Network


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

Hotspot


Dive into the research topics where Hayley D. McKeen is active.

Publication


Featured researches published by Hayley D. McKeen.


Endocrinology | 2008

A Novel FK506-Like Binding Protein Interacts with the Glucocorticoid Receptor and Regulates Steroid Receptor Signaling

Hayley D. McKeen; Kerry McAlpine; Andrea Valentine; Derek J. Quinn; Keeva McClelland; Christopher Byrne; Martin O'Rourke; Sheila Young; Christopher J. Scott; H. McCarthy; David Hirst; Tracy Robson

FKBP-like (FKBPL) protein is a novel immunophilin-like protein that plays a role in the cellular stress response. Its three tetratricopeptide repeat motifs are homologous to the heat shock protein 90 interaction sites of other immunophilins that have roles in steroid hormone receptor signaling. In this study, using biomolecular complementation and coimmunoprecipitation techniques, we show that FKBPL also colocalizes and interacts with the components of the heat shock protein 90-glucocorticoid receptor (GR) complex and demonstrate that the PPIase domain of FKBPL is important for the interaction between this complex and the dynein motor protein, dynamitin. Treatment of DU145 cells with the GR ligand, dexamethasone, induced a rapid and coordinated translocation of both GR and FKBPL to the nucleus; this response was perturbed when FKBPL was knocked down with a targeted small interfering RNA. Furthermore, overexpression of FKBPL increased GR protein levels and transactivation of a luciferase reporter gene in response to dexamethasone in DU145 cells. However, these responses were cell line dependent. In summary, these data suggest that FKBPL can be classed as a new member of the FKBP protein family with a role in steroid receptor complexes and signaling.


Cancer Research | 2009

FKBPL Regulates Estrogen Receptor Signaling and Determines Response to Endocrine Therapy

Hayley D. McKeen; Christopher Byrne; Puthen V. Jithesh; Christopher Donley; Andrea Valentine; Anita Yakkundi; Martin O'Rourke; Charles Swanton; H. McCarthy; David Hirst; Tracy Robson

The HSP90 chaperone and immunophilin FKBPL is an estrogen-responsive gene that interacts with estogen receptor alpha (ERalpha) and regulates its levels. In this study, we explored the effects of FKBPL on breast cancer proliferation. Breast cancer cells stably overexpressing FKBPL became dependent on estrogen for their growth and were dramatically more sensitive to the antiestrogens tamoxifen and fulvestrant, whereas FKBPL knockdown reverses this phenotype. FKBPL knockdown also decreased the levels of the cell cycle inhibitor p21WAF1 and increased ERalpha phosphorylation on Ser(118) in response to 17beta-estradiol and tamoxifen. In support of the likelihood that these effects explained FKBPL-mediated cell growth inhibition and sensitivity to endocrine therapies, FKBPL expression was correlated with increased overall survival and distant metastasis-free survival in breast cancer patients. Our findings suggest that FKBPL may have prognostic value based on its impact on tumor proliferative capacity and sensitivity to endocrine therapies, which improve outcome.


Clinical Cancer Research | 2011

FKBPL and Peptide Derivatives: Novel Biological Agents That Inhibit Angiogenesis by a CD44-Dependent Mechanism

Andrea Valentine; Martin O'Rourke; Anita Yakkundi; Jenny Worthington; Michelle Hookham; Roy Bicknell; H. McCarthy; Keeva McClelland; Lynn McCallum; Hayder Dyer; Hayley D. McKeen; David Waugh; Jennifer Roberts; Joanne McGregor; Graham Cotton; Iain James; David Hirst; Tracy Robson

Purpose: Antiangiogenic therapies can be an important adjunct to the management of many malignancies. Here we investigated a novel protein, FKBPL, and peptide derivative for their antiangiogenic activity and mechanism of action. Experimental Design: Recombinant FKBPL (rFKBPL) and its peptide derivative were assessed in a range of human microvascular endothelial cell (HMEC-1) assays in vitro. Their ability to inhibit proliferation, migration, and Matrigel-dependent tubule formation was determined. They were further evaluated in an ex vivo rat model of neovascularization and in two in vivo mouse models of angiogenesis, that is, the sponge implantation and the intravital microscopy models. Antitumor efficacy was determined in two human tumor xenograft models grown in severe compromised immunodeficient (SCID) mice. Finally, the dependence of peptide on CD44 was determined using a CD44-targeted siRNA approach or in cell lines of differing CD44 status. Results: rFKBPL inhibited endothelial cell migration, tubule formation, and microvessel formation in vitro and in vivo. The region responsible for FKBPLs antiangiogenic activity was identified, and a 24-amino acid peptide (AD-01) spanning this sequence was synthesized. It was potently antiangiogenic and inhibited growth in two human tumor xenograft models (DU145 and MDA-231) when administered systemically, either on its own or in combination with docetaxel. The antiangiogenic activity of FKBPL and AD-01 was dependent on the cell-surface receptor CD44, and signaling downstream of this receptor promoted an antimigratory phenotype. Conclusion: FKBPL and its peptide derivative AD-01 have potent antiangiogenic activity. Thus, these agents offer the potential of an attractive new approach to antiangiogenic therapy. Clin Cancer Res; 17(5); 1044–56. ©2011 AACR.


Biochemical Society Transactions | 2011

The emerging role of FK506-binding proteins as cancer biomarkers: a focus on FKBPL

Hayley D. McKeen; Donal J. Brennan; Shauna Hegarty; Fiona Lanigan; Karin Jirström; Christopher Byrne; Anita Yakkundi; Helen O. McCarthy; William M. Gallagher; Tracy Robson

FKBPs (FK506-binding proteins) have long been recognized as key regulators of the response to immunosuppressant drugs and as co-chaperones of steroid receptor complexes. More recently, evidence has emerged suggesting that this diverse protein family may also represent cancer biomarkers owing to their roles in cancer progression and response to treatment. FKBPL (FKBP-like) is a novel FKBP with roles in GR (glucocorticoid receptor), AR (androgen receptor) and ER (oestrogen receptor) signalling. FKBPL binds Hsp90 (heat-shock protein 90) and modulates translocation, transcriptional activation and phosphorylation of these steroid receptors. It has been proposed as a novel prognostic and predictive biomarker, where high levels predict for increased recurrence-free survival in breast cancer patients and enhanced sensitivity to endocrine therapy. Since this protein family has roles in a plethora of signalling pathways, its members represent novel prognostic markers and therapeutic targets for cancer diagnosis and treatment.


Clinical Cancer Research | 2013

Targeting treatment-resistant breast cancer stem cells with FKBPL and its peptide derivative, AD-01, via the CD44 pathway.

Lana McClements; Anita Yakkundi; Angelos Papaspyropoulos; Hannah Harrison; Matthew P. Ablett; Puthen V. Jithesh; Hayley D. McKeen; Rachel Bennett; Christopher Donley; Adrien Kissenpfennig; Stuart McIntosh; Helen O. McCarthy; Eric O'Neill; Robert B. Clarke; Tracy Robson

Purpose: FK506-binding protein like (FKBPL) and its peptide derivative, AD-01, have already shown tumor growth inhibition and CD44-dependent antiangiogenic activity. Here, we explore the ability of AD-01 to target CD44-positive breast cancer stem cells (BCSC). Experimental Design: Mammosphere assays and flow cytometry were used to analyze the effect of FKBPL overexpression/knockdown and AD-01 treatment ± other anticancer agents on BCSCs using breast cancer cell lines (MCF-7/MDA-231/ZR-75), primary patient samples, and xenografts. Delays in tumor initiation were evaluated in vivo. The anti–stem cell mechanisms were determined using clonogenic assays, quantitative PCR (qPCR), and immunofluorescence. Results: AD-01 treatment was highly effective at inhibiting the BCSC population by reducing mammosphere-forming efficiency and ESA+/CD44+/CD24− or aldehyde dehydrogenase (ALDH)+ cell subpopulations in vitro and tumor initiation in vivo. The ability of AD-01 to inhibit the self-renewal capacity of BCSCs was confirmed; mammospheres were completely eradicated by the third generation. The mechanism seems to be due to AD-01–mediated BCSC differentiation shown by a significant decrease in the number of holoclones and an associated increase in meroclones/paraclones; the stem cell markers, Nanog, Oct4, and Sox2, were also significantly reduced. Furthermore, we showed additive inhibitory effects when AD-01 was combined with the Notch inhibitor, DAPT. AD-01 was also able to abrogate a chemo- and radiotherapy-induced enrichment in BCSCs. Finally, FKBPL knockdown led to an increase in Nanog/Oct4/Sox2 and an increase in BCSCs, highlighting a role for endogenous FKBPL in stem cell signaling. Conclusions: AD-01 has dual antiangiogenic and anti-BCSC activity, which will be advantageous as this agent enters clinical trial. Clin Cancer Res; 19(14); 3881–93. ©2013 AACR.


Oncogene | 2014

Identification of RBCK1 as a novel regulator of FKBPL: implications for tumor growth and response to tamoxifen

Christopher Donley; Keeva McClelland; Hayley D. McKeen; Laura D. Nelson; Anita Yakkundi; Puthen V. Jithesh; James F. Burrows; Lana McClements; Andrea Valentine; Kevin Prise; Helen O. McCarthy; Tracy Robson

FKBPL has been implicated in processes associated with cancer, including regulation of tumor growth and angiogenesis with high levels of FKBPL prognosticating for improved patient survival. Understanding how FKBPL levels are controlled within the cell is therefore critical. We have identified a novel role for RBCK1 as an FKBPL-interacting protein, which regulates FKBPL stability at the post-translational level via ubiquitination. Both RBCK1 and FKBPL are upregulated by 17-β-estradiol and interact within heat shock protein 90 chaperone complexes, together with estrogen receptor-α (ERα). Furthermore, FKBPL and RBCK1 associate with ERα at the promoter of the estrogen responsive gene, pS2, and regulate pS2 levels. MCF-7 clones stably overexpressing RBCK1 were shown to have reduced proliferation and increased levels of FKBPL and p21. Furthermore, these clones were resistant to tamoxifen therapy, suggesting that RBCK1 could be a predictive marker of response to endocrine therapy. RBCK1 knockdown using targeted small interfering RNA resulted in increased proliferation and increased sensitivity to tamoxifen treatment. Moreover, in support of our in vitro data, analysis of mRNA microarray data sets demonstrated that high levels of FKBPL and RBCK1 correlated with increased patient survival, whereas high RBCK1 predicted for a poor response to tamoxifen. Our findings support a role for RBCK1 in the regulation of FKBPL with important implications for estrogen receptor signaling, cell proliferation and response to endocrine therapy.


Nanomedicine: Nanotechnology, Biology and Medicine | 2015

RALA-mediated delivery of FKBPL nucleic acid therapeutics

Rachel Bennett; Anita Yakkundi; Hayley D. McKeen; Lana McClements; Thomas J McKeogh; Cian M. McCrudden; Kenneth Arthur; Tracy Robson; Helen O. McCarthy

AIMS RALA is a novel 30 mer bioinspired amphipathic peptide that is showing promise for gene delivery. Here, we used RALA to deliver the FK506-binding protein like - FKBPL gene (pFKBPL) - a novel member of the immunophilin protein family. FKBPL is a secreted protein, with overexpression shown to inhibit angiogenesis, tumor growth and stemness, through a variety of intra- and extracellular signaling mechanisms. We also elucidated proangiogenic activity and stemness after utilizing RALA to deliver siRNA (siFKBPL). MATERIALS & METHODS The RALA/pFKBPL and RALA/siFKBPL nanoparticles were characterized in terms of size, charge, stability and toxicity. Overexpression and knockdown of FKBPL was assessed in vitro and in vivo. RESULTS RALA delivered both pFKBPL and siFKBPL with less cytotoxicity than commercially available counterparts. In vivo, RALA/pFKBPL delivery retarded tumor growth, and prolonged survival with an associated decrease in angiogenesis, while RALA/siFKBPL had no effect on tumor growth rate or survival, but resulted in an increase in angiogenesis and stemness. CONCLUSION RALA is an effective delivery system for both FKBPL DNA and RNAi and highlights an alternative therapeutic approach to harnessing FKBPLs antiangiogenic and antistemness activity.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2015

FKBPL Is a Critical Antiangiogenic Regulator of Developmental and Pathological Angiogenesis

Anita Yakkundi; Rachel Bennett; Ivette Hernández-Negrete; Jean-Marie Delalande; Mary L. Hanna; Oksana Lyubomska; Kenneth Arthur; Amy Short; Hayley D. McKeen; Laura D. Nelson; Cian M. McCrudden; Ross McNally; Lana McClements; Helen O. McCarthy; Alan J. Burns; Roy Bicknell; Adrien Kissenpfennig; Tracy Robson

Objective— The antitumor effects of FK506-binding protein like (FKBPL) and its extracellular role in angiogenesis are well characterized; however, its role in physiological/developmental angiogenesis and the effect of FKBPL ablation has not been evaluated. This is important as effects of some angiogenic proteins are dosage dependent. Here we evaluate the regulation of FKBPL secretion under angiogenic stimuli, as well as the effect of FKBPL ablation in angiogenesis using mouse and zebrafish models. Approach and Results— FKBPL is secreted maximally by human microvascular endothelial cells and fibroblasts, and this was specifically downregulated by proangiogenic hypoxic signals, but not by the angiogenic cytokines, VEGF or IL8. FKBPL’s critical role in angiogenesis was supported by our inability to generate an Fkbpl knockout mouse, with embryonic lethality occurring before E8.5. However, whilst Fkbpl heterozygotic embryos showed some vasculature irregularities, the mice developed normally. In murine angiogenesis models, including the ex vivo aortic ring assay, in vivo sponge assay, and tumor growth assay, Fkbpl +/− mice exhibited increased sprouting, enhanced vessel recruitment, and faster tumor growth, respectively, supporting the antiangiogenic function of FKBPL. In zebrafish, knockdown of zFkbpl using morpholinos disrupted the vasculature, and the phenotype was rescued with hFKBPL. Interestingly, this vessel disruption was ineffective when zcd44 was knocked-down, supporting the dependency of zFkbpl on zCd44 in zebrafish. Conclusions— FKBPL is an important regulator of angiogenesis, having an essential role in murine and zebrafish blood vessel development. Mouse models of angiogenesis demonstrated a proangiogenic phenotype in Fkbpl heterozygotes.


Molecular Carcinogenesis | 2009

Cytochrome P450 1B1 expression in rat esophageal tumorigenesis promoted by gastric and duodenal reflux

Andrea H. Devlin; Marie McIlroy; Hayley D. McKeen; Pramode Bonde; A.A. Carlos Menezes; Christine Swarbrick; Tracy Robson; David Hirst; F. Charles Campbell; James McGuigan; Stephanie R. McKeown

Cytochrome P450 1B1 (CYP1B1) mRNA is constitutively expressed in most normal extra‐hepatic tissues; however the protein is not detectable in these tissues but is expressed in a wide variety of tumors. CYP1B1 is responsible for the activation of a number of carcinogens present in tobacco smoke and food. A surgical model of rat esophageal tumorigenesis, promoted by gastric or duodenal reflux was used to determine CYP1B1 expression in premalignant esophageal tissue. Immunohistochemistry was performed using a modified amplified fluorescein tyramide protocol. CYP1B1 was not observed in normal esophageal mucosa, submucosa, or muscularis mucosa. Animals exposed to gastric reflux developed mild hyperplasia. Varying degrees of hyperplasia were observed in the duodenal reflux group. All regions of hyperplasia showed moderate or strong CYP1B1 immunoreactivity. Duodenal reflux induced a small number of premalignant changes: immunoreactivity was absent from the epithelium of squamous dysplasia (0/10), Barretts esophagus (0/7), and majority of dysplastic Barretts esophagus (1/4). Moderate or strong immunoreactivity was observed in the majority (7/8) of squamous cell carcinomas (SCCs) in situ. Immunoreactivity was also observed in the lamina propria and submucosa in association with inflammation, regardless of the severity of inflammation. The expression of CYP1B1 in hyperplasia, SCCs in situ, or in association with inflammation may increase the production of carcinogenic metabolites, which may promote esophageal tumorigenesis.


Breast Cancer Research | 2010

FKBPL: a novel prognostic and predictive biomarker?

Hayley D. McKeen; Christopher Byrne; Puthen V. Jithesh; C. Donley; Anita Yakkundi; Lynn McCallum; Helen O. McCarthy; David Hirst; Tracy Robson

Approximately 40% of patients with oestrogen receptor (ER)-positive breast cancers do not respond to endocrine therapies; furthermore, most responsive tumours eventually become resistant. We have identified a novel oestrogen-responsive Hsp90 co-chaperone and immunophilin, FKBPL, which affects the stability and signalling of ER with implications for breast cancer growth and sensitivity to endocrine therapies. MCF7 cells stably overexpressing FKBPL demonstrate a slower rate of proliferation and become highly dependent on oestrogen for their growth. This dependence on oestrogen renders these cells dramatically more sensitive to tamoxifen and fulvestrant. FKBPL overexpressing cells also exhibit decreased levels of ER and an oestrogen-responsive gene, cathepsin D, critical for breast cancer growth, survival and invasion. Moreover, knockdown of FKBPL using a targeted siRNA approach dramatically increases both ER and cathepsin D protein levels and cell resistance to tamoxifen. FKBPL has been previously implicated in the stabilisation of the cyclin-dependent kinase inhibitor, p21 [1]. Loss of p21 has been associated with a tamoxifen growth-inducing phenotype and hyperphosphorylation of ER at Ser118, with increased expression of ER-regulated genes. Following FKBPL knockdown, we have observed a fall in p21 levels and subsequent increase in Ser118 phosphorylation following treatment with 17β-estradiol or tamoxifen while FKBPL overexpressing cells exhibit the reverse effects. Our in vitro data support a model in which high levels of FKBPL would stabilise p21, decrease ER phosphorylation and abrogate tamoxifen-induced agonist potency, thereby increasing drug sensitivity, and suggest that FKBPL may have prognostic value that might impact upon tumour proliferative capacity and improve patient outcome. In addition, analysis of two publically available breast cancer microarray patient cohorts demonstrated that high FKBPL expression was correlated with increased overall and distant metastasis-free survival.

Collaboration


Dive into the Hayley D. McKeen's collaboration.

Top Co-Authors

Avatar

Tracy Robson

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar

Anita Yakkundi

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar

David Hirst

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar

Helen O. McCarthy

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar

Christopher Byrne

Royal College of Surgeons in Ireland

View shared research outputs
Top Co-Authors

Avatar

Andrea Valentine

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar

Lana McClements

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar

Keeva McClelland

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rachel Bennett

Queen's University Belfast

View shared research outputs
Researchain Logo
Decentralizing Knowledge