Network


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

Hotspot


Dive into the research topics where Kyle B. Matchett is active.

Publication


Featured researches published by Kyle B. Matchett.


Cancer Research | 2014

BRCA1 Deficiency Exacerbates Estrogen-Induced DNA Damage and Genomic Instability

Kienan Savage; Kyle B. Matchett; Eliana M. Barros; Kevin M. Cooper; Gareth Irwin; Julia J. Gorski; Katy S. Orr; Jekaterina Vohhodina; Joy N. Kavanagh; Angelina Madden; Alexander Powell; Lorenzo Manti; Simon S. McDade; Ben Ho Park; Kevin Prise; Stuart McIntosh; Manuel Salto-Tellez; Derek J. Richard; Christopher T. Elliott; D. Paul Harkin

Germline mutations in BRCA1 predispose carriers to a high incidence of breast and ovarian cancers. BRCA1 functions to maintain genomic stability through critical roles in DNA repair, cell-cycle arrest, and transcriptional control. A major question has been why BRCA1 loss or mutation leads to tumors mainly in estrogen-regulated tissues, given that BRCA1 has essential functions in all cell types. Here, we report that estrogen and estrogen metabolites can cause DNA double-strand breaks (DSB) in estrogen receptor-α-negative breast cells and that BRCA1 is required to repair these DSBs to prevent metabolite-induced genomic instability. We found that BRCA1 also regulates estrogen metabolism and metabolite-mediated DNA damage by repressing the transcription of estrogen-metabolizing enzymes, such as CYP1A1, in breast cells. Finally, we used a knock-in human cell model with a heterozygous BRCA1 pathogenic mutation to show how BRCA1 haploinsufficiency affects these processes. Our findings provide pivotal new insights into why BRCA1 mutation drives the formation of tumors in estrogen-regulated tissues, despite the general role of BRCA1 in DNA repair in all cell types.


Stem Cells | 2014

Concise Reviews: Cancer Stem Cells: From Concept to Cure

Kyle B. Matchett; Terence Lappin

In 1953, noting a remarkable consistency between the agents causing mutations and those associated with cancer, Carl Nordling, a Finnish‐born architect, proposed that cancer results from an accumulation of genetic mutations. It is now generally accepted that inherited mutations and environmental carcinogens can lead to the development of premalignant clones. After further mutations, one cell reaches a critical state which confers a survival or growth advantage over normal cells. Such cells have the ability to initiate a malignant tumour. They share many of the features of normal stem cells, including the capacity for self‐renewal and differentiation, and are widely termed cancer stem cells (CSCs). Although CSCs have been well characterized in hematological malignancies, their existence in some other tissues has been questioned. Here, we review recent work in which stem cells and stem cell‐like cells have been used to investigate the pathogenesis of cancer and potential anticancer treatment strategies, in the context of both hematological and somatic tissue disease. Stem Cells 2014;32:2563–2570


Advances in Experimental Medicine and Biology | 2014

Ran GTPase in Nuclear Envelope Formation and Cancer Metastasis

Kyle B. Matchett; Suzanne McFarlane; Sophie E. Hamilton; Yousef S. A. Eltuhamy; Matthew A. Davidson; James Murray; Ahmed Faheem; Mohamed El-Tanani

Ran is a small ras-related GTPase that controls the nucleocytoplasmic exchange of macromolecules across the nuclear envelope. It binds to chromatin early during nuclear formation and has important roles during the eukaryotic cell cycle, where it regulates mitotic spindle assembly, nuclear envelope formation and cell cycle checkpoint control. Like other GTPases, Ran relies on the cycling between GTP-bound and GDP-bound conformations to interact with effector proteins and regulate these processes. In nucleocytoplasmic transport, Ran shuttles across the nuclear envelope through nuclear pores. It is concentrated in the nucleus by an active import mechanism where it generates a high concentration of RanGTP by nucleotide exchange. It controls the assembly and disassembly of a range of complexes that are formed between Ran-binding proteins and cellular cargo to maintain rapid nuclear transport. Ran also has been identified as an essential protein in nuclear envelope formation in eukaryotes. This mechanism is dependent on importin-β, which regulates the assembly of further complexes important in this process, such as Nup107-Nup160. A strong body of evidence is emerging implicating Ran as a key protein in the metastatic progression of cancer. Ran is overexpressed in a range of tumors, such as breast and renal, and these perturbed levels are associated with local invasion, metastasis and reduced patient survival. Furthermore, tumors with oncogenic KRAS or PIK3CA mutations are addicted to Ran expression, which yields exciting future therapeutic opportunities.


Oncogene | 2016

Resistance to HSP90 inhibition involving loss of MCL1 addiction

Sara Busacca; E.W.P. Law; Ian R. Powley; D.A. Proia; M. Sequeira; J. Le Quesne; Astero Klabatsa; J.M. Edwards; Kyle B. Matchett; J.L. Luo; James Howard Pringle; Mohamed El-Tanani; Marion MacFarlane; Dean A. Fennell

Inhibition of the chaperone heat-shock protein 90 (HSP90) induces apoptosis, and it is a promising anti-cancer strategy. The mechanisms underpinning apoptosis activation following HSP90 inhibition and how they are modified during acquired drug resistance are unknown. We show for the first time that, to induce apoptosis, HSP90 inhibition requires the cooperation of multi BH3-only proteins (BID, BIK, PUMA) and the reciprocal suppression of the pro-survival BCL-2 family member MCL1, which occurs via inhibition of STAT5A. A subset of tumour cell lines exhibit dependence on MCL1 expression for survival and this dependence is also associated with tumour response to HSP90 inhibition. In the acquired resistance setting, MCL1 suppression in response to HSP90 inhibitors is maintained; however, a switch in MCL1 dependence occurs. This can be exploited by the BH3 peptidomimetic ABT737, through non-BCL-2-dependent synthetic lethality.


British Journal of Haematology | 2015

Novel antibodies directed against the human erythropoietin receptor: creating a basis for clinical implementation

Perry Maxwell; Florinda Meléndez-Rodríguez; Kyle B. Matchett; Julián Aragonés; Nathalie Ben-Califa; Heidelinde Jaekel; Ludger Hengst; Herbert Lindner; André Bernardini; Ulf Brockmeier; Joachim Fandrey; Fritz Grunert; Howard S. Oster; Moshe Mittelman; Mohamed El-Tanani; Markus Thiersch; Edith M. Schneider Gasser; Max Gassmann; David Dangoor; Robert J. Cuthbert; Alexandra Irvine; Anne Jordan; Terence Lappin; John F. Thompson; Drorit Neumann

Recombinant human erythropoietin (rHuEPO) is an effective treatment for anaemia but concerns that it causes disease progression in cancer patients by activation of EPO receptors (EPOR) in tumour tissue have been controversial and have restricted its clinical use. Initial clinical studies were flawed because they used polyclonal antibodies, later shown to lack specificity for EPOR. Moreover, multiple isoforms of EPOR caused by differential splicing have been reported in cancer cell lines at the mRNA level but investigations of these variants and their potential impact on tumour progression, have been hampered by lack of suitable antibodies. The EpoCan consortium seeks to promote improved pathological testing of EPOR, leading to safer clinical use of rHuEPO, by producing well characterized EPOR antibodies. Using novel genetic and traditional peptide immunization protocols, we have produced mouse and rat monoclonal antibodies, and show that several of these specifically recognize EPOR by Western blot, immunoprecipitation, immunofluorescence, flow cytometry and immunohistochemistry in cell lines and clinical material. Widespread availability of these antibodies should enable the research community to gain a better understanding of the role of EPOR in cancer, and eventually to distinguish patients who can be treated safely by rHuEPO from those at increased risk from treatment.


International Journal of Pharmaceutics | 2017

Nano-encapsulation of a novel anti-Ran-GTPase peptide for blockade of regulator of chromosome condensation 1 (RCC1) function in MDA-MB-231 breast cancer cells

Yusuf A. Haggag; Kyle B. Matchett; El-Habib Dakir; Paul J. Buchanan; Mohamed A. Osman; Sanaa A. El-Gizawy; Mohamed El-Tanani; Ahmed Faheem; Paul A. McCarron

Ran is a small ras-related GTPase and is highly expressed in aggressive breast carcinoma. Overexpression induces malignant transformation and drives metastatic growth. We have designed a novel series of anti-Ran-GTPase peptides, which prevents Ran hydrolysis and activation, and although they display effectiveness in silico, peptide activity is suboptimal in vitro due to reduced bioavailability and poor delivery. To overcome this drawback, we delivered an anti-Ran-GTPase peptide using encapsulation in PLGA-based nanoparticles (NP). Formulation variables within a double emulsion solvent evaporation technique were controlled to optimise physicochemical properties. NP were spherical and negatively charged with a mean diameter of 182-277nm. Peptide integrity and stability were maintained after encapsulation and release kinetics followed a sustained profile. We were interested in the relationship between cellular uptake and poly(ethylene glycol) (PEG) in the NP matrix, with results showing enhanced in vitro uptake with increasing PEG content. Peptide-loaded, pegylated (10% PEG)-PLGA NP induced significant cytotoxic and apoptotic effects in MDA-MB-231 breast cancer cells, with no evidence of similar effects in cells pulsed with free peptide. Western blot analysis showed that encapsulated peptide interfered with the proposed signal transduction pathway of the Ran gene. Our novel blockade peptide prevented Ran activation by blockage of regulator of chromosome condensation 1 (RCC1) following peptide release directly in the cytoplasm once endocytosis of the peptide-loaded nanoparticle has occurred. RCC1 blockage was effective only when a nanoparticulate delivery approach was adopted.


Expert Review of Molecular Diagnostics | 2013

Molecular and clinicopathological markers of prognosis in breast cancer

David P. Boyle; Clare McCourt; Kyle B. Matchett; Manuel Salto-Tellez

A vast body of research in breast cancer prognostication has accumulated. Yet despite this, patients within current prognostic categories may have significantly different outcomes. There is a need to more accurately divide those cancer types associated with an excellent prognosis from those requiring more aggressive therapy. Gene expression array studies have revealed the numerous molecular breast cancer subtypes that are associated with differing outcomes. Furthermore, as next generation technologies evolve and further reveal the complexities of breast cancer, it is likely that existing prognostic approaches will become progressively refined. Future prognostication in breast cancer requires a morphomolecular, multifaceted approach involving the assessment of anatomical disease extent and levels of protein, DNA and RNA expression. One of the major challenges in prognostication will be the integration of potential assays into existing clinical systems and identification of appropriate patient subgroups for analysis.


Oncotarget | 2017

Erythropoietin drives breast cancer progression by activation of its receptor EPOR

Ka Kui Chan; Kyle B. Matchett; Jonathan A. Coulter; Hiu-Fung Yuen; Cian M. McCrudden; Shu-Dong Zhang; Gareth Irwin; Matthew A. Davidson; Thomas Rülicke; Sophie Schober; Ludger Hengst; Heidelinde Jaekel; Angela Platt-Higgins; Philip S. Rudland; Ken I. Mills; Perry Maxwell; Mohamed El-Tanani; Terence Lappin

Breast cancer is a leading cause of cancer-related deaths. Anemia is common in breast cancer patients and can be treated with blood transfusions or with recombinant erythropoietin (EPO) to stimulate red blood cell production. Clinical studies have indicated decreased survival in some groups of cancer patients treated with EPO. Numerous tumor cells express the EPO receptor (EPOR), posing a risk that EPO treatment would enhance tumor growth, but the mechanisms involved in breast tumor progression are poorly understood. Here, we have examined the functional role of the EPO-EPOR axis in pre-clinical models of breast cancer. EPO induced the activation of PI3K/AKT and MAPK pathways in human breast cancer cell lines. EPOR knockdown abrogated human tumor cell growth, induced apoptosis through Bim, reduced invasiveness, and caused downregulation of MYC expression. EPO-induced MYC expression is mediated through the PI3K/AKT and MAPK pathways, and overexpression of MYC partially rescued loss of cell proliferation caused by EPOR downregulation. In a xenotransplantation model, designed to simulate recombinant EPO therapy in breast cancer patients, knockdown of EPOR markedly reduced tumor growth. Thus, our experiments in vitro and in vivo demonstrate that functional EPOR signaling is essential for the tumor-promoting effects of EPO and underline the importance of the EPO-EPOR axis in breast tumor progression.


Oncotarget | 2016

Low-dose salinomycin induces anti-leukemic responses in AML and MLL.

Gary D.R. Roulston; Charlotte L. Burt; Laura M.J. Kettyle; Kyle B. Matchett; Heather L. Keenan; Nuala M. Mulgrew; Joanne M. Ramsey; Caoifa Dougan; John McKiernan; Ivan V. Grishagin; Ken I. Mills; Alexander Thompson

Development of anti-cancer drugs towards clinical application is costly and inefficient. Large screens of drugs, efficacious for non-cancer disease, are currently being used to identify candidates for repurposing based on their anti-cancer properties. Here, we show that low-dose salinomycin, a coccidiostat ionophore previously identified in a breast cancer screen, has anti-leukemic efficacy. AML and MLLr cell lines, primary cells and patient samples were sensitive to submicromolar salinomycin. Most strikingly, colony formation of normal hematopoietic cells was unaffected by salinomycin, demonstrating a lack of hemotoxicity at the effective concentrations. Furthermore, salinomycin treatment of primary cells resulted in loss of leukemia repopulation ability following transplantation, as demonstrated by extended recipient survival compared to controls. Bioinformatic analysis of a 17-gene signature identified and validated in primary MLLr cells, uncovered immunomodulatory pathways, hubs and protein interactions as potential transducers of low dose salinomycin treatment. Additionally, increased protein expression of p62/Sqstm1, encoded for by one of the 17 signature genes, demonstrates a role for salinomycin in aggresome/vesicle formation indicative of an autophagic response. Together, the data support the efficacy of salinomycin as an anti-leukemic at non-hemotoxic concentrations. Further investigation alone or in combination with other therapies is warranted for future clinical trial.


Cancers | 2017

Advances in Precision Medicine: Tailoring Individualized Therapies

Kyle B. Matchett; Niamh Lynam-Lennon; R. Watson; James A. L. Brown

The traditional bench-to-bedside pipeline involves using model systems and patient samples to provide insights into pathways deregulated in cancer. This discovery reveals new biomarkers and therapeutic targets, ultimately stratifying patients and informing cohort-based treatment options. Precision medicine (molecular profiling of individual tumors combined with established clinical-pathological parameters) reveals, in real-time, individual patient’s diagnostic and prognostic risk profile, informing tailored and tumor-specific treatment plans. Here we discuss advances in precision medicine presented at the Irish Association for Cancer Research Annual Meeting, highlighting examples where personalized medicine approaches have led to precision discovery in individual tumors, informing customized treatment programs.

Collaboration


Dive into the Kyle B. Matchett's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ahmed Faheem

University of Sunderland

View shared research outputs
Top Co-Authors

Avatar

Ken I. Mills

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar

Terence Lappin

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eliana M. Barros

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar

Gareth Irwin

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar

Julia J. Gorski

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar

Kevin M. Cooper

Queen's University Belfast

View shared research outputs
Researchain Logo
Decentralizing Knowledge