy volunteers. One locus was capable of distinguishing ovarian cancer patients from healthy volunteers with 100% sensitivity and specificity. Conclusions We have identified differentially methylated loci between HGSOC and PBMCs with high sensitivity and specificity. We have shown that these epigenetic changes can be detected in plasma ctDNA from patients with ovarian cancer. References Cancer Genome Atlas Research Network., Integrated genomic analyses of ovarian carcinoma. Nature 2011, 474:7353 Fridley et al., Methylation of leukocyte DNA and ovarian cancer: relationships with disease status and outcome. BMC Med Genomics 2014, 7:21 Citation Format: Kayleigh R. Davis, Kirsty J. Flower, Jane V. Borley, Charlotte SM Wilhelm-Benartzi, Robert Brown. Cell-free circulating tumor DNA methylation in high-grade serous ovarian cancer. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3154.

Collaboration


Dive into the Kirsty Flower's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert Brown

Imperial College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge