60 results on '"McDade SS"'
Search Results
2. Activation of innate-adaptive immune machinery by poly(I:C) exposes a therapeutic vulnerability to prevent relapse in stroma-rich colon cancer
- Author
-
Corry, SM, McCorry, AMB, Lannagan, TRM, Leonard, NA, Fisher, NC, Byrne, RM, Tsantoulis, P, Cortes-Lavaud, X, Amirkhah, R, Redmond, KL, McCooey, AJ, Malla, SB, Rogan, E, Sakhnevych, S, Gillespie, MA, White, M, Richman, SD, Jackstadt, R-F, Campbell, AD, Maguire, S, S:CORT and ACRCelerate consortia, McDade, SS, Longley, DB, Loughrey, MB, Coleman, HG, Kerr, EM, Tejpar, S, Maughan, T, Leedham, SJ, Small, DM, Ryan, AE, Sansom, OJ, Lawler, M, and Dunne, PD
- Abstract
Objective Stroma-rich tumours represent a poor prognostic subtype in stage II/III colon cancer (CC), with high relapse rates and limited response to standard adjuvant chemotherapy. Design To address the lack of efficacious therapeutic options for patients with stroma-rich CC, we stratified our human tumour cohorts according to stromal content, enabling identification of the biology underpinning relapse and potential therapeutic vulnerabilities specifically within stroma-rich tumours that could be exploited clinically. Following human tumour-based discovery and independent clinical validation, we use a series of in vitro and stroma-rich in vivo models to test and validate the therapeutic potential of elevating the biology associated with reduced relapse in human tumours. Results By performing our analyses specifically within the stroma-rich/high-fibroblast (HiFi) subtype of CC, we identify and validate the clinical value of a HiFi-specific prognostic signature (HPS), which stratifies tumours based on STAT1-related signalling (High-HPS v Low-HPS=HR 0.093, CI 0.019 to 0.466). Using in silico, in vitro and in vivo models, we demonstrate that the HPS is associated with antigen processing and presentation within discrete immune lineages in stroma-rich CC, downstream of double-stranded RNA and viral response signalling. Treatment with the TLR3 agonist poly(I:C) elevated the HPS signalling and antigen processing phenotype across in vitro and in vivo models. In an in vivo model of stroma-rich CC, poly(I:C) treatment significantly increased systemic cytotoxic T cell activity (p
- Published
- 2022
3. Regulation of FLIP(L) and TRAIL-R2 signalling by the SCFSkp2Ubiquitin Ligase Complex
- Author
-
Roberts, JZ, primary, Holohan, C, additional, Sessler, T, additional, Fox, J, additional, Higgins, C., additional, Espona-Fiedler, G, additional, Majkut, J, additional, Crawford, N, additional, Riley, JS, additional, Khawaja, H, additional, Humphreys, LM, additional, Ferris, J, additional, Evergren, E, additional, Moynagh, P, additional, McDade, SS, additional, and Longley, DB, additional
- Published
- 2019
- Full Text
- View/download PDF
4. Genome-wide association study identifies a common variant in RAD51B associated with male breast cancer risk
- Author
-
Orr, N, Lemnrau, A, Cooke, R, Fletcher, O, Tomczyk, K, Jones, M, Johnson, N, Lord, CJ, Mitsopoulos, C, Zvelebil, M, McDade, SS, Buck, G, Blancher, C, Trainer, AH, James, PA, Bojesen, SE, Bokmand, S, Nevanlinna, H, Mattson, J, Friedman, E, Laitman, Y, Palli, D, Masala, G, Zanna, I, Ottini, L, Giannini, G, Hollestelle, A, van den Ouweland, AMW, Novakovic, S, Krajc, M, Gago-Dominguez, M, Castelao, JE, Olsson, H, Hedenfalk, I, Easton, DF, Pharoah, PDP, Dunning, AM, Bishop, DT, Neuhausen, SL, Steele, L, Houlston, RS, Garcia-Closas, M, Ashworth, A, Swerdlow, AJ, Orr, N, Lemnrau, A, Cooke, R, Fletcher, O, Tomczyk, K, Jones, M, Johnson, N, Lord, CJ, Mitsopoulos, C, Zvelebil, M, McDade, SS, Buck, G, Blancher, C, Trainer, AH, James, PA, Bojesen, SE, Bokmand, S, Nevanlinna, H, Mattson, J, Friedman, E, Laitman, Y, Palli, D, Masala, G, Zanna, I, Ottini, L, Giannini, G, Hollestelle, A, van den Ouweland, AMW, Novakovic, S, Krajc, M, Gago-Dominguez, M, Castelao, JE, Olsson, H, Hedenfalk, I, Easton, DF, Pharoah, PDP, Dunning, AM, Bishop, DT, Neuhausen, SL, Steele, L, Houlston, RS, Garcia-Closas, M, Ashworth, A, and Swerdlow, AJ
- Abstract
We conducted a genome-wide association study of male breast cancer comprising 823 cases and 2,795 controls of European ancestry, with validation in independent sample sets totaling 438 cases and 474 controls. A SNP in RAD51B at 14q24.1 was significantly associated with male breast cancer risk (P = 3.02 × 10(-13); odds ratio (OR) = 1.57). We also refine association at 16q12.1 to a SNP within TOX3 (P = 3.87 × 10(-15); OR = 1.50).
- Published
- 2012
5. Analysis of the DNA damage-induced TP53 and TP63 cistromes in neonatal keratinocytes
- Author
-
McDade, SS, primary
- Full Text
- View/download PDF
6. ClassifieR 2.0: expanding interactive gene expression-based stratification to prostate and high-grade serous ovarian cancer.
- Author
-
McCabe A, Quinn GP, Jain S, Ó Dálaigh M, Dean K, Murphy RG, and McDade SS
- Subjects
- Humans, Female, Male, Computational Biology methods, Software, Transcriptome genetics, Gene Expression Regulation, Neoplastic genetics, Ovarian Neoplasms genetics, Ovarian Neoplasms classification, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism, Gene Expression Profiling methods
- Abstract
Background: Advances in transcriptional profiling methods have enabled the discovery of molecular subtypes within and across traditional tissue-based cancer classifications. Such molecular subgroups hold potential for improving patient outcomes by guiding treatment decisions and revealing physiological distinctions and targetable pathways. Computational methods for stratifying transcriptomic data into molecular subgroups are increasingly abundant. However, assigning samples to these subtypes and other transcriptionally inferred predictions is time-consuming and requires significant bioinformatics expertise. To address this need, we recently reported "ClassifieR," a flexible, interactive cloud application for the functional annotation of colorectal and breast cancer transcriptomes. Here, we report "ClassifieR 2.0" which introduces additional modules for the molecular subtyping of prostate and high-grade serous ovarian cancer (HGSOC)., Results: ClassifieR 2.0 introduces ClassifieRp and ClassifieRov, two specialised modules specifically designed to address the challenges of prostate and HGSOC molecular classification. ClassifieRp includes sigInfer, a method we developed to infer commercial prognostic prostate gene expression signatures from publicly available gene-lists or indeed any user-uploaded gene-list. ClassifieRov utilizes consensus molecular subtyping methods for HGSOC, including tools like consensusOV, for accurate ovarian cancer stratification. Both modules include functionalities present in the original ClassifieR framework for estimating cellular composition, predicting transcription factor (TF) activity and single sample gene set enrichment analysis (ssGSEA)., Conclusions: ClassifieR 2.0 combines molecular subtyping of prostate cancer and HGSOC with commonly used sample annotation tools in a single, user-friendly platform, allowing scientists without bioinformatics training to explore prostate and HGSOC transcriptional data without the need for extensive bioinformatics knowledge or manual data handling to operate various packages. Our sigInfer method within ClassifieRp enables the inference of commercially available gene signatures for prostate cancer, while ClassifieRov incorporates consensus molecular subtyping for HGSOC. Overall, ClassifieR 2.0 aims to make molecular subtyping more accessible to the wider research community. This is crucial for increased understanding of the molecular heterogeneity of these cancers and developing personalised treatment strategies., Competing Interests: Declarations. Ethics approval and consent to participate: Not applicable. Consent for publication: Not applicable. Competing interests: SSM and GPQ are share-holders of generatR Ltd trading as BlokBio, a cloud genomics data analysis company. All other authors have no competing interests., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
7. Ribosome Quality Control mitigates the cytotoxicity of ribosome collisions induced by 5-Fluorouracil.
- Author
-
Chatterjee S, Naeli P, Onar O, Simms N, Garzia A, Hackett A, Coyle K, Harris Snell P, McGirr T, Sawant TN, Dang K, Stoichkova ZV, Azam Y, Saunders MP, Braun M, Alain T, Tuschl T, McDade SS, Longley DB, Gkogkas CG, Adrain C, Knight JRP, and Jafarnejad SM
- Subjects
- Humans, Signal Transduction drug effects, Antimetabolites, Antineoplastic pharmacology, Antimetabolites, Antineoplastic toxicity, Protein Biosynthesis drug effects, Cell Line, Tumor, RNA, Messenger metabolism, RNA, Messenger genetics, Peptide Chain Initiation, Translational drug effects, Fluorouracil pharmacology, Fluorouracil toxicity, Ribosomes metabolism, Ribosomes drug effects, TOR Serine-Threonine Kinases metabolism
- Abstract
Ribosome quality control (RQC) resolves collided ribosomes, thus preventing their cytotoxic effects. The chemotherapeutic agent 5-Fluorouracil (5FU) is best known for its misincorporation into DNA and inhibition of thymidylate synthase. However, while a major determinant of 5FU's anticancer activity is its misincorporation into RNAs, the mechanisms by which cancer cells overcome the RNA-dependent 5FU toxicity remain ill-defined. Here, we report a role for RQC in mitigating the cytotoxic effects of 5FU. We show that 5FU treatment results in rapid induction of the mTOR signalling pathway, enhanced rate of mRNA translation initiation, and increased ribosome collisions. Consistently, a defective RQC exacerbates the 5FU-induced cell death, which is mitigated by blocking mTOR pathway or mRNA translation initiation. Furthermore, 5FU treatment enhances the expression of the key RQC factors ZNF598 and GIGYF2 via an mTOR-dependent post-translational mechanism. This adaptation likely mitigates the cytotoxic consequences of increased ribosome collisions upon 5FU treatment., (© The Author(s) 2024. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2024
- Full Text
- View/download PDF
8. Dual-Hit Strategy for Therapeutic Targeting of Pancreatic Cancer in Patient-Derived Xenograft Tumors.
- Author
-
Roy Chaudhuri T, Lin Q, Stachowiak EK, Rosario SR, Spernyak JA, Ma WW, Stachowiak MK, Greene MK, Quinn GP, McDade SS, Clynes M, Scott CJ, and Straubinger RM
- Subjects
- Humans, Animals, Heterografts, Hedgehog Proteins genetics, Hedgehog Proteins metabolism, Signal Transduction, Disease Models, Animal, Cell Line, Tumor, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal metabolism
- Abstract
Purpose: Paracrine activation of pro-fibrotic hedgehog (HH) signaling in pancreatic ductal adenocarcinoma (PDAC) results in stromal amplification that compromises tumor drug delivery, efficacy, and patient survival. Interdiction of HH-mediated tumor-stroma crosstalk with smoothened (SMO) inhibitors (SHHi) "primes" PDAC patient-derived xenograft (PDX) tumors for increased drug delivery by transiently increasing vascular patency/permeability, and thereby macromolecule delivery. However, patient tumor isolates vary in their responsiveness, and responders show co-induction of epithelial-mesenchymal transition (EMT). We aimed to identify the signal derangements responsible for EMT induction and reverse them and devise approaches to stratify SHHi-responsive tumors noninvasively based on clinically-quantifiable parameters., Experimental Design: Animals underwent diffusion-weighted magnetic resonance (DW-MR) imaging for measurement of intratumor diffusivity. In parallel, tissue-level deposition of nanoparticle probes was quantified as a marker of vascular permeability/perfusion. Transcriptomic and bioinformatic analysis was employed to investigate SHHi-induced gene reprogramming and identify key "nodes" responsible for EMT induction., Results: Multiple patient tumor isolates responded to short-term SHH inhibitor exposure with increased vascular patency and permeability, with proportionate increases in tumor diffusivity. Nonresponding PDXs did not. SHHi-treated tumors showed elevated FGF drive and distinctly higher nuclear localization of fibroblast growth factor receptor (FGFR1) in EMT-polarized tumor cells. Pan-FGFR inhibitor NVP-BGJ398 (Infigratinib) reversed the SHHi-induced EMT marker expression and nuclear FGFR1 accumulation without compromising the enhanced permeability effect., Conclusions: This dual-hit strategy of SMO and FGFR inhibition provides a clinically-translatable approach to compromise the profound impermeability of PDAC tumors. Furthermore, clinical deployment of DW-MR imaging could fulfill the essential clinical-translational requirement for patient stratification., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
9. Nanoencapsulation of MDM2 Inhibitor RG7388 and Class-I HDAC Inhibitor Entinostat Enhances their Therapeutic Potential Through Synergistic Antitumor Effects and Reduction of Systemic Toxicity.
- Author
-
Abed A, Greene MK, Alsa'd AA, Lees A, Hindley A, Longley DB, McDade SS, and Scott CJ
- Subjects
- Humans, Animals, Mice, Tumor Suppressor Protein p53 metabolism, Apoptosis, Cell Line, Tumor, Drug Combinations, Proto-Oncogene Proteins c-mdm2, Histone Deacetylase Inhibitors pharmacology, Histone Deacetylase Inhibitors therapeutic use, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Benzamides, Pyridines, Pyrrolidines, para-Aminobenzoates
- Abstract
Inhibitors of the p53-MDM2 interaction such as RG7388 have been developed to exploit latent tumor suppressive properties in p53 in 50% of tumors in which p53 is wild-type. However, these agents for the most part activate cell cycle arrest rather than death, and high doses in patients elicit on-target dose-limiting neutropenia. Recent work from our group indicates that combination of p53-MDM2 inhibitors with the class-I HDAC inhibitor Entinostat (which itself has dose-limiting toxicity issues) has the potential to significantly augment cell death in p53 wild-type colorectal cancer cells. We investigated whether coencapsulation of RG7388 and Entinostat within polymeric nanoparticles (NPs) could overcome efficacy and toxicity limitations of this drug combination. Combinations of RG7388 and Entinostat across a range of different molar ratios resulted in synergistic increases in cell death when delivered in both free drug and nanoencapsulated formats in all colorectal cell lines tested. Importantly, we also explored the in vivo impact of the drug combination on murine blood leukocytes, showing that the leukopenia induced by the free drugs could be significantly mitigated by nanoencapsulation. Taken together, this study demonstrates that formulating these agents within a single nanoparticle delivery platform may provide clinical utility beyond use as nonencapsulated agents.
- Published
- 2024
- Full Text
- View/download PDF
10. surviveR: a flexible shiny application for patient survival analysis.
- Author
-
Sessler T, Quinn GP, Wappett M, Rogan E, Sharkey D, Ahmaderaghi B, Lawler M, Longley DB, and McDade SS
- Subjects
- Humans, Survival Analysis, Kaplan-Meier Estimate, Software, Neoplasms genetics
- Abstract
Kaplan-Meier (KM) survival analyses based on complex patient categorization due to the burgeoning volumes of genomic, molecular and phenotypic data, are an increasingly important aspect of the biomedical researcher's toolkit. Commercial statistics and graphing packages for such analyses are functionally limited, whereas open-source tools have a high barrier-to-entry in terms of understanding of methodologies and computational expertise. We developed surviveR to address this unmet need for a survival analysis tool that can enable users with limited computational expertise to conduct routine but complex analyses. surviveR is a cloud-based Shiny application, that addresses our identified unmet need for an easy-to-use web-based tool that can plot and analyse survival based datasets. Integrated customization options allows a user with limited computational expertise to easily filter patients to enable custom cohort generation, automatically calculate log-rank test and Cox hazard ratios. Continuous datasets can be integrated, such as RNA or protein expression measurements which can be then used as categories for survival plotting. We further demonstrate the utility through exemplifying its application to a clinically relevant colorectal cancer patient dataset. surviveR is a cloud-based web application available at https://generatr.qub.ac.uk/app/surviveR , that can be used by non-experts users to perform complex custom survival analysis., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
11. Circulating tumour DNA kinetics in recurrent/metastatic head and neck squamous cell cancer patients.
- Author
-
Taylor K, Zou J, Magalhaes M, Oliva M, Spreafico A, Hansen AR, McDade SS, Coyle VM, Lawler M, Elimova E, Bratman SV, and Siu LL
- Subjects
- Humans, Squamous Cell Carcinoma of Head and Neck drug therapy, Squamous Cell Carcinoma of Head and Neck genetics, Kinetics, Biomarkers, Tumor genetics, Neoplasm Recurrence, Local genetics, Circulating Tumor DNA genetics, Head and Neck Neoplasms drug therapy, Head and Neck Neoplasms genetics, Neoplasms, Squamous Cell
- Abstract
Purpose: Immune checkpoint blockade (ICB) has become a standard of care in the treatment of recurrent/metastatic head and neck squamous cell cancer (R/M HNSCC). However, only a subset of patients benefit from treatment. Quantification of plasma circulating tumour DNA (ctDNA) levels and on-treatment kinetics may permit real-time assessment of disease burden under selective pressures of treatment., Patients and Methods: R/M HNSCC patients treated with systemic therapy, platinum-based chemotherapy (CT) or ICB, underwent serial liquid biopsy sampling. Biomarkers tested included ctDNA measured by CAncer Personalized Profiling by deep Sequencing (CAPP-Seq) and markers of host inflammation measured by neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR)., Results: Among 53 eligible patients, 16 (30%) received CT, 30 (57%) ICB [anti-PD1/L1] monotherapy and 7 (13%) combination immunotherapy (IO). Median progression-free survival (PFS) and overall survival (OS) were 2.8 months (95% CI, 1.3-4.3) and 8.2 months (95% CI, 5.6-10.8), respectively. Seven (13%) patients experienced a partial response and 21 (40%) derived clinical benefit. At baseline, median ctDNA variant allele frequency (VAF) was 4.3%. Baseline ctDNA abundance was not associated with OS (p = 0.56) nor PFS (p = 0.54). However, a change in ctDNA VAF after one cycle of treatment (ΔVAF (T1-2)) was predictive of both PFS (p< 0.01) and OS (p< 0.01). Additionally, decrease in ΔVAF identified patients with longer OS despite early radiological progression, 8.2 vs 4.6 months, hazard ratio 0.44 (95% CI, 0.19-0.87) p = 0.03. After incorporating NLR and PLR into multivariable Cox models, ctDNA ∆VAF retained an association with OS., Conclusions: Early dynamic changes in ctDNA abundance, after one cycle of treatment, compared to baseline predicted both OS and PFS in R/M HNSCC patients on systemic therapy., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: KT – Nothing to declare. JZ – Nothing to declare. MM – Nothing to declare. MO reports consultant/advisory role for: Merck, EMD Serono, Transgene; Grant/Research support from (Clinical Trials): Merck, Boerhinger-Ingelheim, GlaxoSmithKline, Roche/Genentech, Bayer, Abbvie, EMD Serono, ALX Oncology, ISA Therapeutics, Ayala Therapeutics, Debiopharm; Honoraria: Merck, EMD Serono, BMS outside the submitted work. AS reports grants and personal fees from Novartis, Merck, BMS, JNJ, and Oncorus and grants from Symphogen, Roche, Northern Biologics, Regeneron, AstraZeneca MedImmune, ArrayBiopharma/Pfizer, GSK, Bayer, Surface Oncology, and Treadwell outside the submitted work. AH reports grants and other support from GSK, Merck, Eisai, and Novartis, as well as grants from MedImmune, Roche, Boehringer Ingelheim, Pfizer, Janssen, and BMS outside the submitted work. SMcD reports Co-founder/shareholder: AilseVax outside of submitted work. VC reports research grants: Astex Pharmaceuticals, Cancer Research UK, NIHR and honoraria: Servier, outside of submitted work. ML reports educational grant from Pfizer and honoraria: Pfizer, EMD Serono, Roche, Carnall Farrar outside the submitted work. EE reports receiving research funding from Bristol Myers Squibb and Zymeworks; serving as a consultant or in an advisory role for Bristol Myers Squibb, Zymeworks, and Adaptimmune; and having an immediate family member employed by Merck, outside the submitted work. SVB is inventor on patents related to cell-free DNA mutation and methylation analysis technologies that have been licensed to Roche Molecular Diagnostics and Adela, respectively. SVB is a cofounder of, has ownership in, and serves in a leadership role at Adela. LLS reports personal fees from Merck, Pfizer, Celgene, AstraZeneca, Morphosys, Roche, Loxo, Oncorus, Symphogen, Seattle Genetics, GlaxoSmithKline, Voronoi, Treadwell Therapeutics, Arvinas, Tessa, Navire, Relay Therapeutics, Rubius, Janpix, Agios, and Treadwell Therapeutics and grants from Merck, Novartis, Bristol-Myers Squibb, Pfizer, Boerhinger-Ingelheim, GlaxoSmithKline, Roche/Genentech, Karyopharm, AstraZeneca, Astellas, Bayer, AbbVie, Amgen, Symphogen, Intensity Therapeutics, Mirati, Shattucks, and Avid outside the submitted work., (Copyright © 2023 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
12. Autocrine activation of MAPK signaling mediates intrinsic tolerance to androgen deprivation in LY6D prostate cancer cells.
- Author
-
Steiner I, Flores-Tellez TDNJ, Mevel R, Ali A, Wang P, Schofield P, Behan C, Forsythe N, Ashton G, Taylor C, Mills IG, Oliveira P, McDade SS, Zaiss DM, Choudhury A, Lacaud G, and Baena E
- Subjects
- Animals, Male, Mice, Androgen Antagonists pharmacology, Cell Adhesion Molecules metabolism, Cell Line, Tumor, GPI-Linked Proteins metabolism, Mitogen-Activated Protein Kinases metabolism, Prostate metabolism, Receptors, Androgen metabolism, Androgens metabolism, Prostatic Neoplasms, Castration-Resistant metabolism
- Abstract
The emergence of castration-resistant prostate cancer remains an area of unmet clinical need. We recently identified a subpopulation of normal prostate progenitor cells, characterized by an intrinsic resistance to androgen deprivation and expression of LY6D. We here demonstrate that conditional deletion of PTEN in the murine prostate epithelium causes an expansion of transformed LY6D
+ progenitor cells without impairing stem cell properties. Transcriptomic analyses of LY6D+ luminal cells identified an autocrine positive feedback loop, based on the secretion of amphiregulin (AREG)-mediated activation of mitogen-activated protein kinase (MAPK) signaling, increasing cellular fitness and organoid formation. Pharmacological interference with this pathway overcomes the castration-resistant properties of LY6D+ cells with a suppression of organoid formation and loss of LY6D+ cells in vivo. Notably, LY6D+ tumor cells are enriched in high-grade and androgen-resistant prostate cancer, providing clinical evidence for their contribution to advanced disease. Our data indicate that early interference with MAPK inhibitors can prevent progression of castration-resistant prostate cancer., Competing Interests: Declaration of interests All authors declare no competing interests., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
13. Investigating the suitability of in vitro cell lines as models for the major subtypes of epithelial ovarian cancer.
- Author
-
McCabe A, Zaheed O, McDade SS, and Dean K
- Abstract
Epithelial ovarian cancer (EOC) is the most fatal gynaecological malignancy, accounting for over 200,000 deaths worldwide per year. EOC is a highly heterogeneous disease, classified into five major histological subtypes-high-grade serous (HGSOC), clear cell (CCOC), endometrioid (ENOC), mucinous (MOC) and low-grade serous (LGSOC) ovarian carcinomas. Classification of EOCs is clinically beneficial, as the various subtypes respond differently to chemotherapy and have distinct prognoses. Cell lines are often used as in vitro models for cancer, allowing researchers to explore pathophysiology in a relatively cheap and easy to manipulate system. However, most studies that make use of EOC cell lines fail to recognize the importance of subtype. Furthermore, the similarity of cell lines to their cognate primary tumors is often ignored. Identification of cell lines with high molecular similarity to primary tumors is needed in order to better guide pre-clinical EOC research and to improve development of targeted therapeutics and diagnostics for each distinctive subtype. This study aims to generate a reference dataset of cell lines representative of the major EOC subtypes. We found that non-negative matrix factorization (NMF) optimally clustered fifty-six cell lines into five groups, putatively corresponding to each of the five EOC subtypes. These clusters validated previous histological groupings, while also classifying other previously unannotated cell lines. We analysed the mutational and copy number landscapes of these lines to investigate whether they harboured the characteristic genomic alterations of each subtype. Finally we compared the gene expression profiles of cell lines with 93 primary tumor samples stratified by subtype, to identify lines with the highest molecular similarity to HGSOC, CCOC, ENOC, and MOC. In summary, we examined the molecular features of both EOC cell lines and primary tumors of multiple subtypes. We recommend a reference set of cell lines most suited to represent four different subtypes of EOC for both in silico and in vitro studies. We also identify lines displaying poor overall molecular similarity to EOC tumors, which we argue should be avoided in pre-clinical studies. Ultimately, our work emphasizes the importance of choosing suitable cell line models to maximise clinical relevance of experiments., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 McCabe, Zaheed, McDade and Dean.)
- Published
- 2023
- Full Text
- View/download PDF
14. Bcl-xL Is a Key Mediator of Apoptosis Following KRASG12C Inhibition in KRASG12C-mutant Colorectal Cancer.
- Author
-
Khawaja H, Briggs R, Latimer CH, Rassel M, Griffin D, Hanson L, Bardelli A, Di Nicolantonio F, McDade SS, Scott CJ, Lambe S, Maurya M, Lindner AU, Prehn JHM, Sousa J, Winnington C, LaBonte MJ, Ross S, and Van Schaeybroeck S
- Subjects
- Humans, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Cell Line, Tumor, Apoptosis, bcl-X Protein genetics, bcl-X Protein metabolism, Proto-Oncogene Proteins c-bcl-2 genetics, Proto-Oncogene Proteins c-bcl-2 metabolism, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics
- Abstract
Novel covalent inhibitors of KRASG12C have shown limited response rates in patients with KRASG12C-mutant (MT) colorectal cancer. Thus, novel KRASG12C inhibitor combination strategies that can achieve deep and durable responses are needed. Small-molecule KRASG12C inhibitors AZ'1569 and AZ'8037 were used. To identify novel candidate combination strategies for AZ'1569, we performed RNA sequencing, siRNA, and high-throughput drug screening. Top hits were validated in a panel of KRASG12CMT colorectal cancer cells and in vivo. AZ'1569-resistant colorectal cancer cells were generated and characterized. We found that response to AZ'1569 was heterogeneous across the KRASG12CMT models. AZ'1569 was ineffective at inducing apoptosis when used as a single agent or combined with chemotherapy or agents targeting the EGFR/KRAS/AKT axis. Using a systems biology approach, we identified the antiapoptotic BH3-family member BCL2L1/Bcl-xL as a top hit mediating resistance to AZ'1569. Further analyses identified acute increases in the proapoptotic protein BIM following AZ'1569 treatment. ABT-263 (navitoclax), a pharmacologic Bcl-2 family inhibitor that blocks the ability of Bcl-xL to bind and inhibit BIM, led to dramatic and universal apoptosis when combined with AZ'1569. Furthermore, this combination also resulted in dramatically attenuated tumor growth in KRASG12CMT xenografts. Finally, AZ'1569-resistant cells showed amplification of KRASG12C, EphA2/c-MET activation, increased proinflammatory chemokine profile and cross-resistance to several targeted agents. Importantly, KRAS amplification and AZ'1569 resistance were reversible upon drug withdrawal, arguing strongly for the use of drug holidays in the case of KRAS amplification. Taken together, combinatorial targeting of Bcl-xL and KRASG12C is highly effective, suggesting a novel therapeutic strategy for patients with KRASG12CMT colorectal cancer., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
15. Activation of innate-adaptive immune machinery by poly(I:C) exposes a therapeutic vulnerability to prevent relapse in stroma-rich colon cancer.
- Author
-
Corry SM, McCorry AM, Lannagan TR, Leonard NA, Fisher NC, Byrne RM, Tsantoulis P, Cortes-Lavaud X, Amirkhah R, Redmond KL, McCooey AJ, Malla SB, Rogan E, Sakhnevych S, Gillespie MA, White M, Richman SD, Jackstadt RF, Campbell AD, Maguire S, McDade SS, Longley DB, Loughrey MB, Coleman HG, Kerr EM, Tejpar S, Maughan T, Leedham SJ, Small DM, Ryan AE, Sansom OJ, Lawler M, and Dunne PD
- Subjects
- Humans, Stromal Cells pathology, Neoplasm Recurrence, Local prevention & control, Neoplasm Recurrence, Local pathology, Prognosis, Biomarkers, Tumor genetics, Colonic Neoplasms pathology
- Abstract
Objective: Stroma-rich tumours represent a poor prognostic subtype in stage II/III colon cancer (CC), with high relapse rates and limited response to standard adjuvant chemotherapy., Design: To address the lack of efficacious therapeutic options for patients with stroma-rich CC, we stratified our human tumour cohorts according to stromal content, enabling identification of the biology underpinning relapse and potential therapeutic vulnerabilities specifically within stroma-rich tumours that could be exploited clinically. Following human tumour-based discovery and independent clinical validation, we use a series of in vitro and stroma-rich in vivo models to test and validate the therapeutic potential of elevating the biology associated with reduced relapse in human tumours., Results: By performing our analyses specifically within the stroma-rich/high-fibroblast (HiFi) subtype of CC, we identify and validate the clinical value of a HiFi-specific prognostic signature (HPS), which stratifies tumours based on STAT1-related signalling (High-HPS v Low-HPS=HR 0.093, CI 0.019 to 0.466). Using in silico, in vitro and in vivo models, we demonstrate that the HPS is associated with antigen processing and presentation within discrete immune lineages in stroma-rich CC, downstream of double-stranded RNA and viral response signalling. Treatment with the TLR3 agonist poly(I:C) elevated the HPS signalling and antigen processing phenotype across in vitro and in vivo models. In an in vivo model of stroma-rich CC, poly(I:C) treatment significantly increased systemic cytotoxic T cell activity (p<0.05) and reduced liver metastases (p<0.0002)., Conclusion: This study reveals new biological insight that offers a novel therapeutic option to reduce relapse rates in patients with the worst prognosis CC., Competing Interests: Competing interests: ML has received honoraria from Pfizer, EMF Serono and Roche for presentations unrelated to this work; ML has received an unrestricted educational grant from Pfizer for research unrelated to this work. PT has received honoraria and travel expenses from BMS, Merck, Roche, Lilly and Sanofi-Aventis for contributions that are not related to the present work. The authors declare no other potential conflicts of interest., (© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY. Published by BMJ.)
- Published
- 2022
- Full Text
- View/download PDF
16. VPRBP Functions Downstream of the Androgen Receptor and OGT to Restrict p53 Activation in Prostate Cancer.
- Author
-
Poulose N, Forsythe N, Polonski A, Gregg G, Maguire S, Fuchs M, Minner S, Sauter G, McDade SS, and Mills IG
- Subjects
- Humans, Male, N-Acetylglucosaminyltransferases genetics, N-Acetylglucosaminyltransferases metabolism, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Receptors, Androgen genetics, Receptors, Androgen metabolism, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism
- Abstract
Androgen receptor (AR) is a major driver of prostate cancer initiation and progression. O-GlcNAc transferase (OGT), the enzyme that catalyzes the covalent addition of UDP-N-acetylglucosamine (UDP-GlcNAc) to serine and threonine residues of proteins, is often highly expressed in prostate cancer with its expression correlated with high Gleason score. In this study, we have identified an AR and OGT coregulated factor, Vpr (HIV-1) binding protein (VPRBP) also known as DDB1 and CUL4 Associated Factor 1 (DCAF1). We show that VPRBP is regulated by the AR at the transcript level, and stabilized by OGT at the protein level. VPRBP knockdown in prostate cancer cells led to a significant decrease in cell proliferation, p53 stabilization, nucleolar fragmentation, and increased p53 recruitment to the chromatin. In human prostate tumor samples, VPRBP protein overexpression correlated with AR amplification, OGT overexpression, a shorter time to postoperative biochemical progression and poor clinical outcome. In clinical transcriptomic data, VPRBP expression was positively correlated with the AR and also with AR activity gene signatures., Implications: In conclusion, we have shown that VPRBP/DCAF1 promotes prostate cancer cell proliferation by restraining p53 activation under the influence of the AR and OGT., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
17. Functional Genomic Identification of Predictors of Sensitivity and Mechanisms of Resistance to Multivalent Second-Generation TRAIL-R2 Agonists.
- Author
-
Grinkevitch V, Wappett M, Crawford N, Price S, Lees A, McCann C, McAllister K, Prehn J, Young J, Bateson J, Gallagher L, Michaut M, Iyer V, Chatzipli A, Barthorpe S, Ciznadija D, Sloma I, Wesa A, Tice DA, Wessels L, Garnett M, Longley DB, McDermott U, and McDade SS
- Subjects
- Apoptosis, CASP8 and FADD-Like Apoptosis Regulating Protein genetics, CASP8 and FADD-Like Apoptosis Regulating Protein metabolism, Caspase 8 genetics, Cell Line, Tumor, Genomics, Humans, Proto-Oncogene Proteins c-bcl-2 metabolism, TNF-Related Apoptosis-Inducing Ligand metabolism, TNF-Related Apoptosis-Inducing Ligand pharmacology
- Abstract
Multivalent second-generation TRAIL-R2 agonists are currently in late preclinical development and early clinical trials. Herein, we use a representative second-generation agent, MEDI3039, to address two major clinical challenges facing these agents: lack of predictive biomarkers to enable patient selection and emergence of resistance. Genome-wide CRISPR knockout screens were notable for the lack of resistance mechanisms beyond the canonical TRAIL-R2 pathway (caspase-8, FADD, BID) as well as p53 and BAX in TP53 wild-type models, whereas a CRISPR activatory screen identified cell death inhibitors MCL-1 and BCL-XL as mechanisms to suppress MEDI3039-induced cell death. High-throughput drug screening failed to identify genomic alterations associated with response to MEDI3039; however, transcriptomics analysis revealed striking association between MEDI3039 sensitivity and expression of core components of the extrinsic apoptotic pathway, most notably its main apoptotic effector caspase-8 in solid tumor cell lines. Further analyses of colorectal cell lines and patient-derived xenografts identified caspase-8 expression ratio to its endogenous regulator FLIP(L) as predictive of sensitivity to MEDI3039 in several major solid tumor types and a further subset indicated by caspase-8:MCL-1 ratio. Subsequent MEDI3039 combination screening of TRAIL-R2, caspase-8, FADD, and BID knockout models with 60 compounds with varying mechanisms of action identified two inhibitor of apoptosis proteins (IAP) that exhibited strong synergy with MEDI3039 that could reverse resistance only in BID-deleted models. In summary, we identify the ratios of caspase-8:FLIP(L) and caspase-8:MCL-1 as potential predictive biomarkers for second-generation TRAIL-R2 agonists and loss of key effectors such as FADD and caspase-8 as likely drivers of clinical resistance in solid tumors., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
18. classifieR a flexible interactive cloud-application for functional annotation of cancer transcriptomes.
- Author
-
Quinn GP, Sessler T, Ahmaderaghi B, Lambe S, VanSteenhouse H, Lawler M, Wappett M, Seligmann B, Longley DB, and McDade SS
- Subjects
- Computational Biology methods, Female, Gene Expression Profiling methods, Humans, Software, Breast Neoplasms genetics, Transcriptome
- Abstract
Background: Transcriptionally informed predictions are increasingly important for sub-typing cancer patients, understanding underlying biology and to inform novel treatment strategies. For instance, colorectal cancers (CRCs) can be classified into four CRC consensus molecular subgroups (CMS) or five intrinsic (CRIS) sub-types that have prognostic and predictive value. Breast cancer (BRCA) has five PAM50 molecular subgroups with similar value, and the OncotypeDX test provides transcriptomic based clinically actionable treatment-risk stratification. However, assigning samples to these subtypes and other transcriptionally inferred predictions is time consuming and requires significant bioinformatics experience. There is no "universal" method of using data from diverse assay/sequencing platforms to provide subgroup classification using the established classifier sets of genes (CMS, CRIS, PAM50, OncotypeDX), nor one which in provides additional useful functional annotations such as cellular composition, single-sample Gene Set Enrichment Analysis, or prediction of transcription factor activity., Results: To address this bottleneck, we developed classifieR, an easy-to-use R-Shiny based web application that supports flexible rapid single sample annotation of transcriptional profiles derived from cancer patient samples form diverse platforms. We demonstrate the utility of the " classifieR" framework to applications focused on the analysis of transcriptional profiles from colorectal (classifieRc) and breast (classifieRb). Samples are annotated with disease relevant transcriptional subgroups (CMS/CRIS sub-types in classifieRc and PAM50/inferred OncotypeDX in classifieRb), estimation of cellular composition using MCP-counter and xCell, single-sample Gene Set Enrichment Analysis (ssGSEA) and transcription factor activity predictions with Discriminant Regulon Expression Analysis (DoRothEA)., Conclusions: classifieR provides a framework which enables labs without access to a dedicated bioinformation can get information on the molecular makeup of their samples, providing an insight into patient prognosis, druggability and also as a tool for analysis and discovery. Applications are hosted online at https://generatr.qub.ac.uk/app/classifieRc and https://generatr.qub.ac.uk/app/classifieRb after signing up for an account on https://generatr.qub.ac.uk ., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
19. Molecular Subtyping Resource: a user-friendly tool for rapid biological discovery from transcriptional data.
- Author
-
Ahmaderaghi B, Amirkhah R, Jackson J, Lannagan TRM, Gilroy K, Malla SB, Redmond KL, Quinn G, McDade SS, ACRCelerate Consortium, Maughan T, Leedham S, Campbell ASD, Sansom OJ, Lawler M, and Dunne PD
- Subjects
- Algorithms, Animals, Computational Biology, Humans, Mice, Sequence Analysis, RNA, Gene Expression Profiling, Software
- Abstract
Generation of transcriptional data has dramatically increased in the past decade, driving the development of analytical algorithms that enable interrogation of the biology underpinning the profiled samples. However, these resources require users to have expertise in data wrangling and analytics, reducing opportunities for biological discovery by 'wet-lab' users with a limited programming skillset. Although commercial solutions exist, costs for software access can be prohibitive for academic research groups. To address these challenges, we have developed an open source and user-friendly data analysis platform for on-the-fly bioinformatic interrogation of transcriptional data derived from human or mouse tissue, called Molecular Subtyping Resource (MouSR). This internet-accessible analytical tool, https://mousr.qub.ac.uk/, enables users to easily interrogate their data using an intuitive 'point-and-click' interface, which includes a suite of molecular characterisation options including quality control, differential gene expression, gene set enrichment and microenvironmental cell population analyses from RNA sequencing. The MouSR online tool provides a unique freely available option for users to perform rapid transcriptomic analyses and comprehensive interrogation of the signalling underpinning transcriptional datasets, which alleviates a major bottleneck for biological discovery. This article has an associated First Person interview with the first author of the paper., Competing Interests: Competing interests M.L. has received honoraria from Pfizer, EMF Serono and Roche for presentations unrelated to this work, and received an unrestricted educational grant from Pfizer for research unrelated to this work. O.J.S. has received funding from Novartis, Astra Zeneca, Cancer Research Technology and Redex for research unrelated to this work., (© 2022. Published by The Company of Biologists Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
20. TNF-α synergises with IFN-γ to induce caspase-8-JAK1/2-STAT1-dependent death of intestinal epithelial cells.
- Author
-
Woznicki JA, Saini N, Flood P, Rajaram S, Lee CM, Stamou P, Skowyra A, Bustamante-Garrido M, Regazzoni K, Crawford N, McDade SS, Longley DB, Aza-Blanc P, Shanahan F, Zulquernain SA, McCarthy J, Melgar S, McRae BL, and Nally K
- Subjects
- Apoptosis, Biopsy, Cell Death, Cell Line, Tumor, Colon pathology, Cytoprotection, Epithelial Cells metabolism, Humans, Janus Kinase 2 metabolism, Mitochondria metabolism, Organoids pathology, RNA Interference, Receptors, Tumor Necrosis Factor, Type I metabolism, Signal Transduction, Caspase 8 metabolism, Epithelial Cells pathology, Interferon-gamma metabolism, Intestines pathology, Janus Kinase 1 metabolism, STAT1 Transcription Factor metabolism, Tumor Necrosis Factor-alpha metabolism
- Abstract
Rewiring of host cytokine networks is a key feature of inflammatory bowel diseases (IBD) such as Crohn's disease (CD). Th1-type cytokines-IFN-γ and TNF-α-occupy critical nodes within these networks and both are associated with disruption of gut epithelial barrier function. This may be due to their ability to synergistically trigger the death of intestinal epithelial cells (IECs) via largely unknown mechanisms. In this study, through unbiased kinome RNAi and drug repurposing screens we identified JAK1/2 kinases as the principal and nonredundant drivers of the synergistic killing of human IECs by IFN-γ/TNF-α. Sensitivity to IFN-γ/TNF-α-mediated synergistic IEC death was retained in primary patient-derived intestinal organoids. Dependence on JAK1/2 was confirmed using genetic loss-of-function studies and JAK inhibitors (JAKinibs). Despite the presence of biochemical features consistent with canonical TNFR1-mediated apoptosis and necroptosis, IFN-γ/TNF-α-induced IEC death was independent of RIPK1/3, ZBP1, MLKL or caspase activity. Instead, it involved sustained activation of JAK1/2-STAT1 signalling, which required a nonenzymatic scaffold function of caspase-8 (CASP8). Further modelling in gut mucosal biopsies revealed an intercorrelated induction of the lethal CASP8-JAK1/2-STAT1 module during ex vivo stimulation of T cells. Functional studies in CD-derived organoids using inhibitors of apoptosis, necroptosis and JAKinibs confirmed the causative role of JAK1/2-STAT1 in cytokine-induced death of primary IECs. Collectively, we demonstrate that TNF-α synergises with IFN-γ to kill IECs via the CASP8-JAK1/2-STAT1 module independently of canonical TNFR1 and cell death signalling. This non-canonical cell death pathway may underpin immunopathology driven by IFN-γ/TNF-α in diverse autoinflammatory diseases such as IBD, and its inhibition may contribute to the therapeutic efficacy of anti-TNFs and JAKinibs., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
21. Clinical Positioning of the IAP Antagonist Tolinapant (ASTX660) in Colorectal Cancer.
- Author
-
Crawford N, Stott KJ, Sessler T, McCann C, McDaid W, Lees A, Latimer C, Fox JP, Munck JM, Smyth T, Shah A, Martins V, Lawler M, Dunne PD, Kerr EM, McDade SS, Coyle VM, and Longley DB
- Subjects
- Animals, Apoptosis, Biomarkers, Tumor genetics, Cell Proliferation, Colorectal Neoplasms metabolism, Colorectal Neoplasms pathology, Humans, Male, Mice, Mice, Inbred BALB C, Mice, Nude, Prognosis, Survival Rate, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Baculoviral IAP Repeat-Containing 3 Protein antagonists & inhibitors, Biomarkers, Tumor metabolism, Colorectal Neoplasms drug therapy, Gene Expression Regulation, Neoplastic drug effects, Inhibitor of Apoptosis Proteins antagonists & inhibitors, Morpholines pharmacology, Piperazines pharmacology, Pyrroles pharmacology
- Abstract
Inhibitors of apoptosis proteins (IAPs) are intracellular proteins, with important roles in regulating cell death, inflammation, and immunity. Here, we examined the clinical and therapeutic relevance of IAPs in colorectal cancer. We found that elevated expression of cIAP1 and cIAP2 (but not XIAP) significantly correlated with poor prognosis in patients with microsatellite stable (MSS) stage III colorectal cancer treated with 5-fluorouracil (5FU)-based adjuvant chemotherapy, suggesting their involvement in promoting chemoresistance. A novel IAP antagonist tolinapant (ASTX660) potently and rapidly downregulated cIAP1 in colorectal cancer models, demonstrating its robust on-target efficacy. In cells co-cultured with TNFα to mimic an inflammatory tumor microenvironment, tolinapant induced caspase-8-dependent apoptosis in colorectal cancer cell line models; however, the extent of apoptosis was limited because of inhibition by the caspase-8 paralogs FLIP and, unexpectedly, caspase-10. Importantly, tolinapant-induced apoptosis was augmented by FOLFOX in human colorectal cancer and murine organoid models in vitro and in vivo , due (at least in part) to FOLFOX-induced downregulation of class I histone deacetylases (HDAC), leading to acetylation of the FLIP-binding partner Ku70 and downregulation of FLIP. Moreover, the effects of FOLFOX could be phenocopied using the clinically relevant class I HDAC inhibitor, entinostat, which also induced acetylation of Ku70 and FLIP downregulation. Further analyses revealed that caspase-8 knockout RIPK3-positive colorectal cancer models were sensitive to tolinapant-induced necroptosis, an effect that could be exploited in caspase-8-proficient models using the clinically relevant caspase inhibitor emricasan. Our study provides evidence for immediate clinical exploration of tolinapant in combination with FOLFOX in poor prognosis MSS colorectal cancer with elevated cIAP1/2 expression., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
22. Dissecting the DNA binding landscape and gene regulatory network of p63 and p53.
- Author
-
Riege K, Kretzmer H, Sahm A, McDade SS, Hoffmann S, and Fischer M
- Subjects
- Binding Sites, Biomarkers, Tumor metabolism, Computational Biology, DNA genetics, Databases, Genetic, Gene Expression Regulation, Head and Neck Neoplasms genetics, Head and Neck Neoplasms metabolism, Head and Neck Neoplasms mortality, Humans, Prognosis, Protein Binding, Squamous Cell Carcinoma of Head and Neck genetics, Squamous Cell Carcinoma of Head and Neck metabolism, Squamous Cell Carcinoma of Head and Neck mortality, Transcription Factors metabolism, Tumor Suppressor Protein p53 metabolism, Tumor Suppressor Proteins metabolism, Biomarkers, Tumor genetics, DNA metabolism, Gene Regulatory Networks, Transcription Factors genetics, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Proteins genetics
- Abstract
The transcription factor p53 is the best-known tumor suppressor, but its sibling p63 is a master regulator of epidermis development and a key oncogenic driver in squamous cell carcinomas (SCC). Despite multiple gene expression studies becoming available, the limited overlap of reported p63-dependent genes has made it difficult to decipher the p63 gene regulatory network. Particularly, analyses of p63 response elements differed substantially among the studies. To address this intricate data situation, we provide an integrated resource that enables assessing the p63-dependent regulation of any human gene of interest. We use a novel iterative de novo motif search approach in conjunction with extensive ChIP-seq data to achieve a precise global distinction between p53-and p63-binding sites, recognition motifs, and potential co-factors. We integrate these data with enhancer:gene associations to predict p63 target genes and identify those that are commonly de-regulated in SCC representing candidates for prognosis and therapeutic interventions., Competing Interests: KR, HK, AS, SM, SH, MF No competing interests declared, (© 2020, Riege et al.)
- Published
- 2020
- Full Text
- View/download PDF
23. Tumor suppressor p53: from engaging DNA to target gene regulation.
- Author
-
Sammons MA, Nguyen TT, McDade SS, and Fischer M
- Subjects
- Animals, Binding Sites, Cell Line, Humans, Mice, Protein Binding, Rats, Chromatin metabolism, DNA metabolism, Gene Expression Regulation, Gene Regulatory Networks, Tumor Suppressor Protein p53 physiology
- Abstract
The p53 transcription factor confers its potent tumor suppressor functions primarily through the regulation of a large network of target genes. The recent explosion of next generation sequencing protocols has enabled the study of the p53 gene regulatory network (GRN) and underlying mechanisms at an unprecedented depth and scale, helping us to understand precisely how p53 controls gene regulation. Here, we discuss our current understanding of where and how p53 binds to DNA and chromatin, its pioneer-like role, and how this affects gene regulation. We provide an overview of the p53 GRN and the direct and indirect mechanisms through which p53 affects gene regulation. In particular, we focus on delineating the ubiquitous and cell type-specific network of regulatory elements that p53 engages; reviewing our understanding of how, where, and when p53 binds to DNA and the mechanisms through which these events regulate transcription. Finally, we discuss the evolution of the p53 GRN and how recent work has revealed remarkable differences between vertebrates, which are of particular importance to cancer researchers using mouse models., (© The Author(s) 2020. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2020
- Full Text
- View/download PDF
24. Chronic loss of STAG2 leads to altered chromatin structure contributing to de-regulated transcription in AML.
- Author
-
Smith JS, Lappin KM, Craig SG, Liberante FG, Crean CM, McDade SS, Thompson A, Mills KI, and Savage KI
- Subjects
- Cell Cycle Proteins genetics, Chromatin genetics, Humans, Mutation, Leukemia, Myeloid, Acute genetics, Myelodysplastic Syndromes
- Abstract
Background: The cohesin complex plays a major role in folding the human genome into 3D structural domains. Mutations in members of the cohesin complex are known early drivers of myelodysplastic syndromes (MDS) and acute myeloid leukaemia (AML), with STAG2 the most frequently mutated complex member., Methods: Here we use functional genomics (RNA-seq, ChIP-seq and HiChIP) to investigate the impact of chronic STAG2 loss on three-dimensional genome structure and transcriptional programming in a clinically relevant model of chronic STAG2 loss., Results: The chronic loss of STAG2 led to loss of smaller loop domains and the maintenance/formation of large domains that, in turn, led to altered genome compartmentalisation. These changes in genome structure resulted in altered gene expression, including deregulation of the HOXA locus and the MAPK signalling pathway, resulting in increased sensitivity to MEK inhibition., Conclusions: The altered genomic architecture driven by the chronic loss of STAG2 results in altered gene expression that may contribute to leukaemogenesis and may be therapeutically targeted.
- Published
- 2020
- Full Text
- View/download PDF
25. The SCF Skp2 ubiquitin ligase complex modulates TRAIL-R2-induced apoptosis by regulating FLIP(L).
- Author
-
Roberts JZ, Holohan C, Sessler T, Fox J, Crawford N, Riley JS, Khawaja H, Majkut J, Evergren E, Humphreys LM, Ferris J, Higgins C, Espona-Fiedler M, Moynagh P, McDade SS, and Longley DB
- Subjects
- Caspase 8 metabolism, Cell Line, Tumor, Cullin Proteins metabolism, Cyclopentanes pharmacology, Death Domain Receptor Signaling Adaptor Proteins metabolism, Humans, Protein Binding drug effects, Protein Interaction Mapping, Proteolysis drug effects, Pyrimidines pharmacology, Signal Transduction drug effects, TNF Receptor-Associated Factor 2 metabolism, TNF-Related Apoptosis-Inducing Ligand pharmacology, Apoptosis drug effects, CASP8 and FADD-Like Apoptosis Regulating Protein metabolism, Receptors, TNF-Related Apoptosis-Inducing Ligand metabolism, S-Phase Kinase-Associated Proteins metabolism
- Abstract
TRAIL-R2 (DR5) is a clinically-relevant therapeutic target and a key target for immune effector cells. Herein, we identify a novel interaction between TRAIL-R2 and the Skp1-Cullin-1-F-box (SCF) Cullin-Ring E3 Ubiquitin Ligase complex containing Skp2 (SCF
Skp2 ). We find that SCFSkp2 can interact with both TRAIL-R2's pre-ligand association complex (PLAC) and ligand-activated death-inducing signalling complex (DISC). Moreover, Cullin-1 interacts with TRAIL-R2 in its active NEDDylated form. Inhibiting Cullin-1's DISC recruitment using the NEDDylation inhibitor MLN4924 (Pevonedistat) or siRNA increased apoptosis induction in response to TRAIL. This correlated with enhanced levels of the caspase-8 regulator FLIP at the TRAIL-R2 DISC, particularly the long splice form, FLIP(L). We subsequently found that FLIP(L) (but not FLIP(S), caspase-8, nor the other core DISC component FADD) interacts with Cullin-1 and Skp2. Importantly, this interaction is enhanced when FLIP(L) is in its DISC-associated, C-terminally truncated p43-form. Prevention of FLIP(L) processing to its p43-form stabilises the protein, suggesting that by enhancing its interaction with SCFSkp2 , cleavage to the p43-form is a critical step in FLIP(L) turnover. In support of this, we found that silencing any of the components of the SCFSkp2 complex inhibits FLIP ubiquitination, while overexpressing Cullin-1/Skp2 enhances its ubiquitination in a NEDDylation-dependent manner. DISC recruitment of TRAF2, previously identified as an E3 ligase for caspase-8 at the DISC, was also enhanced when Cullin-1's recruitment was inhibited, although its interaction with Cullin-1 was found to be mediated indirectly via FLIP(L). Notably, the interaction of p43-FLIP(L) with Cullin-1 disrupts its ability to interact with FADD, caspase-8 and TRAF2. Collectively, our results suggest that processing of FLIP(L) to p43-FLIP(L) at the TRAIL-R2 DISC enhances its interaction with co-localised SCFSkp2 , leading to disruption of p43-FLIP(L)'s interactions with other DISC components and promoting its ubiquitination and degradation, thereby modulating TRAIL-R2-mediated apoptosis.- Published
- 2020
- Full Text
- View/download PDF
26. The pseudo-caspase FLIP(L) regulates cell fate following p53 activation.
- Author
-
Lees A, McIntyre AJ, Crawford NT, Falcone F, McCann C, Holohan C, Quinn GP, Roberts JZ, Sessler T, Gallagher PF, Gregg GMA, McAllister K, McLaughlin KM, Allen WL, Egan LJ, Ryan AE, Labonte-Wilson MJ, Dunne PD, Wappett M, Coyle VM, Johnston PG, Kerr EM, Longley DB, and McDade SS
- Subjects
- Acetylation, Antineoplastic Agents pharmacology, Apoptosis drug effects, Apoptosis genetics, Benzamides pharmacology, Caspase 8 metabolism, Cell Cycle drug effects, Cell Cycle genetics, Cell Line, Tumor, Drug Synergism, Gene Expression Regulation, Humans, Imidazoles metabolism, Models, Biological, Piperazines metabolism, Protein Binding, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-mdm2 metabolism, Pyridines pharmacology, Receptors, TNF-Related Apoptosis-Inducing Ligand metabolism, TNF-Related Apoptosis-Inducing Ligand metabolism, Tumor Suppressor Protein p53 genetics, CASP8 and FADD-Like Apoptosis Regulating Protein metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
p53 is the most frequently mutated, well-studied tumor-suppressor gene, yet the molecular basis of the switch from p53-induced cell-cycle arrest to apoptosis remains poorly understood. Using a combination of transcriptomics and functional genomics, we unexpectedly identified a nodal role for the caspase-8 paralog and only human pseudo-caspase, FLIP(L), in regulating this switch. Moreover, we identify FLIP(L) as a direct p53 transcriptional target gene that is rapidly up-regulated in response to Nutlin-3A, an MDM2 inhibitor that potently activates p53. Genetically or pharmacologically inhibiting expression of FLIP(L) using siRNA or entinostat (a clinically relevant class-I HDAC inhibitor) efficiently promoted apoptosis in colorectal cancer cells in response to Nutlin-3A, which otherwise predominantly induced cell-cycle arrest. Enhanced apoptosis was also observed when entinostat was combined with clinically relevant, p53-activating chemotherapy in vitro, and this translated into enhanced in vivo efficacy. Mechanistically, FLIP(L) inhibited p53-induced apoptosis by blocking activation of caspase-8 by the TRAIL-R2/DR5 death receptor; notably, this activation was not dependent on receptor engagement by its ligand, TRAIL. In the absence of caspase-8, another of its paralogs, caspase-10 (also transcriptionally up-regulated by p53), induced apoptosis in Nutlin-3A-treated, FLIP(L)-depleted cells, albeit to a lesser extent than in caspase-8-proficient cells. FLIP(L) depletion also modulated transcription of canonical p53 target genes, suppressing p53-induced expression of the cell-cycle regulator p21 and enhancing p53-induced up-regulation of proapoptotic PUMA. Thus, even in the absence of caspase-8/10, FLIP(L) silencing promoted p53-induced apoptosis by enhancing PUMA expression. Thus, we report unexpected, therapeutically relevant roles for FLIP(L) in determining cell fate following p53 activation., Competing Interests: The authors declare no competing interest.
- Published
- 2020
- Full Text
- View/download PDF
27. Pevonedistat (MLN4924): mechanism of cell death induction and therapeutic potential in colorectal cancer.
- Author
-
Ferris J, Espona-Fiedler M, Hamilton C, Holohan C, Crawford N, McIntyre AJ, Roberts JZ, Wappett M, McDade SS, Longley DB, and Coyle V
- Abstract
Pevonedistat (MLN4924), a selective inhibitor of the NEDD8-activating enzyme E1 regulatory subunit (NAE1), has demonstrated significant therapeutic potential in several malignancies. Although multiple mechanisms-of-action have been identified, how MLN4924 induces cell death and its potential as a combinatorial agent with standard-of-care (SoC) chemotherapy in colorectal cancer (CRC) remains largely undefined. In an effort to understand MLN4924-induced cell death in CRC, we identified p53 as an important mediator of the apoptotic response to MLN4924. We also identified roles for the extrinsic (TRAIL-R2/caspase-8) and intrinsic (BAX/BAK) apoptotic pathways in mediating the apoptotic effects of MLN4924 in CRC cells, as well as a role for BID, which modulates a cross-talk between these pathways. Depletion of the anti-apoptotic protein FLIP, which we identify as a novel mediator of resistance to MLN4924, enhanced apoptosis in a p53-, TRAIL-R2/DR5-, and caspase-8-dependent manner. Notably, TRAIL-R2 was involved in potentiating the apoptotic response to MLN4924 in the absence of FLIP, in a ligand-independent manner. Moreoever, when paired with SoC chemotherapies, MLN4924 demonstrated synergy with the irinotecan metabolite SN38. The cell death induced by MLN4924/SN38 combination was dependent on activation of mitochondria through BAX/BAK, but in a p53-independent manner, an important observation given the high frequency of TP53 mutation(s) in advanced CRC. These results uncover mechanisms of cell death induced by MLN4924 and suggest that this second-generation proteostasis-disrupting agent may have its most widespread activity in CRC, in combination with irinotecan-containing treatment regimens., Competing Interests: Conflict of interestThe authors declare that they have no conflict of interest., (© The Author(s) 2020.)
- Published
- 2020
- Full Text
- View/download PDF
28. A revised model of TRAIL-R2 DISC assembly explains how FLIP(L) can inhibit or promote apoptosis.
- Author
-
Humphreys LM, Fox JP, Higgins CA, Majkut J, Sessler T, McLaughlin K, McCann C, Roberts JZ, Crawford NT, McDade SS, Scott CJ, Harrison T, and Longley DB
- Subjects
- Caspase 8 genetics, Caspase 8 metabolism, Humans, Receptors, TNF-Related Apoptosis-Inducing Ligand, Signal Transduction, TNF-Related Apoptosis-Inducing Ligand genetics, Apoptosis genetics, CASP8 and FADD-Like Apoptosis Regulating Protein genetics
- Abstract
The long FLIP splice form FLIP(L) can act as both an inhibitor and promoter of caspase-8 at death-inducing signalling complexes (DISCs) formed by death receptors such as TRAIL-R2 and related intracellular complexes such as the ripoptosome. Herein, we describe a revised DISC assembly model that explains how FLIP(L) can have these opposite effects by defining the stoichiometry (with respect to caspase-8) at which it converts from being anti- to pro-apoptotic at the DISC. We also show that in the complete absence of FLIP(L), procaspase-8 activation at the TRAIL-R2 DISC has significantly slower kinetics, although ultimately the extent of apoptosis is significantly greater. This revised model of DISC assembly also explains why FLIP's recruitment to the TRAIL-R2 DISC is impaired in the absence of caspase-8 despite showing that it can interact with the DISC adaptor protein FADD and why the short FLIP splice form FLIP(S) is the more potent inhibitor of DISC-mediated apoptosis., (© 2020 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2020
- Full Text
- View/download PDF
29. Comparison of Molecular Assays for HPV Testing in Oropharyngeal Squamous Cell Carcinomas: A Population-Based Study in Northern Ireland.
- Author
-
Craig SG, Anderson LA, Moran M, Graham L, Currie K, Rooney K, Robinson M, Bingham V, Cuschieri KS, McQuaid S, Schache AG, Jones TM, McCance D, Salto-Tellez M, McDade SS, and James JA
- Subjects
- Age Factors, Alphapapillomavirus genetics, Alphapapillomavirus immunology, Biomarkers, Tumor analysis, Biomarkers, Tumor immunology, Cyclin-Dependent Kinase Inhibitor p16 analysis, Cyclin-Dependent Kinase Inhibitor p16 immunology, DNA, Viral isolation & purification, Female, Follow-Up Studies, Humans, Immunohistochemistry, In Situ Hybridization, Kaplan-Meier Estimate, Male, Middle Aged, Neoplasm Staging, Northern Ireland epidemiology, Oropharyngeal Neoplasms immunology, Oropharyngeal Neoplasms pathology, Oropharyngeal Neoplasms virology, Papillomavirus Infections mortality, Papillomavirus Infections pathology, Papillomavirus Infections virology, Prognosis, RNA, Viral isolation & purification, Retrospective Studies, Sensitivity and Specificity, Squamous Cell Carcinoma of Head and Neck mortality, Squamous Cell Carcinoma of Head and Neck pathology, Squamous Cell Carcinoma of Head and Neck virology, Alphapapillomavirus isolation & purification, Human Papillomavirus DNA Tests methods, Oropharyngeal Neoplasms diagnosis, Papillomavirus Infections diagnosis, Squamous Cell Carcinoma of Head and Neck diagnosis
- Abstract
Background: Determination of human papillomavirus (HPV) status has become clinically relevant for patient stratification under UICC TNM8 staging. Within the United Kingdom, a combination of p16 IHC and HPV DNA-ISH is recommended for classifying HPV status. This study will assess a series of clinically applicable second-line molecular tests to run in combination with p16 IHC to optimally determine HPV status., Methods: The ability of HPV RNA-ISH, HPV DNA-ISH, and HPV DNA-PCR to identify p16-positive/HPV-positive patients was investigated in a population-based oropharyngeal squamous cell carcinoma (OPSCC) cohort of patients diagnosed in Northern Ireland from 2000 to 2011., Results: Only 41% of the Northern Irish OPSCC patient population was associated with HPV-driven carcinogenesis. Both ISH assays were more specific than the DNA-PCR assay (100% and 95% vs. 67%) and were less likely to be affected by preanalytic factors such as increasing block age. A pooled HPV genotype probe for RNA-ISH was found to be the most accurate molecular assay assessed (95% accuracy) when compared with p16 positivity., Conclusions: Our study demonstrates the advantage of tissue-based molecular assays when determining HPV status in retrospective samples. Specifically, we demonstrate the enhanced sensitivity and specificity of ISH techniques compared with PCR-based methodology when working with formalin-fixed paraffin-embedded tissue, and found HPV RNA-ISH to be the most effective assay for determining HPV status., Impact: As p16 IHC is a relatively inexpensive, accessible, and sensitive test for stratifying patients by HPV status, this study finds that more patients would benefit from first-line p16 IHC followed by specific HPV testing using HPV RNA-ISH to confirm HPV status., (©2019 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
30. ACE: A Workbench Using Evolutionary Genetic Algorithms for Analyzing Association in TCGA.
- Author
-
Gilmore AR, Alderdice M, Savage KI, O'Reilly PG, Roddy AC, Dunne PD, Lawler M, McDade SS, Waugh DJ, and McArt DG
- Subjects
- Cohort Studies, Female, Humans, Software, User-Computer Interface, Algorithms, Biomarkers, Tumor genetics, Breast Neoplasms genetics, Evolution, Molecular, Genomics methods, Transcriptome
- Abstract
Modern methods of acquiring molecular data have improved rapidly in recent years, making it easier for researchers to collect large volumes of information. However, this has increased the challenge of recognizing interesting patterns within the data. Atlas Correlation Explorer (ACE) is a user-friendly workbench for seeking associations between attributes in The Cancer Genome Atlas (TCGA) database. It allows any combination of clinical and genomic data streams to be searched using an evolutionary algorithm approach. To showcase ACE, we assessed which RNA sequencing transcripts were associated with estrogen receptor (ESR1) in the TCGA breast cancer cohort. The analysis revealed already well-established associations with XBP1 and FOXA1, but also identified a strong association with CT62, a potential immunotherapeutic target with few previous associations with breast cancer. In conclusion, ACE can produce results for very large searches in a short time and will serve as an increasingly useful tool for biomarker discovery in the big data era. SIGNIFICANCE: ACE uses an evolutionary algorithm approach to perform large searches for associations between any combinations of data in the TCGA database., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
31. Recommendations for determining HPV status in patients with oropharyngeal cancers under TNM8 guidelines: a two-tier approach.
- Author
-
Craig SG, Anderson LA, Schache AG, Moran M, Graham L, Currie K, Rooney K, Robinson M, Upile NS, Brooker R, Mesri M, Bingham V, McQuaid S, Jones T, McCance DJ, Salto-Tellez M, McDade SS, and James JA
- Subjects
- Adolescent, Adult, Child, Child, Preschool, Disease-Free Survival, Female, Gene Expression Regulation, Neoplastic genetics, Humans, Infant, Infant, Newborn, Male, Middle Aged, Neoplasm Staging, Oropharyngeal Neoplasms mortality, Oropharyngeal Neoplasms pathology, Oropharyngeal Neoplasms virology, Papillomavirus Infections pathology, Papillomavirus Infections virology, Young Adult, Oropharyngeal Neoplasms genetics, Papillomavirus Infections genetics, Viral Proteins genetics
- Abstract
Background: TNM8 staging for oropharyngeal squamous cell carcinomas (OPSCC) surrogates p16 immunohistochemistry for HPV testing. Patients with p16+ OPSCC may lack HPV aetiology. Here, we evaluate the suitability of TNM8 staging for guiding prognosis in such patients., Methods: HPV status was ascertained using p16 immunohistochemistry and high-risk HPV RNA and DNA in situ hybridisation. Survival by stage in a cohort of OPSCC patients was evaluated using TNM7/TNM8 staging. Survival of p16+/HPV- patients was compared to p16 status., Results: TNM8 staging was found to improve on TNM7 (log rank p = 0·0190 for TNM8 compared with p = 0·0530 for TNM7) in p16+ patients. Patients who tested p16+ but were HPV- (n = 20) had significantly reduced five-year survival (33%) compared to p16+ patients (77%) but not p16- patients (35%). Cancer stage was reduced in 95% of p16+/HPV- patients despite having a mortality rate twice (HR 2.66 [95% CI: 1.37-5.15]) that of p16+/HPV+ patients under new TNM8 staging criteria., Conclusion: Given the significantly poorer survival of p16+/HPV- OPSCCs, these data provide compelling evidence for use of an HPV-specific test for staging classification. This has particular relevance in light of potential treatment de-escalation that could expose these patients to inappropriately reduced treatment intensity as treatment algorithms evolve.
- Published
- 2019
- Full Text
- View/download PDF
32. Cytoplasmic FLIP(S) and nuclear FLIP(L) mediate resistance of castrate-resistant prostate cancer to apoptosis induced by IAP antagonists.
- Author
-
McCann C, Crawford N, Majkut J, Holohan C, Armstrong CWD, Maxwell PJ, Ong CW, LaBonte MJ, McDade SS, Waugh DJ, and Longley DB
- Subjects
- Animals, Caspase 8 metabolism, Cell Line, Tumor, Cell Nucleus metabolism, Cytoplasm metabolism, Histone Deacetylases metabolism, Humans, Male, Mice, Mice, Inbred BALB C, Mice, SCID, NF-kappa B metabolism, PC-3 Cells, Prostatic Neoplasms, Castration-Resistant metabolism, THP-1 Cells, Tumor Necrosis Factor-alpha metabolism, Apoptosis drug effects, CASP8 and FADD-Like Apoptosis Regulating Protein metabolism, Cell Nucleus drug effects, Cytoplasm drug effects, Drug Resistance, Neoplasm drug effects, Histone Deacetylase Inhibitors pharmacology, Inhibitor of Apoptosis Proteins antagonists & inhibitors, Prostatic Neoplasms, Castration-Resistant drug therapy
- Abstract
Expression of tumor necrosis factor-α (TNFα) in the serum of prostate cancer patients is associated with poorer outcome and progression to castrate-resistant (CRPC) disease. TNFα promotes the activity of NFκB, which regulates a number of anti-apoptotic and proinflammatory genes, including those encoding the inhibitor of apoptosis proteins (IAPs); however, in the presence of IAP antagonists, TNFα can induce cell death. In the presence of recombinant or macrophage-derived TNFα, we found that IAP antagonists triggered degradation of cIAP1 and induced formation of Complex-IIb, consisting of caspase-8, FADD and RIPK1 in CRPC models; however, no, or modest levels of apoptosis were induced. This resistance was found to be mediated by both the long (L) and short (S) splice forms of the caspase-8 inhibitor, FLIP, another NFκB-regulated protein frequently overexpressed in CRPC. By decreasing FLIP expression at the post-transcriptional level in PC3 and DU145 cells (but not VCaP), the Class-I histone deacetylase (HDAC) inhibitor Entinostat promoted IAP antagonist-induced cell death in these models in a manner dependent on RIPK1, FADD and Caspase-8. Of note, Entinostat primarily targeted the nuclear rather than cytoplasmic pool of FLIP(L). While the cytoplasmic pool of FLIP(L) was highly stable, the nuclear pool was more labile and regulated by the Class-I HDAC target Ku70, which we have previously shown regulates FLIP stability. The efficacy of IAP antagonist (TL32711) and Entinostat combination and their effects on cIAP1 and FLIP respectively were confirmed in vivo, highlighting the therapeutic potential for targeting IAPs and FLIP in proinflammatory CRPC.
- Published
- 2018
- Full Text
- View/download PDF
33. ΔNp63γ/SRC/Slug Signaling Axis Promotes Epithelial-to-Mesenchymal Transition in Squamous Cancers.
- Author
-
Srivastava K, Pickard A, Craig SG, Quinn GP, Lambe SM, James JA, McDade SS, and McCance DJ
- Subjects
- Cell Line, Tumor, Cell Movement genetics, Cell Proliferation genetics, Epithelial-Mesenchymal Transition genetics, Gene Expression Regulation, Neoplastic genetics, Human papillomavirus 16 genetics, Human papillomavirus 16 pathogenicity, Humans, Keratinocytes virology, Neoplasm Invasiveness genetics, Neoplasm Invasiveness pathology, Oncogene Proteins, Viral genetics, Protein Isoforms genetics, Repressor Proteins genetics, Signal Transduction genetics, Squamous Cell Carcinoma of Head and Neck pathology, Squamous Cell Carcinoma of Head and Neck virology, Snail Family Transcription Factors genetics, Squamous Cell Carcinoma of Head and Neck genetics, Transcription Factors genetics, Tumor Suppressor Proteins genetics, src-Family Kinases genetics
- Abstract
Purpose: To investigate the regulation of epithelial-to-mesenchymal transition (EMT) in head and neck squamous cell carcinoma (HNSCC) and its importance in tumor invasion. Experimental Design: We use a three-dimensional invasive organotypic raft culture model of human foreskin keratinocytes expressing the E6/E7 genes of the human papilloma virus-16, coupled with bioinformatic and IHC analysis of patient samples to investigate the role played by EMT in invasion and identify effectors and upstream regulatory pathways. Results: We identify SNAI2 (Slug) as a critical effector of EMT-activated downstream of TP63 overexpression in HNSCC. Splice-form-specific depletion and rescue experiments further identify the ΔNp63γ isoform as both necessary and sufficient to activate the SRC signaling axis and SNAI2-mediated EMT and invasion. Moreover, elevated SRC levels are associated with poor outcome in patients with HNSCC in The Cancer Genome Atlas dataset. Importantly, the effects on EMT and invasions and SNAI2 expression can be reversed by genetic or pharmacologic inhibition of SRC. Conclusions: Overexpression of ΔNp63γ modulates cell invasion by inducing targetable SRC-Slug-evoked EMT in HNSCC, which can be reversed by inhibitors of the SRC signaling. Clin Cancer Res; 24(16); 3917-27. ©2018 AACR ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
34. Transcription Factor Activities Enhance Markers of Drug Sensitivity in Cancer.
- Author
-
Garcia-Alonso L, Iorio F, Matchan A, Fonseca N, Jaaks P, Peat G, Pignatelli M, Falcone F, Benes CH, Dunham I, Bignell G, McDade SS, Garnett MJ, and Saez-Rodriguez J
- Subjects
- Apoptosis, Cell Proliferation, Humans, Neoplasms genetics, Neoplasms pathology, Transcription Factors metabolism, Tumor Cells, Cultured, Antineoplastic Agents pharmacology, Biomarkers, Tumor genetics, Gene Expression Regulation, Neoplastic drug effects, Neoplasms drug therapy, Pharmacogenetics, Small Molecule Libraries pharmacology, Transcription Factors genetics
- Abstract
Transcriptional dysregulation induced by aberrant transcription factors (TF) is a key feature of cancer, but its global influence on drug sensitivity has not been examined. Here, we infer the transcriptional activity of 127 TFs through analysis of RNA-seq gene expression data newly generated for 448 cancer cell lines, combined with publicly available datasets to survey a total of 1,056 cancer cell lines and 9,250 primary tumors. Predicted TF activities are supported by their agreement with independent shRNA essentiality profiles and homozygous gene deletions, and recapitulate mutant-specific mechanisms of transcriptional dysregulation in cancer. By analyzing cell line responses to 265 compounds, we uncovered numerous TFs whose activity interacts with anticancer drugs. Importantly, combining existing pharmacogenomic markers with TF activities often improves the stratification of cell lines in response to drug treatment. Our results, which can be queried freely at dorothea.opentargets.io, offer a broad foundation for discovering opportunities to refine personalized cancer therapies. Significance: Systematic analysis of transcriptional dysregulation in cancer cell lines and patient tumor specimens offers a publicly searchable foundation to discover new opportunities to refine personalized cancer therapies. Cancer Res; 78(3); 769-80. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
35. Validation of a Metastatic Assay using biopsies to improve risk stratification in patients with prostate cancer treated with radical radiation therapy.
- Author
-
Jain S, Lyons CA, Walker SM, McQuaid S, Hynes SO, Mitchell DM, Pang B, Logan GE, McCavigan AM, O'Rourke D, McArt DG, McDade SS, Mills IG, Prise KM, Knight LA, Steele CJ, Medlow PW, Berge V, Katz B, Loblaw DA, Harkin DP, James JA, O'Sullivan JM, Kennedy RD, and Waugh DJ
- Subjects
- Aged, Cohort Studies, Disease-Free Survival, Gene Expression Profiling, Humans, Kaplan-Meier Estimate, Male, Middle Aged, Neoplasm Metastasis, Neoplasm Staging, Proportional Hazards Models, Prostatic Neoplasms genetics, Reproducibility of Results, Retrospective Studies, Risk Assessment methods, Risk Factors, Biopsy methods, Prostatic Neoplasms pathology, Prostatic Neoplasms radiotherapy
- Abstract
Background: Radiotherapy is an effective treatment of intermediate/high-risk locally advanced prostate cancer, however, >30% of patients relapse within 5 years. Clinicopathological parameters currently fail to identify patients prone to systemic relapse and those whom treatment intensification may be beneficial. The purpose of this study was to independently validate the performance of a 70-gene Metastatic Assay in a cohort of diagnostic biopsies from patients treated with radical radiotherapy and androgen deprivation therapy., Patients and Methods: A bridging cohort of prostate cancer diagnostic biopsy specimens was profiled to enable optimization of the Metastatic Assay threshold before further independent clinical validation in a cohort of diagnostic biopsies from patients treated with radical radiotherapy and androgen deprivation therapy. Multivariable Cox proportional hazard regression analysis was used to assess assay performance in predicting biochemical failure-free survival (BFFS) and metastasis-free survival (MFS)., Results: Gene expression analysis was carried out in 248 patients from the independent validation cohort and the Metastatic Assay applied. Ten-year MFS was 72% for Metastatic Assay positive patients and 94% for Metastatic Assay negative patients [HR = 3.21 (1.35-7.67); P = 0.003]. On multivariable analysis the Metastatic Assay remained predictive for development of distant metastases [HR = 2.71 (1.11-6.63); P = 0.030]. The assay retained independent prognostic performance for MFS when assessed with the Cancer of the Prostate Assessment Score (CAPRA) [HR = 3.23 (1.22-8.59); P = 0.019] whilst CAPRA itself was not significant [HR = 1.88, (0.52-6.77); P = 0.332]. A high concordance [100% (61.5-100)] for the assay result was noted between two separate foci taken from 11 tumours, whilst Gleason score had low concordance., Conclusions: The Metastatic Assay demonstrated significant prognostic performance in patients treated with radical radiotherapy both alone and independent of standard clinical and pathological variables. The Metastatic Assay could have clinical utility when deciding upon treatment intensification in high-risk patients. Genomic and clinical data are available as a public resource., (© The Author 2017. Published by Oxford University Press on behalf of the European Society for Medical Oncology.)
- Published
- 2018
- Full Text
- View/download PDF
36. Molecular Subgroup of Primary Prostate Cancer Presenting with Metastatic Biology.
- Author
-
Walker SM, Knight LA, McCavigan AM, Logan GE, Berge V, Sherif A, Pandha H, Warren AY, Davidson C, Uprichard A, Blayney JK, Price B, Jellema GL, Steele CJ, Svindland A, McDade SS, Eden CG, Foster C, Mills IG, Neal DE, Mason MD, Kay EW, Waugh DJ, Harkin DP, Watson RW, Clarke NW, and Kennedy RD
- Subjects
- Cluster Analysis, Genetic Predisposition to Disease, Humans, Least-Squares Analysis, Lymphatic Metastasis, Male, Multivariate Analysis, Phenotype, Proportional Hazards Models, Prostatic Neoplasms pathology, Retrospective Studies, Risk Assessment, Risk Factors, Time Factors, Treatment Outcome, Biomarkers, Tumor genetics, Lymph Node Excision adverse effects, Prostatectomy adverse effects, Prostatic Neoplasms genetics, Prostatic Neoplasms surgery, Transcriptome
- Abstract
Background: Approximately 4-25% of patients with early prostate cancer develop disease recurrence following radical prostatectomy., Objective: To identify a molecular subgroup of prostate cancers with metastatic potential at presentation resulting in a high risk of recurrence following radical prostatectomy., Design, Setting, and Participants: Unsupervised hierarchical clustering was performed using gene expression data from 70 primary resections, 31 metastatic lymph nodes, and 25 normal prostate samples. Independent assay validation was performed using 322 radical prostatectomy samples from four sites with a mean follow-up of 50.3 months., Outcome Measurements and Statistical Analysis: Molecular subgroups were identified using unsupervised hierarchical clustering. A partial least squares approach was used to generate a gene expression assay. Relationships with outcome (time to biochemical and metastatic recurrence) were analysed using multivariable Cox regression and log-rank analysis., Results and Limitations: A molecular subgroup of primary prostate cancer with biology similar to metastatic disease was identified. A 70-transcript signature (metastatic assay) was developed and independently validated in the radical prostatectomy samples. Metastatic assay positive patients had increased risk of biochemical recurrence (multivariable hazard ratio [HR] 1.62 [1.13-2.33]; p=0.0092) and metastatic recurrence (multivariable HR=3.20 [1.76-5.80]; p=0.0001). A combined model with Cancer of the Prostate Risk Assessment post surgical (CAPRA-S) identified patients at an increased risk of biochemical and metastatic recurrence superior to either model alone (HR=2.67 [1.90-3.75]; p<0.0001 and HR=7.53 [4.13-13.73]; p<0.0001, respectively). The retrospective nature of the study is acknowledged as a potential limitation., Conclusions: The metastatic assay may identify a molecular subgroup of primary prostate cancers with metastatic potential., Patient Summary: The metastatic assay may improve the ability to detect patients at risk of metastatic recurrence following radical prostatectomy. The impact of adjuvant therapies should be assessed in this higher-risk population., (Copyright © 2017 European Association of Urology. Published by Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
37. Cancer-cell intrinsic gene expression signatures overcome intratumoural heterogeneity bias in colorectal cancer patient classification.
- Author
-
Dunne PD, Alderdice M, O'Reilly PG, Roddy AC, McCorry AMB, Richman S, Maughan T, McDade SS, Johnston PG, Longley DB, Kay E, McArt DG, and Lawler M
- Subjects
- Algorithms, Biomarkers, Tumor genetics, Cohort Studies, Gene Expression Profiling, Humans, Lymph Nodes pathology, Lymphatic Metastasis, Neoplasm Metastasis, Oligonucleotide Array Sequence Analysis, Prognosis, Transcriptome, Colorectal Neoplasms diagnosis, Colorectal Neoplasms metabolism, Gene Expression Regulation, Neoplastic
- Abstract
Stromal-derived intratumoural heterogeneity (ITH) has been shown to undermine molecular stratification of patients into appropriate prognostic/predictive subgroups. Here, using several clinically relevant colorectal cancer (CRC) gene expression signatures, we assessed the susceptibility of these signatures to the confounding effects of ITH using gene expression microarray data obtained from multiple tumour regions of a cohort of 24 patients, including central tumour, the tumour invasive front and lymph node metastasis. Sample clustering alongside correlative assessment revealed variation in the ability of each signature to cluster samples according to patient-of-origin rather than region-of-origin within the multi-region dataset. Signatures focused on cancer-cell intrinsic gene expression were found to produce more clinically useful, patient-centred classifiers, as exemplified by the CRC intrinsic signature (CRIS), which robustly clustered samples by patient-of-origin rather than region-of-origin. These findings highlight the potential of cancer-cell intrinsic signatures to reliably stratify CRC patients by minimising the confounding effects of stromal-derived ITH.
- Published
- 2017
- Full Text
- View/download PDF
38. Challenging the Cancer Molecular Stratification Dogma: Intratumoral Heterogeneity Undermines Consensus Molecular Subtypes and Potential Diagnostic Value in Colorectal Cancer.
- Author
-
Dunne PD, McArt DG, Bradley CA, O'Reilly PG, Barrett HL, Cummins R, O'Grady T, Arthur K, Loughrey MB, Allen WL, McDade SS, Waugh DJ, Hamilton PW, Longley DB, Kay EW, Johnston PG, Lawler M, Salto-Tellez M, and Van Schaeybroeck S
- Subjects
- Gene Expression Regulation, Neoplastic, Humans, Lymphatic Metastasis, Neoplasm Staging, Organ Specificity genetics, Stromal Cells metabolism, Transcriptome, Biomarkers, Tumor, Colorectal Neoplasms diagnosis, Colorectal Neoplasms genetics, Gene Expression Profiling methods
- Abstract
Purpose: A number of independent gene expression profiling studies have identified transcriptional subtypes in colorectal cancer with potential diagnostic utility, culminating in publication of a colorectal cancer Consensus Molecular Subtype classification. The worst prognostic subtype has been defined by genes associated with stem-like biology. Recently, it has been shown that the majority of genes associated with this poor prognostic group are stromal derived. We investigated the potential for tumor misclassification into multiple diagnostic subgroups based on tumoral region sampled., Experimental Design: We performed multiregion tissue RNA extraction/transcriptomic analysis using colorectal-specific arrays on invasive front, central tumor, and lymph node regions selected from tissue samples from 25 colorectal cancer patients., Results: We identified a consensus 30-gene list, which represents the intratumoral heterogeneity within a cohort of primary colorectal cancer tumors. Using a series of online datasets, we showed that this gene list displays prognostic potential HR = 2.914 (confidence interval 0.9286-9.162) in stage II/III colorectal cancer patients, but in addition, we demonstrated that these genes are stromal derived, challenging the assumption that poor prognosis tumors with stem-like biology have undergone a widespread epithelial-mesenchymal transition. Most importantly, we showed that patients can be simultaneously classified into multiple diagnostically relevant subgroups based purely on the tumoral region analyzed., Conclusions: Gene expression profiles derived from the nonmalignant stromal region can influence assignment of colorectal cancer transcriptional subtypes, questioning the current molecular classification dogma and highlighting the need to consider pathology sampling region and degree of stromal infiltration when employing transcription-based classifiers to underpin clinical decision making in colorectal cancer. Clin Cancer Res; 22(16); 4095-104. ©2016 AACRSee related commentary by Morris and Kopetz, p. 3989., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
39. A chromatin-independent role of Polycomb-like 1 to stabilize p53 and promote cellular quiescence.
- Author
-
Brien GL, Healy E, Jerman E, Conway E, Fadda E, O'Donovan D, Krivtsov AV, Rice AM, Kearney CJ, Flaus A, McDade SS, Martin SJ, McLysaght A, O'Connell DJ, Armstrong SA, and Bracken AP
- Subjects
- Animals, Cell Proliferation genetics, Cells, Cultured, Chromatin metabolism, Cyclin-Dependent Kinase Inhibitor p16 genetics, Cyclin-Dependent Kinase Inhibitor p16 metabolism, DNA-Binding Proteins genetics, E2F Transcription Factors metabolism, Humans, Mice, Polycomb-Group Proteins genetics, Protein Binding, Protein Stability, Protein Structure, Tertiary genetics, DNA-Binding Proteins metabolism, Gene Expression Regulation, Neoplastic, Polycomb-Group Proteins metabolism, Transcription Factors metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
Polycomb-like proteins 1-3 (PCL1-3) are substoichiometric components of the Polycomb-repressive complex 2 (PRC2) that are essential for association of the complex with chromatin. However, it remains unclear why three proteins with such apparent functional redundancy exist in mammals. Here we characterize their divergent roles in both positively and negatively regulating cellular proliferation. We show that while PCL2 and PCL3 are E2F-regulated genes expressed in proliferating cells, PCL1 is a p53 target gene predominantly expressed in quiescent cells. Ectopic expression of any PCL protein recruits PRC2 to repress the INK4A gene; however, only PCL2 and PCL3 confer an INK4A-dependent proliferative advantage. Remarkably, PCL1 has evolved a PRC2- and chromatin-independent function to negatively regulate proliferation. We show that PCL1 binds to and stabilizes p53 to induce cellular quiescence. Moreover, depletion of PCL1 phenocopies the defects in maintaining cellular quiescence associated with p53 loss. This newly evolved function is achieved by the binding of the PCL1 N-terminal PHD domain to the C-terminal domain of p53 through two unique serine residues, which were acquired during recent vertebrate evolution. This study illustrates the functional bifurcation of PCL proteins, which act in both a chromatin-dependent and a chromatin-independent manner to regulate the INK4A and p53 pathways., (© 2015 Brien et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2015
- Full Text
- View/download PDF
40. HPV16 Down-Regulates the Insulin-Like Growth Factor Binding Protein 2 to Promote Epithelial Invasion in Organotypic Cultures.
- Author
-
Pickard A, McDade SS, McFarland M, McCluggage WG, Wheeler CM, and McCance DJ
- Subjects
- Cells, Cultured, Down-Regulation, Female, Humans, Oncogene Proteins, Viral metabolism, Papillomavirus E7 Proteins genetics, Repressor Proteins metabolism, Transcription, Genetic genetics, Uterine Cervical Neoplasms metabolism, Uterine Cervical Neoplasms pathology, Uterine Cervical Neoplasms virology, Epithelial Cells metabolism, Human papillomavirus 16 genetics, Insulin-Like Growth Factor Binding Protein 2 metabolism
- Abstract
Cervical cancer is a multi-stage disease caused by human papillomaviruses (HPV) infection of cervical epithelial cells, but the mechanisms regulating disease progression are not clearly defined. Using 3-dimensional organotypic cultures, we demonstrate that HPV16 E6 and E7 proteins alter the secretome of primary human keratinocytes resulting in local epithelial invasion. Mechanistically, absence of the IGF-binding protein 2 (IGFBP2) caused increases in IGFI/II signalling and through crosstalk with KGF/FGFR2b/AKT, cell invasion. Repression of IGFBP2 is mediated by histone deacetylation at the IGFBP2 promoter and was reversed by treatment with histone deacetylase (HDAC) inhibitors. Our in vitro findings were confirmed in 50 invasive cancers and 79 cervical intra-epithelial neoplastic lesions caused by HPV16 infection, where IGFBP2 levels were reduced with increasing disease severity. In summary, the loss of IGFBP2 is associated with progression of premalignant disease, and sensitises cells to pro-invasive IGF signalling, and together with stromal derived factors promotes epithelial invasion.
- Published
- 2015
- Full Text
- View/download PDF
41. Genome-wide characterization reveals complex interplay between TP53 and TP63 in response to genotoxic stress.
- Author
-
McDade SS, Patel D, Moran M, Campbell J, Fenwick K, Kozarewa I, Orr NJ, Lord CJ, Ashworth AA, and McCance DJ
- Subjects
- Binding Sites, Cells, Cultured, Cisplatin toxicity, DNA Breaks, Double-Stranded, DNA Repair, Doxorubicin toxicity, Genome, Human, Humans, Keratinocytes drug effects, Keratinocytes metabolism, Transcription, Genetic, Mutagens toxicity, Stress, Physiological genetics, Transcription Factors metabolism, Tumor Suppressor Protein p53 metabolism, Tumor Suppressor Proteins metabolism
- Abstract
In response to genotoxic stress the TP53 tumour suppressor activates target gene expression to induce cell cycle arrest or apoptosis depending on the extent of DNA damage. These canonical activities can be repressed by TP63 in normal stratifying epithelia to maintain proliferative capacity or drive proliferation of squamous cell carcinomas, where TP63 is frequently overexpressed/amplified. Here we use ChIP-sequencing, integrated with microarray analysis, to define the genome-wide interplay between TP53 and TP63 in response to genotoxic stress in normal cells. We reveal that TP53 and TP63 bind to overlapping, but distinct cistromes of sites through utilization of distinctive consensus motifs and that TP53 is constitutively bound to a number of sites. We demonstrate that cisplatin and adriamycin elicit distinct effects on TP53 and TP63 binding events, through which TP53 can induce or repress transcription of an extensive network of genes by direct binding and/or modulation of TP63 activity. Collectively, this results in a global TP53-dependent repression of cell cycle progression, mitosis and DNA damage repair concomitant with activation of anti-proliferative and pro-apoptotic canonical target genes. Further analyses reveal that in the absence of genotoxic stress TP63 plays an important role in maintaining expression of DNA repair genes, loss of which results in defective repair., (© The Author(s) 2014. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2014
- Full Text
- View/download PDF
42. BRCA1 deficiency exacerbates estrogen-induced DNA damage and genomic instability.
- Author
-
Savage KI, Matchett KB, Barros EM, Cooper KM, Irwin GW, Gorski JJ, Orr KS, Vohhodina J, Kavanagh JN, Madden AF, Powell A, Manti L, McDade SS, Park BH, Prise KM, McIntosh SA, Salto-Tellez M, Richard DJ, Elliott CT, and Harkin DP
- Subjects
- BRCA1 Protein genetics, Breast Neoplasms chemically induced, Breast Neoplasms genetics, Breast Neoplasms metabolism, Cytochrome P-450 CYP1A1 genetics, Cytochrome P-450 CYP1A1 metabolism, DNA Repair, Estradiol analogs & derivatives, Estradiol pharmacology, Estrogens metabolism, Estrogens, Catechol pharmacology, Female, Genomic Instability, Humans, MCF-7 Cells, BRCA1 Protein deficiency, Breast drug effects, Breast physiology, DNA Breaks, Double-Stranded, Estrogens pharmacology
- Abstract
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., (©2014 American Association for Cancer Research.)
- Published
- 2014
- Full Text
- View/download PDF
43. Identification of a BRCA1-mRNA splicing complex required for efficient DNA repair and maintenance of genomic stability.
- Author
-
Savage KI, Gorski JJ, Barros EM, Irwin GW, Manti L, Powell AJ, Pellagatti A, Lukashchuk N, McCance DJ, McCluggage WG, Schettino G, Salto-Tellez M, Boultwood J, Richard DJ, McDade SS, and Harkin DP
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Basic-Leucine Zipper Transcription Factors genetics, Basic-Leucine Zipper Transcription Factors metabolism, Cell Survival radiation effects, DNA Damage, DNA Repair Enzymes genetics, DNA Repair Enzymes metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Exodeoxyribonucleases genetics, Exodeoxyribonucleases metabolism, Fanconi Anemia Complementation Group Proteins genetics, Fanconi Anemia Complementation Group Proteins metabolism, Genome, Human, HEK293 Cells, Humans, Phosphorylation, Protein Processing, Post-Translational, RNA Splicing, Radiation Tolerance, Repressor Proteins metabolism, Tumor Suppressor Proteins metabolism, BRCA1 Protein metabolism, DNA Repair, Genomic Instability, RNA, Messenger metabolism
- Abstract
Mutations within BRCA1 predispose carriers to a high risk of breast and ovarian cancers. BRCA1 functions to maintain genomic stability through the assembly of multiple protein complexes involved in DNA repair, cell-cycle arrest, and transcriptional regulation. Here, we report the identification of a DNA damage-induced BRCA1 protein complex containing BCLAF1 and other key components of the mRNA-splicing machinery. In response to DNA damage, this complex regulates pre-mRNA splicing of a number of genes involved in DNA damage signaling and repair, thereby promoting the stability of these transcripts/proteins. Further, we show that abrogation of this complex results in sensitivity to DNA damage, defective DNA repair, and genomic instability. Interestingly, mutations in a number of proteins found within this complex have been identified in numerous cancer types. These data suggest that regulation of splicing by the BRCA1-mRNA splicing complex plays an important role in the cellular response to DNA damage., (Copyright © 2014 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
44. Androgens and estrogens stimulate ribosome biogenesis in prostate and breast cancer cells in receptor dependent manner.
- Author
-
Ray S, Johnston R, Campbell DC, Nugent S, McDade SS, Waugh D, and Panov KI
- Subjects
- Androgen Antagonists pharmacology, Anilides pharmacology, Breast Neoplasms genetics, Cell Line, Tumor, Estradiol analogs & derivatives, Estradiol pharmacology, Estrogen Receptor Modulators pharmacology, Female, Fulvestrant, Humans, MCF-7 Cells, Male, Neoplasms, Hormone-Dependent genetics, Neoplasms, Hormone-Dependent metabolism, Nitriles pharmacology, Prostatic Neoplasms genetics, RNA, Neoplasm genetics, RNA, Neoplasm metabolism, RNA, Ribosomal genetics, RNA, Ribosomal metabolism, Receptors, Androgen metabolism, Receptors, Estrogen metabolism, Ribosomes drug effects, Signal Transduction, Tosyl Compounds pharmacology, Transcription, Genetic drug effects, Androgens metabolism, Breast Neoplasms metabolism, Estrogens metabolism, Prostatic Neoplasms metabolism, Ribosomes metabolism
- Abstract
Ribosome biogenesis is a fundamental cellular process intimately linked to cell growth and proliferation, which is upregulated in most of cancers especially in aggressive cancers. In breast and prostate cancers steroid hormone receptor signalling is the principal stimulus for cancer growth and progression. Here we investigated the link between estrogen and androgen receptor signalling and the initial stage of ribosome biogenesis - transcription of rRNA genes. We have discovered that oestrogen or androgen treatment can positively regulate rRNA synthesis in breast and prostate cancer cells respectively and that this effect is receptor dependent. This novel and interesting finding suggests a previously unidentified link between steroid hormone receptor signalling pathways and the regulation of ribosome biogenesis., (Copyright © 2013 Elsevier B.V. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
45. AKT in stromal fibroblasts controls invasion of epithelial cells.
- Author
-
Cichon AC, Pickard A, McDade SS, Sharpe DJ, Moran M, James JA, and McCance DJ
- Subjects
- Carcinogenesis metabolism, Carcinogenesis pathology, Cells, Cultured, Disease Progression, Epithelial Cells enzymology, Epithelial Cells pathology, Humans, Keratinocytes enzymology, Keratinocytes pathology, PTEN Phosphohydrolase genetics, PTEN Phosphohydrolase metabolism, Stromal Cells enzymology, Stromal Cells pathology, Tumor Microenvironment, Fibroblasts enzymology, Fibroblasts pathology, Oropharyngeal Neoplasms enzymology, Oropharyngeal Neoplasms pathology, Proto-Oncogene Proteins c-akt metabolism
- Abstract
The tumour microenvironment has an important role in cancer progression and recent reports have proposed that stromal AKT is activated and regulates tumourigenesis and invasion. We have shown, by immuno-fluorescent analysis of oro-pharyngeal cancer biopsies, an increase in AKT activity in tumour associated stromal fibroblasts compared to normal stromal fibroblasts. Using organotypic raft co-cultures, we show that activation of stromal AKT can induce the invasion of keratinocytes expressing the HPV type 16 E6 and E7 proteins, in a Keratinocyte Growth Factor (KGF) dependent manner. By depleting stromal fibroblasts of each of the three AKT isoforms independently, or through using isoform specific inhibitors, we determined that stromal AKT2 is an essential regulator of invasion and show in oro-pharyngeal cancers that AKT2 specific phosphorylation events are also identified in stromal fibroblasts. Depletion of stromal AKT2 inhibits epithelial invasion through activating a protective pathway counteracting KGF mediated invasions. AKT2 depletion in fibroblasts stimulates the cleavage and release of IL1B from stromal fibroblasts resulting in down-regulation of the KGF receptor (fibroblast growth factor receptor 2B (FGFR2B)) expression in the epithelium. We also show that high IL1B is associated with increased overall survival in a cohort of patients with oro-pharyngeal cancers. Our findings demonstrate the importance of stromal derived growth factors and cytokines in regulating the process of tumour cell invasion.
- Published
- 2013
- Full Text
- View/download PDF
46. Genome-wide association study identifies a common variant in RAD51B associated with male breast cancer risk.
- Author
-
Orr N, Lemnrau A, Cooke R, Fletcher O, Tomczyk K, Jones M, Johnson N, Lord CJ, Mitsopoulos C, Zvelebil M, McDade SS, Buck G, Blancher C, Trainer AH, James PA, Bojesen SE, Bokmand S, Nevanlinna H, Mattson J, Friedman E, Laitman Y, Palli D, Masala G, Zanna I, Ottini L, Giannini G, Hollestelle A, Ouweland AM, Novaković S, Krajc M, Gago-Dominguez M, Castelao JE, Olsson H, Hedenfalk I, Easton DF, Pharoah PD, Dunning AM, Bishop DT, Neuhausen SL, Steele L, Houlston RS, Garcia-Closas M, Ashworth A, and Swerdlow AJ
- Subjects
- Chromosomes, Human, Pair 14, Genetic Predisposition to Disease, Humans, Male, Polymorphism, Single Nucleotide, Risk Factors, White People, Breast Neoplasms, Male genetics, DNA-Binding Proteins genetics, Genome-Wide Association Study
- Abstract
We conducted a genome-wide association study of male breast cancer comprising 823 cases and 2,795 controls of European ancestry, with validation in independent sample sets totaling 438 cases and 474 controls. A SNP in RAD51B at 14q24.1 was significantly associated with male breast cancer risk (P = 3.02 × 10(-13); odds ratio (OR) = 1.57). We also refine association at 16q12.1 to a SNP within TOX3 (P = 3.87 × 10(-15); OR = 1.50).
- Published
- 2012
- Full Text
- View/download PDF
47. Genome-wide analysis of p63 binding sites identifies AP-2 factors as co-regulators of epidermal differentiation.
- Author
-
McDade SS, Henry AE, Pivato GP, Kozarewa I, Mitsopoulos C, Fenwick K, Assiotis I, Hakas J, Zvelebil M, Orr N, Lord CJ, Patel D, Ashworth A, and McCance DJ
- Subjects
- Binding Sites, Cell Differentiation, Cells, Cultured, Cleft Palate genetics, Gene Expression Regulation, Genome, Human, Humans, Keratinocytes cytology, Molecular Sequence Annotation, Regulatory Elements, Transcriptional, Transcription Factor AP-2 antagonists & inhibitors, Tumor Suppressor Protein p53 metabolism, Epidermal Cells, Keratinocytes metabolism, Transcription Factor AP-2 metabolism, Transcription Factors metabolism, Tumor Suppressor Proteins metabolism
- Abstract
The p63 transcription factor (TP63) is critical in development, growth and differentiation of stratifying epithelia. This is highlighted by the severity of congenital abnormalities caused by TP63 mutations in humans, the dramatic phenotypes in knockout mice and de-regulation of TP63 expression in neoplasia altering the tumour suppressive roles of the TP53 family. In order to define the normal role played by TP63 and provide the basis for better understanding how this network is perturbed in disease, we used chromatin immunoprecipitation combined with massively parallel sequencing (ChIP-seq) to identify >7500 high-confidence TP63-binding regions across the entire genome, in primary human neonatal foreskin keratinocytes (HFKs). Using integrative strategies, we demonstrate that only a subset of these sites are bound by TP53 in response to DNA damage. We identify a role for TP63 in transcriptional regulation of multiple genes genetically linked to cleft palate and identify AP-2alpha (TFAP2A) as a co-regulator of a subset of these genes. We further demonstrate that AP-2gamma (TFAP2C) can bind a subset of these regions and that acute depletion of either TFAP2A or TFAP2C alone is sufficient to reduce terminal differentiation of organotypic epidermal skin equivalents, indicating overlapping physiological functions with TP63.
- Published
- 2012
- Full Text
- View/download PDF
48. Expression of the SEPT9_i4 isoform confers resistance to microtubule-interacting drugs.
- Author
-
Chacko AD, McDade SS, Chanduloy S, Church SW, Kennedy R, Price J, Hall PA, and Russell SE
- Subjects
- Cell Line, Tumor, DNA Damage, Flow Cytometry, Humans, Ice, Polymerization drug effects, Protein Isoforms metabolism, Vinblastine pharmacology, Vinorelbine, Drug Resistance, Neoplasm drug effects, Microtubules drug effects, Microtubules metabolism, Paclitaxel pharmacology, Septins metabolism, Vinblastine analogs & derivatives
- Abstract
Background: The evolutionarily conserved septin family of genes encode GTP binding proteins involved in a variety of cellular functions including cytokinesis, apoptosis, membrane dynamics and vesicle trafficking. Septin proteins can form hetero-oligomeric complexes and interact with other proteins including actin and tubulin. The human SEPT9 gene on chromosome 17q25.3 has a complex genomic architecture with 18 different transcripts that can encode 15 distinct polypeptides. Two distinct transcripts with unique 5' ends (SEPT9_v4 and SEPT9_v4*) encode the same protein. In tumours the ratio of these transcripts changes with elevated levels of SEPT9_v4* mRNA, a transcript that is translated with enhanced efficiency leading to increased SEPT9_i4 protein., Methods: We have examined the effect of over-expression of SEPT9_i4 on the dynamics of microtubule polymer mass in cultured cells., Results: We show that the microtubule network in SEPT9_i4 over-expressing cells resists disruption by paclitaxel or cold incubation but also repolymerises tubulin more slowly after microtubule depolymerisation. Finally we show that SEPT9_i4 over-expressing cells have enhanced survival in the presence of clinically relevant microtubule acting drugs but not after treatment with DNAinteracting agents., Conclusions: Given that SEPT9 over-expression is seen in diverse tumours and in particular ovarian and breast cancer, such data indicate that SEPT9_v4 expression may be clinically relevant and contribute to some forms of drug resistance.
- Published
- 2012
- Full Text
- View/download PDF
49. Profiling of the BRCA1 transcriptome through microarray and ChIP-chip analysis.
- Author
-
Gorski JJ, Savage KI, Mulligan JM, McDade SS, Blayney JK, Ge Z, and Harkin DP
- Subjects
- BRCA1 Protein antagonists & inhibitors, BRCA1 Protein metabolism, Cell Line, Tumor, Chromatin Immunoprecipitation, Gene Expression Profiling, Gene Expression Regulation, Humans, Oligonucleotide Array Sequence Analysis, Stress, Physiological genetics, Transcription Factors metabolism, BRCA1 Protein physiology, Transcriptome
- Abstract
A role for BRCA1 in the direct and indirect regulation of transcription is well established. However, a comprehensive view of the degree to which BRCA1 impacts transcriptional regulation on a genome-wide level has not been defined. We performed genome-wide expression profiling and ChIP-chip analysis, comparison of which revealed that although BRCA1 depletion results in transcriptional changes in 1294 genes, only 44 of these are promoter bound by BRCA1. However, 27% of these transcripts were linked to transcriptional regulation possibly explaining the large number of indirect transcriptional changes observed by microarray analysis. We show that no specific consensus sequence exists for BRCA1 DNA binding but rather demonstrate the presence of a number of known and novel transcription factor (TF)- binding sites commonly found on BRCA1 bound promoters. Co-immunoprecipitations confirmed that BRCA1 interacts with a number of these TFs including AP2-α, PAX2 and ZF5. Finally, we show that BRCA1 is bound to a subset of promoters of genes that are not altered by BRCA1 loss, but are transcriptionally regulated in a BRCA1-dependent manner upon DNA damage. These data suggest a model, whereby BRCA1 is present on defined promoters as part of an inactive complex poised to respond to various genotoxic stimuli., (© The Author(s) 2011. Published by Oxford University Press.)
- Published
- 2011
- Full Text
- View/download PDF
50. Evidence for alteration of EZH2, BMI1, and KDM6A and epigenetic reprogramming in human papillomavirus type 16 E6/E7-expressing keratinocytes.
- Author
-
Hyland PL, McDade SS, McCloskey R, Dickson GJ, Arthur K, McCance DJ, and Patel D
- Subjects
- Cells, Cultured, Enhancer of Zeste Homolog 2 Protein, Gene Expression Profiling, Host-Pathogen Interactions, Humans, Keratinocytes virology, Polycomb Repressive Complex 1, Polycomb Repressive Complex 2, DNA-Binding Proteins metabolism, Epigenesis, Genetic, Histone Demethylases metabolism, Human papillomavirus 16 pathogenicity, Nuclear Proteins metabolism, Oncogene Proteins, Viral metabolism, Papillomavirus E7 Proteins metabolism, Proto-Oncogene Proteins metabolism, Repressor Proteins metabolism, Transcription Factors metabolism
- Abstract
A number of epigenetic alterations occur in both the virus and host cellular genomes during human papillomavirus (HPV)-associated carcinogenesis, and investigations of such alterations, including changes in chromatin proteins and histone modifications, have the potential to lead to therapeutic epigenetic reversion. We report here that transformed HPV16 E6/E7-expressing primary human foreskin keratinocytes (HFKs) (E6/E7 cells) demonstrate increased expression of the PRC2 methyltransferase EZH2 at both the mRNA and protein levels but do not exhibit the expected increase in trimethylated H3K27 (H3K27me3) compared to normal keratinocytes. In contrast, these cells show a reduction in global H3K27me3 levels in vitro, as well as upregulation of the KDM6A demethylase. We further show for the first time that transformation with the HPV16 E6 and E7 oncogenes also results in an increase in phosphorylated EZH2 serine 21 (P-EZH2-Ser21), mediated by active Akt, and in a downregulation of the PRC1 protein BMI1 in these cells. High-grade squamous cervical intraepithelial lesions also showed a loss of H3K27me3 in the presence of increased expression of EZH2. Correlating with the loss of H3K27me3, E6/E7 cells exhibited derepression of specific EZH2-, KMD6A-, and BMI1-targeted HOX genes. These results suggest that the observed reduction in H3K27me3 may be due to a combination of reduced activities/levels of specific polycomb proteins and increases in demethylases. The dysregulation of multiple chromatin proteins resulting in the loss of global H3K27me3 and the transcriptional reprogramming in HPV16 E6/E7-infected cells could provide an epigenetic signature associated with risk and/or progression of HPV16-associated cancers, as well as the potential for epigenetic reversion in the future.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.