51 results on '"Aengus Stewart"'
Search Results
2. The combination of CHK1 inhibitor with G-CSF overrides cytarabine resistance in human acute myeloid leukemia
- Author
-
Alessandro Di Tullio, Kevin Rouault-Pierre, Ander Abarrategi, Syed Mian, William Grey, John Gribben, Aengus Stewart, Elizabeth Blackwood, and Dominique Bonnet
- Subjects
Science - Abstract
Overriding cytarabine resistance in AML remains an unmet medical need. Here, the authors show that the CHK1 inhibitor GDC-0575 in combination with cytarabine and G-CSF has a significant anti-leukemic effect without toxicity to normal marrow stem and progenitor cells.
- Published
- 2017
- Full Text
- View/download PDF
3. Multiomic Analysis of the UV-Induced DNA Damage Response
- Author
-
Stefan Boeing, Laura Williamson, Vesela Encheva, Ilaria Gori, Rebecca E. Saunders, Rachael Instrell, Ozan Aygün, Marta Rodriguez-Martinez, Juston C. Weems, Gavin P. Kelly, Joan W. Conaway, Ronald C. Conaway, Aengus Stewart, Michael Howell, Ambrosius P. Snijders, and Jesper Q. Svejstrup
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: In order to facilitate the identification of factors and pathways in the cellular response to UV-induced DNA damage, several descriptive proteomic screens and a functional genomics screen were performed in parallel. Numerous factors could be identified with high confidence when the screen results were superimposed and interpreted together, incorporating biological knowledge. A searchable database, bioLOGIC, which provides access to relevant information about a protein or process of interest, was established to host the results and facilitate data mining. Besides uncovering roles in the DNA damage response for numerous proteins and complexes, including Integrator, Cohesin, PHF3, ASC-1, SCAF4, SCAF8, and SCAF11, we uncovered a role for the poorly studied, melanoma-associated serine/threonine kinase 19 (STK19). Besides effectively uncovering relevant factors, the multiomic approach also provides a systems-wide overview of the diverse cellular processes connected to the transcription-related DNA damage response. : Boeing et al. investigate the UV-induced DNA damage response by combining a range of proteomic and genomic screens. A function in this response for the melanoma driver STK19 as well as a number of other factors are uncovered.
- Published
- 2016
- Full Text
- View/download PDF
4. Pharmacological Bypass of Cockayne Syndrome B Function in Neuronal Differentiation
- Author
-
Yuming Wang, Jace Jones-Tabah, Probir Chakravarty, Aengus Stewart, Alysson Muotri, Rebecca R. Laposa, and Jesper Q. Svejstrup
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Cockayne syndrome (CS) is a severe neurodevelopmental disorder characterized by growth abnormalities, premature aging, and photosensitivity. Mutation of Cockayne syndrome B (CSB) affects neuronal gene expression and differentiation, so we attempted to bypass its function by expressing downstream target genes. Intriguingly, ectopic expression of Synaptotagmin 9 (SYT9), a key component of the machinery controlling neurotrophin release, bypasses the need for CSB in neuritogenesis. Importantly, brain-derived neurotrophic factor (BDNF), a neurotrophin implicated in neuronal differentiation and synaptic modulation, and pharmacological mimics such as 7,8-dihydroxyflavone and amitriptyline can compensate for CSB deficiency in cell models of neuronal differentiation as well. SYT9 and BDNF are downregulated in CS patient brain tissue, further indicating that sub-optimal neurotrophin signaling underlies neurological defects in CS. In addition to shedding light on cellular mechanisms underlying CS and pointing to future avenues for pharmacological intervention, these data suggest an important role for SYT9 in neuronal differentiation.
- Published
- 2016
- Full Text
- View/download PDF
5. Distinct modes of SMAD2 chromatin binding and remodeling shape the transcriptional response to NODAL/Activin signaling
- Author
-
Davide M Coda, Tessa Gaarenstroom, Philip East, Harshil Patel, Daniel S J Miller, Anna Lobley, Nik Matthews, Aengus Stewart, and Caroline S Hill
- Subjects
SMAD2 ,chromatin remodeling ,NODAL/Activin ,transcription ,signaling dynamics ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
NODAL/Activin signaling orchestrates key processes during embryonic development via SMAD2. How SMAD2 activates programs of gene expression that are modulated over time however, is not known. Here we delineate the sequence of events that occur from SMAD2 binding to transcriptional activation, and the mechanisms underlying them. NODAL/Activin signaling induces dramatic chromatin landscape changes, and a dynamic transcriptional network regulated by SMAD2, acting via multiple mechanisms. Crucially we have discovered two modes of SMAD2 binding. SMAD2 can bind pre-acetylated nucleosome-depleted sites. However, it also binds to unacetylated, closed chromatin, independently of pioneer factors, where it induces nucleosome displacement and histone acetylation. For a subset of genes, this requires SMARCA4. We find that long term modulation of the transcriptional responses requires continued NODAL/Activin signaling. Thus SMAD2 binding does not linearly equate with transcriptional kinetics, and our data suggest that SMAD2 recruits multiple co-factors during sustained signaling to shape the downstream transcriptional program.
- Published
- 2017
- Full Text
- View/download PDF
6. A simple biophysical model emulates budding yeast chromosome condensation
- Author
-
Tammy MK Cheng, Sebastian Heeger, Raphaël AG Chaleil, Nik Matthews, Aengus Stewart, Jon Wright, Carmay Lim, Paul A Bates, and Frank Uhlmann
- Subjects
chromosome architecture ,mitosis ,condensin ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Mitotic chromosomes were one of the first cell biological structures to be described, yet their molecular architecture remains poorly understood. We have devised a simple biophysical model of a 300 kb-long nucleosome chain, the size of a budding yeast chromosome, constrained by interactions between binding sites of the chromosomal condensin complex, a key component of interphase and mitotic chromosomes. Comparisons of computational and experimental (4C) interaction maps, and other biophysical features, allow us to predict a mode of condensin action. Stochastic condensin-mediated pairwise interactions along the nucleosome chain generate native-like chromosome features and recapitulate chromosome compaction and individualization during mitotic condensation. Higher order interactions between condensin binding sites explain the data less well. Our results suggest that basic assumptions about chromatin behavior go a long way to explain chromosome architecture and are able to generate a molecular model of what the inside of a chromosome is likely to look like.
- Published
- 2015
- Full Text
- View/download PDF
7. An Important Role for RPRD1B in the Heat Shock Response
- Author
-
Simona Cugusi, Prashanth Kumar Bajpe, Richard Mitter, Harshil Patel, Aengus Stewart, and Jesper Q. Svejstrup
- Subjects
Proteomics ,Mammals ,Cell Biology ,HSF1 ,heat shock ,Heat Shock Transcription Factors ,HSF ,Animals ,RPRD1B ,HSP70 Heat-Shock Proteins ,RNA polymerase II ,transcription ,Molecular Biology ,Heat-Shock Response ,Heat-Shock Proteins ,Transcription Factors - Abstract
During the heat shock response (HSR), heat shock factor (HSF1 in mammals) binds to target gene promoters, resulting in increased expression of heat shock proteins that help maintain protein homeostasis and ensure cell survival. Besides HSF1, only a relatively few transcription factors with a specific role in ensuring correctly regulated gene expression during the HSR have been described. Here, we use proteomic and genomic (CRISPR) screening to identify a role for RPRD1B in the response to heat shock. Indeed, cells depleted for RPRD1B are heat shock sensitive and show decreased expression of key heat shock proteins (HSPs). These results add to our understanding of the connection between basic gene expression mechanisms and the HSR.
- Published
- 2022
- Full Text
- View/download PDF
8. Heat shock induces premature transcript termination and reconfigures the human transcriptome
- Author
-
Simona Cugusi, Richard Mitter, Gavin P. Kelly, Jane Walker, Zhong Han, Paola Pisano, Michael Wierer, Aengus Stewart, and Jesper Q. Svejstrup
- Subjects
Model organisms ,U1 snRNA ,Gene Expression ,CDK9 ,CPSF3 ,elongation ,Biochemistry & Proteomics ,Polyadenylation ,transcriptional repression ,Humans ,RNA, Messenger ,Molecular Biology ,Computational & Systems Biology ,Chemical Biology & High Throughput ,premature termination ,SCAF8 ,Genome Integrity & Repair ,alternative polyadenylation ,SCAF4 ,Cell Biology ,telescripting ,Tumour Biology ,pTEFb ,heat shock ,cryptic polyadenylation sites ,RNA Polymerase II ,Transcriptome ,Genetics & Genomics ,Heat-Shock Response ,pause release ,TT-seq - Abstract
The heat shock (HS) response involves rapid induction of HS genes, whereas transcriptional repression is established more slowly at most other genes. Previous data suggested that such repression results from inhibition of RNA polymerase II (RNAPII) pause release, but here, we show that HS strongly affects other phases of the transcription cycle. Intriguingly, while elongation rates increase upon HS, processivity markedly decreases, so that RNAPII frequently fails to reach the end of genes. Indeed, HS results in widespread premature transcript termination at cryptic, intronic polyadenylation (IPA) sites near gene 5′-ends, likely via inhibition of U1 telescripting. This results in dramatic reconfiguration of the human transcriptome with production of new, previously unannotated, short mRNAs that accumulate in the nucleus. Together, these results shed new light on the basic transcription mechanisms induced by growth at elevated temperature and show that a genome-wide shift toward usage of IPA sites can occur under physiological conditions.
- Published
- 2022
- Full Text
- View/download PDF
9. Integrated Genome and Transcriptome Analyses Reveal the Mechanism of Genome Instability in Ataxia with Oculomotor Apraxia 2
- Author
-
Probir Chakravarty, Matthew M. Edwards, Becherel O, Benitez A, Fengtang Yang, Kanagaraj R, Stephen C. West, Amnon Koren, Beiyuan Fu, Aengus Stewart, Lavin Mf, Richard Mitter, and Theodoros Kantidakis
- Subjects
Genome instability ,genetic structures ,Cerebellar Ataxia ,DNA Repair ,ataxia with oculomotor apraxia ,Apraxias ,Primary Cell Culture ,Biology ,Genomic Instability ,Cockayne syndrome ,Cell Line ,Mice ,Chromosomal Instability ,Chromosome instability ,medicine ,Animals ,Humans ,Spinocerebellar Ataxias ,Oculomotor apraxia ,Promoter Regions, Genetic ,DNA repair senataxin ,Gene ,Genetics ,Multidisciplinary ,Gene Expression Profiling ,DNA Helicases ,Mouse Embryonic Stem Cells ,Neurodegenerative Diseases ,Promoter ,Genomics ,Cell Biology ,Chromosome Fragility ,Biological Sciences ,medicine.disease ,Multifunctional Enzymes ,transcription stress ,Chromosome Fragile Site ,Mutation ,Ataxia ,Transcriptome ,RNA Helicases - Abstract
Significance Ataxia with oculomotor apraxia (AOA) is a progressive neurodegenerative disease characterized by early‐onset autosomal recessive cerebellar ataxia with oculomotor apraxia, peripheral axonal neuropathy, and impaired motor functions. The AOA-2 subgroup results from mutations in an RNA/DNA helicase, Senataxin, which is encoded by the SETX gene. Here, we carried out integrated genome and transcriptome analyses of cell lines derived from individuals with AOA2, as well as CRISPR/Cas9 generated SETX knockouts, and observed genome-wide chromosome fragility. Genome instability was caused by increased transcription stress and the accumulation of RNA/DNA hybrids near gene promotors, resulting in aberrant DNA repair that led to changes in gene-expression profiles. The results indicate that SETX-defective cells exhibit transcription stress that leads to chromosome fragility., Mutations in the SETX gene, which encodes Senataxin, are associated with the progressive neurodegenerative diseases ataxia with oculomotor apraxia 2 (AOA2) and amyotrophic lateral sclerosis 4 (ALS4). To identify the causal defect in AOA2, patient-derived cells and SETX knockouts (human and mouse) were analyzed using integrated genomic and transcriptomic approaches. A genome-wide increase in chromosome instability (gains and losses) within genes and at chromosome fragile sites was observed, resulting in changes to gene-expression profiles. Transcription stress near promoters correlated with high GCskew and the accumulation of R-loops at promoter-proximal regions, which localized with chromosomal regions where gains and losses were observed. In the absence of Senataxin, the Cockayne syndrome protein CSB was required for the recruitment of the transcription-coupled repair endonucleases (XPG and XPF) and RAD52 recombination protein to target and resolve transcription bubbles containing R-loops, leading to genomic instability. These results show that transcription stress is an important contributor to SETX mutation-associated chromosome fragility and AOA2.
- Published
- 2021
- Full Text
- View/download PDF
10. Molecular Characterization of Influenza C Viruses from Outbreaks in Hong Kong SAR, China
- Author
-
Jeremy Guntoro, Jerome Nicod, Saira Hussain, Burcu Ermetal, Janice Lo, Zheng Xiang, Herman Tse, Deborah Jackson, John W. McCauley, Rodney S. Daniels, Karen J Cross, and Aengus Stewart
- Subjects
Male ,Models, Molecular ,Protein Conformation, alpha-Helical ,Influenzavirus C ,viruses ,Gene Expression ,Disease ,Disease Outbreaks ,outbreak surveillance ,Spotlight ,Child ,Phylogeny ,0303 health sciences ,Molecular Epidemiology ,High-Throughput Nucleotide Sequencing ,Middle Aged ,genome sequencing ,Viral evolution ,Child, Preschool ,Epidemiological Monitoring ,Hong Kong ,Female ,Adult ,Adolescent ,Immunology ,Hemagglutinins, Viral ,Biology ,Microbiology ,Antigenic drift ,Virus ,03 medical and health sciences ,Genetic drift ,Virology ,Influenza, Human ,Humans ,Gene ,030304 developmental biology ,Aged ,Retrospective Studies ,virus evolution ,030306 microbiology ,Outbreak ,Infant ,influenza C virus ,Genetic Diversity and Evolution ,Amino Acid Substitution ,Insect Science ,Mutation ,Protein Conformation, beta-Strand ,Influenza C Virus ,Viral Fusion Proteins - Abstract
Influenza C virus infection of humans is common, and reinfection can occur throughout life. While symptoms are generally mild, severe disease cases have been reported, but knowledge of the virus is limited, as little systematic surveillance for influenza C virus is conducted and the virus cannot be studied by classical virologic methods because it cannot be readily isolated in laboratories. A combination of systematic surveillance in Hong Kong SAR, China, and new gene sequencing methods has been used in this study to assess influenza C virus evolution and provides evidence for a 2-year cycle of disease outbreaks. The results of studies like that reported here are key to developing an understanding of the impact of influenza C virus infection in humans and how virus evolution might be associated with epidemics., In 2014, the Centre for Health Protection in Hong Kong introduced screening for influenza C virus (ICV) as part of its routine surveillance for infectious agents in specimens collected from patients presenting with symptoms of respiratory viral infection, including influenza-like illness (ILI). A retrospective analysis of ICV detections up to week 26 of 2019 revealed persistent low-level circulation, with two outbreaks having occurred in the winters of 2015 to 2016 and 2017 to 2018. These outbreaks occurred at the same time as, and were dwarfed by, seasonal epidemics of influenza types A and B. Gene sequencing studies on stored ICV-positive clinical specimens from the two outbreaks have shown that the hemagglutinin-esterase (HE) genes of the viruses fall into two of the six recognized genetic lineages (represented by C/Kanagawa/1/76 and C/São Paulo/378/82), with there being significant genetic drift compared to earlier circulating viruses within both lineages. The location of a number of encoded amino acid substitutions in hemagglutinin-esterase fusion (HEF) glycoproteins suggests that antigenic drift may also have occurred. Observations of ICV outbreaks in other countries, with some of the infections being associated with severe disease, indicates that ICV infection has the potential to have significant clinical and health care impacts in humans. IMPORTANCE Influenza C virus infection of humans is common, and reinfection can occur throughout life. While symptoms are generally mild, severe disease cases have been reported, but knowledge of the virus is limited, as little systematic surveillance for influenza C virus is conducted and the virus cannot be studied by classical virologic methods because it cannot be readily isolated in laboratories. A combination of systematic surveillance in Hong Kong SAR, China, and new gene sequencing methods has been used in this study to assess influenza C virus evolution and provides evidence for a 2-year cycle of disease outbreaks. The results of studies like that reported here are key to developing an understanding of the impact of influenza C virus infection in humans and how virus evolution might be associated with epidemics.
- Published
- 2020
11. Regulation of the RNAPII Pool Is Integral to the DNA Damage Response
- Author
-
Ana Tufegdžić Vidaković, Ronald C. Conaway, Laura Milligan, Joan W. Conaway, Juston C. Weems, Stefan Boeing, Aengus Stewart, Michelle Harreman, Michelle Neumann, Jesper Q. Svejstrup, Ambrosius P. Snijders, David Tollervey, Richard Mitter, Vesela Encheva, Gavin Kelly, Anna Herlihy, Marta Rodríguez-Martínez, and Liam Gaul
- Subjects
Model organisms ,Genome instability ,DNA Repair ,Transcription, Genetic ,DNA repair ,DNA damage ,Gene Expression ,RNA polymerase II ,Biochemistry & Proteomics ,General Biochemistry, Genetics and Molecular Biology ,Article ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,Transcription (biology) ,ubiquitin ,ubiquitylation ,Computational & Systems Biology ,030304 developmental biology ,Chemical Biology & High Throughput ,0303 health sciences ,biology ,Biochemistry, Genetics and Molecular Biology(all) ,Genome Integrity & Repair ,UV irradiation ,Ubiquitination ,Tumour Biology ,Cell biology ,biology.protein ,transcription ,Genetics & Genomics ,Developmental biology ,030217 neurology & neurosurgery - Abstract
Summary In response to transcription-blocking DNA damage, cells orchestrate a multi-pronged reaction, involving transcription-coupled DNA repair, degradation of RNA polymerase II (RNAPII), and genome-wide transcription shutdown. Here, we provide insight into how these responses are connected by the finding that ubiquitylation of RNAPII itself, at a single lysine (RPB1 K1268), is the focal point for DNA-damage-response coordination. K1268 ubiquitylation affects DNA repair and signals RNAPII degradation, essential for surviving genotoxic insult. RNAPII degradation results in a shutdown of transcriptional initiation, in the absence of which cells display dramatic transcriptome alterations. Additionally, regulation of RNAPII stability is central to transcription recovery—persistent RNAPII depletion underlies the failure of this process in Cockayne syndrome B cells. These data expose regulation of global RNAPII levels as integral to the cellular DNA-damage response and open the intriguing possibility that RNAPII pool size generally affects cell-specific transcription programs in genome instability disorders and even normal cells., Graphical Abstract, Highlights • Specific RPB1 K1268 ubiquitylation targets RNAPII for UV-induced proteolysis • RPB1 K1268 ubiquitylation is required for surviving DNA damage • Control of the RNAPII pool via degradation regulates the transcriptome after UV • Lack of transcription recovery in Cockayne syndrome is caused by unstable RNAPII, Control of the pool of available RNA polymerase II shapes how cells respond to UV stress and the efficacy of the resulting damage response.
- Published
- 2020
- Full Text
- View/download PDF
12. The combination of CHK1 inhibitor with G-CSF overrides cytarabine resistance in human acute myeloid leukemia
- Author
-
Syed A Mian, Kevin Rouault-Pierre, Dominique Bonnet, William Grey, Ander Abarrategi, Aengus Stewart, Elizabeth Blackwood, John G. Gribben, and Alessandro Di Tullio
- Subjects
0301 basic medicine ,Male ,Myeloid ,Pyridines ,General Physics and Astronomy ,Mice, SCID ,Mice ,0302 clinical medicine ,Piperidines ,Mice, Inbred NOD ,Antineoplastic Combined Chemotherapy Protocols ,Granulocyte Colony-Stimulating Factor ,lcsh:Science ,Multidisciplinary ,Cytarabine ,Myeloid leukemia ,U937 Cells ,3. Good health ,Granulocyte colony-stimulating factor ,Leukemia ,Haematopoiesis ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,medicine.drug ,Science ,HL-60 Cells ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Pyrroles ,CHEK1 ,Progenitor cell ,Protein Kinase Inhibitors ,business.industry ,General Chemistry ,medicine.disease ,Xenograft Model Antitumor Assays ,Hematopoiesis ,carbohydrates (lipids) ,030104 developmental biology ,Drug Resistance, Neoplasm ,Checkpoint Kinase 1 ,Mutation ,Cancer research ,lcsh:Q ,business - Abstract
Cytarabine (AraC) represents the most effective single agent treatment for AML. Nevertheless, overriding AraC resistance in AML remains an unmet medical need. Here we show that the CHK1 inhibitor (CHK1i) GDC-0575 enhances AraC-mediated killing of AML cells both in vitro and in vivo, thus abrogating any potential chemoresistance mechanisms involving DNA repair. Importantly, this combination of drugs does not affect normal long-term hematopoietic stem/progenitors. Moreover, the addition of CHK1i to AraC does not generate de novo mutations and in patients’ samples where AraC is mutagenic, addition of CHK1i appears to eliminate the generation of mutant clones. Finally, we observe that persistent residual leukemic cells are quiescent and can become responsive to the treatment when forced into cycle via granulocyte colony-stimulating factor (G-CSF) administration. This drug combination (AraC+CHK1i+G-CSF) will open the doors for a more efficient treatment of AML in the clinic., Overriding cytarabine resistance in AML remains an unmet medical need. Here, the authors show that the CHK1 inhibitor GDC-0575 in combination with cytarabine and G-CSF has a significant anti-leukemic effect without toxicity to normal marrow stem and progenitor cells.
- Published
- 2017
13. Tracking the Evolution of Non–Small-Cell Lung Cancer
- Author
-
Mariam, Jamal-Hanjani, Gareth A, Wilson, Nicholas, McGranahan, Nicolai J, Birkbak, Thomas B K, Watkins, Selvaraju, Veeriah, Seema, Shafi, Diana H, Johnson, Richard, Mitter, Rachel, Rosenthal, Max, Salm, Stuart, Horswell, Mickael, Escudero, Nik, Matthews, Andrew, Rowan, Tim, Chambers, David A, Moore, Samra, Turajlic, Hang, Xu, Siow-Ming, Lee, Martin D, Forster, Tanya, Ahmad, Crispin T, Hiley, Christopher, Abbosh, Mary, Falzon, Elaine, Borg, Teresa, Marafioti, David, Lawrence, Martin, Hayward, Shyam, Kolvekar, Nikolaos, Panagiotopoulos, Sam M, Janes, Ricky, Thakrar, Asia, Ahmed, Fiona, Blackhall, Yvonne, Summers, Rajesh, Shah, Leena, Joseph, Anne M, Quinn, Phil A, Crosbie, Babu, Naidu, Gary, Middleton, Gerald, Langman, Simon, Trotter, Marianne, Nicolson, Hardy, Remmen, Keith, Kerr, Mahendran, Chetty, Lesley, Gomersall, Dean A, Fennell, Apostolos, Nakas, Sridhar, Rathinam, Girija, Anand, Sajid, Khan, Peter, Russell, Veni, Ezhil, Babikir, Ismail, Melanie, Irvin-Sellers, Vineet, Prakash, Jason F, Lester, Malgorzata, Kornaszewska, Richard, Attanoos, Haydn, Adams, Helen, Davies, Stefan, Dentro, Philippe, Taniere, Brendan, O'Sullivan, Helen L, Lowe, John A, Hartley, Natasha, Iles, Harriet, Bell, Yenting, Ngai, Jacqui A, Shaw, Javier, Herrero, Zoltan, Szallasi, Roland F, Schwarz, Aengus, Stewart, Sergio A, Quezada, John, Le Quesne, Peter, Van Loo, Caroline, Dive, Allan, Hackshaw, Charles, Swanton, and Karl, Peggs
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,DNA Copy Number Variations ,Somatic cell ,Bioinformatics ,medicine.disease_cause ,Disease-Free Survival ,Evolution, Molecular ,Genetic Heterogeneity ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Carcinoma, Non-Small-Cell Lung ,Chromosomal Instability ,Internal medicine ,Chromosome instability ,Carcinoma ,medicine ,Humans ,Exome ,Prospective Studies ,Prospective cohort study ,Lung cancer ,Phylogeny ,Mutation ,Manchester Cancer Research Centre ,Genetic heterogeneity ,business.industry ,ResearchInstitutes_Networks_Beacons/mcrc ,Sequence Analysis, DNA ,General Medicine ,Prognosis ,medicine.disease ,Editorial ,030104 developmental biology ,030220 oncology & carcinogenesis ,Female ,Neoplasm Recurrence, Local ,business - Abstract
Background: \ud Among patients with non–small-cell lung cancer (NSCLC), data on intratumor heterogeneity and cancer genome evolution have been limited to small retrospective cohorts. We wanted to prospectively investigate intratumor heterogeneity in relation to clinical outcome and to determine the clonal nature of driver events and evolutionary processes in early-stage NSCLC.\ud \ud Methods: \ud In this prospective cohort study, we performed multiregion whole-exome sequencing on 100 early-stage NSCLC tumors that had been resected before systemic therapy. We sequenced and analyzed 327 tumor regions to define evolutionary histories, obtain a census of clonal and subclonal events, and assess the relationship between intratumor heterogeneity and recurrence-free survival.\ud \ud Results: \ud We observed widespread intratumor heterogeneity for both somatic copy-number alterations and mutations. Driver mutations in EGFR, MET, BRAF, and TP53 were almost always clonal. However, heterogeneous driver alterations that occurred later in evolution were found in more than 75% of the tumors and were common in PIK3CA and NF1 and in genes that are involved in chromatin modification and DNA damage response and repair. Genome doubling and ongoing dynamic chromosomal instability were associated with intratumor heterogeneity and resulted in parallel evolution of driver somatic copy-number alterations, including amplifications in CDK4, FOXA1, and BCL11A. Elevated copy-number heterogeneity was associated with an increased risk of recurrence or death (hazard ratio, 4.9; P=4.4×10−4), which remained significant in multivariate analysis.\ud \ud Conclusions: \ud Intratumor heterogeneity mediated through chromosome instability was associated with an increased risk of recurrence or death, a finding that supports the potential value of chromosome instability as a prognostic predictor. (Funded by Cancer Research UK and others; TRACERx ClinicalTrials.gov number, NCT01888601.)
- Published
- 2017
- Full Text
- View/download PDF
14. SRF Co-factors Control the Balance between Cell Proliferation and Contractility
- Author
-
Aengus Stewart, Francesco Gualdrini, Nik Matthews, Richard Treisman, Stuart Horswell, Cyril Esnault, Imagerie et Modélisation en Neurobiologie et Cancérologie (IMNC (UMR_8165)), Université Paris-Sud - Paris 11 (UP11)-Institut National de Physique Nucléaire et de Physique des Particules du CNRS (IN2P3)-Université Paris Diderot - Paris 7 (UPD7)-Centre National de la Recherche Scientifique (CNRS), Laboratory for Atmospheric and Space Physics [Boulder] (LASP), and University of Colorado [Boulder]
- Subjects
0301 basic medicine ,Serum Response Factor ,Transcription, Genetic ,Mice ,Elk-1 ,Hi-C ,Transcription (biology) ,Gene expression ,Ras/MAPK signaling ,Extracellular Signal-Regulated MAP Kinases ,3. Good health ,Cell biology ,Cytoskeletal Gene ,embryonic structures ,cardiovascular system ,Tetradecanoylphorbol Acetate ,SRF ,transcription ,Signal Transduction ,animal structures ,education ,cell contraction ,Ternary Complex Factors ,Biology ,immediate-early genes ,Article ,Cell Line ,Contractility ,03 medical and health sciences ,transcription factors ,Serum response factor ,Animals ,Humans ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Molecular Biology ,Gene ,Transcription factor ,Cell Proliferation ,ets-Domain Protein Elk-1 ,Base Sequence ,Cell growth ,Gene Expression Profiling ,Genetic Complementation Test ,Cell Biology ,Fibroblasts ,Molecular biology ,030104 developmental biology ,Gene Expression Regulation ,Ternary Complex Factor ,Trans-Activators - Abstract
Summary The ERK-regulated ternary complex factors (TCFs) act with the transcription factor serum response factor (SRF) to activate mitogen-induced transcription. However, the extent of their involvement in the immediate-early transcriptional response, and their wider functional significance, has remained unclear. We show that, in MEFs, TCF inactivation significantly inhibits over 60% of TPA-inducible gene transcription and impairs cell proliferation. Using integrated SRF ChIP-seq and Hi-C data, we identified over 700 TCF-dependent SRF direct target genes involved in signaling, transcription, and proliferation. These also include a significant number of cytoskeletal gene targets for the Rho-regulated myocardin-related transcription factor (MRTF) SRF cofactor family. The TCFs act as general antagonists of MRTF-dependent SRF target gene expression, competing directly with the MRTFs for access to SRF. As a result, TCF-deficient MEFs exhibit hypercontractile and pro-invasive behavior. Thus, competition between TCFs and MRTFs for SRF determines the balance between antagonistic proliferative and contractile programs of gene expression., Graphical Abstract, Highlights • Integrated ChIP-seq Hi-C analysis identifies over 700 TCF-dependent SRF target genes • Over 60% of TPA-inducible gene transcription is TCF-dependent • TCF-dependent transcription potentiates cell proliferation • TCF/MRTF competition for SRF determines contractility and pro-invasive behavior, Two cofactor families, the TCFs and the MRTFs, compete for binding to SRF, a key regulator of the mitogen-responsive transcription. TCFs not only control proliferative gene expression programs but also regulate access of the MRTFs to SRF, thereby regulating cell contractility and pro-invasive behavior.
- Published
- 2016
- Full Text
- View/download PDF
15. A CRISPR platform for targeted in vivo screens identifies Toxoplasma gondii virulence factors in mice
- Author
-
Xingda Ye, Gavin Kelly, Caia Dominicus, Simon Butterworth, Aengus Stewart, Jeanette Wagener, Becky Saunders, Merav Ordan, Moritz Treeck, Rachael Instrell, Joanna Young, and Michael Howell
- Subjects
0301 basic medicine ,Virulence Factors ,Science ,General Physics and Astronomy ,Virulence ,02 engineering and technology ,Computational biology ,Biology ,Genome ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Gene Knockout Techniques ,03 medical and health sciences ,Single-cell analysis ,parasitic diseases ,Parasite immune evasion ,Parasite genetics ,Animals ,Humans ,CRISPR ,Clustered Regularly Interspaced Short Palindromic Repeats ,Genomic library ,lcsh:Science ,Pathogen ,Gene Library ,Multidisciplinary ,Cas9 ,General Chemistry ,021001 nanoscience & nanotechnology ,3. Good health ,Mice, Inbred C57BL ,030104 developmental biology ,lcsh:Q ,CRISPR-Cas Systems ,0210 nano-technology ,Genome, Protozoan ,Toxoplasma ,Toxoplasmosis ,Parasite host response ,RNA, Guide, Kinetoplastida ,Genetic screen - Abstract
Genome-wide CRISPR screening is a powerful tool to identify genes required under selective conditions. However, the inherent scale of genome-wide libraries can limit their application in experimental settings where cell numbers are restricted, such as in vivo infections or single cell analysis. The use of small scale CRISPR libraries targeting gene subsets circumvents this problem. Here we develop a method for rapid generation of custom guide RNA (gRNA) libraries using arrayed single-stranded oligonucleotides for reproducible pooled cloning of CRISPR/Cas9 libraries. We use this system to generate mutant pools of different sizes in the protozoan parasite Toxoplasma gondi and describe optimised analysis methods for small scale libraries. An in vivo genetic screen in the murine host identifies novel and known virulence factors and we confirm results using cloned knock-out parasites. Our study also reveals a potential trans-rescue of individual knock-out parasites in pools of mutants compared to homogenous knock-out lines of the key virulence factor MYR1., Targeted CRISPR libraries expand the use of genetic screens across experimental conditions. Here, the authors develop a method for generating and analysing small scale custom CRISPR libraries and use it in the human and livestock pathogen Toxoplasma gondii to identify virulence factors in mice.
- Published
- 2019
- Full Text
- View/download PDF
16. Pharmacological Bypass of Cockayne Syndrome B Function in Neuronal Differentiation
- Author
-
Alysson R. Muotri, Probir Chakravarty, Jace Jones-Tabah, Yuming Wang, Rebecca R. Laposa, Aengus Stewart, and Jesper Q. Svejstrup
- Subjects
0301 basic medicine ,Premature aging ,Chromatin Immunoprecipitation ,Amitriptyline ,Down-Regulation ,Tropomyosin receptor kinase B ,Biology ,Real-Time Polymerase Chain Reaction ,Bioinformatics ,General Biochemistry, Genetics and Molecular Biology ,Synaptotagmin 1 ,Cockayne syndrome ,Synaptotagmins ,03 medical and health sciences ,0302 clinical medicine ,Neurodevelopmental disorder ,Neurotrophic factors ,Report ,Cell Line, Tumor ,medicine ,Humans ,Receptor, trkB ,RNA, Small Interfering ,Cockayne Syndrome ,Poly-ADP-Ribose Binding Proteins ,lcsh:QH301-705.5 ,Membrane Glycoproteins ,Brain-Derived Neurotrophic Factor ,DNA Helicases ,Cell Differentiation ,Protein-Tyrosine Kinases ,Flavones ,medicine.disease ,3. Good health ,DNA Repair Enzymes ,030104 developmental biology ,lcsh:Biology (General) ,Microscopy, Fluorescence ,nervous system ,biology.protein ,RNA Interference ,Ectopic expression ,Neuroscience ,030217 neurology & neurosurgery ,Neurotrophin - Abstract
Summary Cockayne syndrome (CS) is a severe neurodevelopmental disorder characterized by growth abnormalities, premature aging, and photosensitivity. Mutation of Cockayne syndrome B (CSB) affects neuronal gene expression and differentiation, so we attempted to bypass its function by expressing downstream target genes. Intriguingly, ectopic expression of Synaptotagmin 9 (SYT9), a key component of the machinery controlling neurotrophin release, bypasses the need for CSB in neuritogenesis. Importantly, brain-derived neurotrophic factor (BDNF), a neurotrophin implicated in neuronal differentiation and synaptic modulation, and pharmacological mimics such as 7,8-dihydroxyflavone and amitriptyline can compensate for CSB deficiency in cell models of neuronal differentiation as well. SYT9 and BDNF are downregulated in CS patient brain tissue, further indicating that sub-optimal neurotrophin signaling underlies neurological defects in CS. In addition to shedding light on cellular mechanisms underlying CS and pointing to future avenues for pharmacological intervention, these data suggest an important role for SYT9 in neuronal differentiation., Graphical Abstract, Highlights • Neuritogenesis defects in CS cell lines can be overcome by overexpression of SYT9 • SYT9 is crucial for neuritogenesis and involved in neurotrophin (BDNF) signaling • Neuritogenesis defects in CS cell lines can be overcome by BDNF treatment • They can also be overcome by treatment with amitriptyline, an FDA-approved BDNF mimic, Wang et al. show that Cockayne syndrome cell lines have defects in gene regulatory loops, resulting in sub-optimal neurotrophin signaling and explaining their defects in neurogenesis. These defects can be overcome by Synaptotagmin 9 overexpression or by treatment with NTRK2 (TrkB) agonists, pointing to future disease intervention.
- Published
- 2016
- Full Text
- View/download PDF
17. Elongation Factor TFIIS Prevents Transcription Stress and R-Loop Accumulation to Maintain Genome Stability
- Author
-
Andrés Aguilera, Jesper Q. Svejstrup, Diana Zatreanu, Richard Mitter, Stefania Roma, Emanuela Tumini, Hannah Williams, Lea H. Gregersen, and Aengus Stewart
- Subjects
Genome instability ,Elongation factor ,chemistry.chemical_compound ,biology ,chemistry ,Transcription (biology) ,RNA splicing ,biology.protein ,RNA polymerase II ,Gene ,Polymerase ,DNA ,Cell biology - Abstract
While intriguing connections between RNA polymerase II (RNAPII) transcription stress, R-loops, and genome instability have been established, the underlying mechanisms remain poorly understood. Here we used a mutant version of elongation factor TFIIS (TFIISmut) to specifically induce increased levels of RNAPII pausing, arrest, and/or backtracking in human cells. TFIISmut expression results in slower elongation rates, relative depletion of polymerases from the end of genes, and increased levels of stopped RNAPII. It affects mRNA splicing and termination as well. Remarkably, however, TFIISmut expression also dramatically increases R-loops, which may form at the anterior end of backtracked RNAPII and trigger genome instability, including DNA strand breaks. These results shed new light on the relationship between transcription stress and R-loops, and suggest that different classes of R-loops exist, potentially with distinct consequences for genome instability.
- Published
- 2018
- Full Text
- View/download PDF
18. Timing the Landmark Events in the Evolution of Clear Cell Renal Cell Cancer: TRACERx Renal
- Author
-
Nicholas McGranahan, Inigo Martincorena, Kevin Litchfield, Aengus Stewart, P. Andrew Futreal, Ben Challacombe, Ashish Chandra, José I. López, István Csabai, Hang Xu, David Nicol, David C. Wedge, Martin Gore, Tim O'Brien, James H.R. Farmery, Andrew Rowan, Archana Fernando, Charles Swanton, Stuart Horswell, Stéphane Oudard, Grant D. Stewart, Lisa Pickering, Francesco Maura, Andy G. Lynch, Joanna Lynch, James Larkin, Nicos Fotiadis, Thomas J. Mitchell, Sarah Rudman, Lewis Au, Lavinia Spain, Zoltan Szallasi, Aspasia Soultati, Reza Elaidi, Simon Chowdhury, Patrick S. Tarpey, Nicos Angelopoulos, Steve Hazell, Peter J. Campbell, Keiran Raine, Samra Turajlic, Tim Chambers, Gordon Stamp, Lucy R. Yates, Adam Butler, Jon W. Teague, Mark Stares, University of St Andrews. Statistics, University of St Andrews. School of Medicine, and University of St Andrews. Cellular Medicine Division
- Subjects
Male ,0301 basic medicine ,Clear cell renal cell carcinoma ,Telomerase ,tert promoter mutations ,Gene Dosage ,clear cell renal cell carcinoma ,carcinoma ,Prospective Studies ,Aged, 80 and over ,cancer evolution ,Chromothripsis ,Manchester Cancer Research Centre ,kidney cancer ,11 Medical And Health Sciences ,Middle Aged ,Kidney Neoplasms ,3. Good health ,Von Hippel-Lindau Tumor Suppressor Protein ,Disease Progression ,Chromosomes, Human, Pair 5 ,Female ,Chromosomes, Human, Pair 3 ,transcription ,Life Sciences & Biomedicine ,Adult ,Biochemistry & Molecular Biology ,hTERT gene ,Biology ,TRACERx Renal Consortium ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cancer evolution ,RC0254 ,03 medical and health sciences ,SDG 3 - Good Health and Well-being ,Carcinoma ,medicine ,copy-number ,C-Myc ,Humans ,patterns ,Carcinoma, Renal Cell ,Aged ,Science & Technology ,Genome, Human ,RC0254 Neoplasms. Tumors. Oncology (including Cancer) ,Point mutation ,ResearchInstitutes_Networks_Beacons/mcrc ,DAS ,Cell Biology ,06 Biological Sciences ,medicine.disease ,030104 developmental biology ,Chromosome 3 ,Mutation ,Cancer research ,somatic mutations ,chromothripsis ,5' Untranslated Regions ,Kidney cancer ,Clear cell ,Developmental Biology - Abstract
Summary Clear cell renal cell carcinoma (ccRCC) is characterized by near-universal loss of the short arm of chromosome 3, deleting several tumor suppressor genes. We analyzed whole genomes from 95 biopsies across 33 patients with clear cell renal cell carcinoma. We find hotspots of point mutations in the 5′ UTR of TERT, targeting a MYC-MAX-MAD1 repressor associated with telomere lengthening. The most common structural abnormality generates simultaneous 3p loss and 5q gain (36% patients), typically through chromothripsis. This event occurs in childhood or adolescence, generally as the initiating event that precedes emergence of the tumor’s most recent common ancestor by years to decades. Similar genomic changes drive inherited ccRCC. Modeling differences in age incidence between inherited and sporadic cancers suggests that the number of cells with 3p loss capable of initiating sporadic tumors is no more than a few hundred. Early development of ccRCC follows well-defined evolutionary trajectories, offering opportunity for early intervention., Graphical Abstract, Highlights • Novel hotspot of driver mutations in 5′-UTR repressor of TERT, expanding telomeres • Most common cause of 3p loss is a chromothripsis event, generating concurrent 5q gain • t(3;5) event occurs in childhood or adolescence, decades before tumor diagnosed • Initial clonal expansion after 3p loss starts from only a few hundred cells, Combination of whole-genome sequencing analysis and a multi-region sampling approach provides insights into the nature and timing of key oncogenic events in clear cell renal cell carcinoma, depicts the evolutionary trajectories of tumors in patients and highlights the opportunity for early intervention.
- Published
- 2018
- Full Text
- View/download PDF
19. Deterministic Evolutionary Trajectories Influence Primary Tumor Growth: TRACERx Renal
- Author
-
Emma Nye, Ben Phillimore, Nicholas McGranahan, Gordon Stamp, Archana Fernando, José I. López, Thomas B.K. Watkins, Hang Xu, Lisa Pickering, Kevin Litchfield, David Nicol, Sharmin Begum, Mariam Jamal-Hanjani, Lewis Au, Sharanpreet Lall, Lavinia Spain, Mary Varia, Nicolai Juul Birkbak, Steve Hazell, Eva Grönroos, Nicholas M. Luscombe, Mark Stares, Martin Gore, Alexander Polson, Aspasia Soultati, Ben Challacombe, Marcin Krzystanek, Nicos Fotiadis, Tim Chambers, Aengus Stewart, Ismaeel Aurangzeb, James Larkin, Dezso Ribli, Adam Huffman, Rachel Rosenthal, Orsolya Pipek, Max Salm, Peter J. Campbell, Charles Swanton, Catherine Horsfield, Samra Turajlic, Mickael Escudero, Bruno Silva, Faiz Jabbar, Thomas J. Mitchell, Zoltan Szallasi, Roland F. Schwarz, Simon Chowdhury, Sarkhara Sharma, Ashish Chandra, Sebastijan Hobor, Stuart Horswell, Wei Xing, Gareth A. Wilson, Sophia Ward, Tim O'Brien, Stefan Boeing, Claudia Eichler-Jonsson, Javier Herrero, Andrew Rowan, Nik Matthews, István Csabai, Peter Van Loo, Jonathan C. Smith, Carolina Navas, Greg Elgar, Joanna Lynch, Marta Costa, Sarah Rudman, and Rosalie Fisher
- Subjects
0301 basic medicine ,Cancer Research ,cell carcinoma ,Computational biology ,Biology ,rhabdoid differentiation ,Somatic evolution in cancer ,General Biochemistry, Genetics and Molecular Biology ,copy-number alterations ,03 medical and health sciences ,Renal cell carcinoma ,medicine ,BAP1 ,Allele ,gene ,Genetic heterogeneity ,active surveillance ,driver mutation ,sequencing data ,kidney cancer ,medicine.disease ,Primary tumor ,international society ,3. Good health ,030104 developmental biology ,Biomarker (medicine) ,Kidney cancer - Abstract
The evolutionary features of clear-cell renal cell carcinoma (ccRCC) have not been systematically studied to date. We analyzed 1,206 primary tumor regions from 101 patients recruited into the multi-center prospective study, TRACERx Renal. We observe up to 30 driver events per tumor and show that subclonal diversification is associated with known prognostic parameters. By resolving the patterns of driver event ordering, co-occurrence, and mutual exclusivity at clone level, we show the deterministic nature of clonal evolution. ccRCC can be grouped into seven evolutionary subtypes, ranging from tumors characterized by early fixation of multiple mutational and copy number drivers and rapid metastases to highly branched tumors with > 10 subclonal drivers and extensive parallel evolution associated with attenuated progression. We identify genetic diversity and chromosomal complexity as determinants of patient outcome. Our insights reconcile the variable clinical behavior of ccRCC and suggest evolutionary potential as a biomarker for both intervention and surveillance. We thank Aida Murra, Naheed Shaikh, Justine Korteweg, Jeremy Tai, Eleanor Carlyle, Leonora Conneely, KimEdmonds, Karla Lingard, Karen O'Meara, Helen Breeze, Sarah Sarker, Lesley Cooper, Linda Shephard, Susie Slater, and Catherine Rogers for study support. We thank the patients and their families. S.T. and H.X. are funded by Cancer Research UK (CRUK) (C50947/A18176). S.T., T.C., J.L., and M.G. are funded by the National Institute for Health Research (NIHR) Biomedical Research Centre (BRC) at the Royal Marsden Hospital and Institute of Cancer Research (A109). J.I.L. is funded by the Ministerio de Economia y Competitividad (MINECO, SAF2016-79847-R). M.S., A.S., J. Lynch, R.F., L.A., and L.S. are funded by the Royal Marsden Cancer Charity. K.L. is funded by UK Medical Research Council (MR/P014712/1). T.B.K.W. is funded by the European Union Seventh Framework Programme (FP7-People-2013-ITN). M.J.-H. is funded by the NIHR. N.M.L. is a Winton Group Leader in recognition of the Winton Charitable Foundation's support towards the establishment of The Francis Crick Institute. N.M.L. is additionally funded by a Wellcome Trust Joint Investigator Award (103760/Z/14/Z) and the MRC eMedLab Medical Bioinformatics Infrastructure Award (MR/L016311/1). M.K. is funded by the Danish Cancer Society grant (R90-A6213). P.C. is funded by the Wellcome Trust (WT088340MA). N.M. receives funding from CRUK, Rosetrees, and the NIHR BRC at University College London Hospitals. C.S. is a Royal Society Napier Research Professor. C.S. is funded by Cancer Research UK (TRACERx and CRUK Cancer Immunotherapy Catalyst Network), the CRUK Lung Cancer Centre of Excellence, Stand Up 2 Cancer (SU2C), the Rosetrees and Stoneygate Trusts, NovoNordisk Foundation (THESEUS), Marie Curie Network PloidyNet, the NIHR BRC at University College London Hospitals, and the CRUK University College London Experimental Cancer Medicine Centre. C.S., O.P., D.R., I.C., and Z.S. are funded by NovoNordisk Foundation (16584). O.P., D.R., and I.C. are funded by the National Research, Development and Innovation Office of Hungary (NVKP_16-1-20160004). This work was supported by CRUK (Clinical Scientist Fellowship to S.T., C50947/A18176), The Francis Crick Institute, which receives its core funding from Cancer Reseach UK (FC010110), the UK Medical Research Council (FC010110), the Wellcome Trust (FC010110), and NIHR BRC at the Royal Marsden Hospital and Institute of Cancer Research (A109). High-performance computing was supported by eMedLab. The results published here are in whole or part based upon data generated by the TCGA Research Network: http://cancergenome.nih.gov/.
- Published
- 2018
- Full Text
- View/download PDF
20. Allele-Specific HLA Loss and Immune Escape in Lung Cancer Evolution
- Author
-
Nicholas McGranahan, Rachel Rosenthal, Crispin T. Hiley, Andrew J. Rowan, Thomas B.K. Watkins, Gareth A. Wilson, Nicolai J. Birkbak, Selvaraju Veeriah, Peter Van Loo, Javier Herrero, Charles Swanton, Mariam Jamal-Hanjani, Seema Shafi, Justyna Czyzewska-Khan, Diana Johnson, Joanne Laycock, Leticia Bosshard-Carter, Pat Gorman, Robert E. Hynds, Gareth Wilson, Stuart Horswell, Richard Mitter, Mickael Escudero, Aengus Stewart, Andrew Rowan, Hang Xu, Samra Turajlic, Crispin Hiley, Christopher Abbosh, Jacki Goldman, Richard Kevin Stone, Tamara Denner, Nik Matthews, Greg Elgar, Sophia Ward, Marta Costa, Sharmin Begum, Ben Phillimore, Tim Chambers, Emma Nye, Sofia Graca, Maise Al Bakir, Kroopa Joshi, Andrew Furness, Assma Ben Aissa, Yien Ning Sophia Wong, Andy Georgiou, Sergio Quezada, John A. Hartley, Helen L. Lowe, David Lawrence, Martin Hayward, Nikolaos Panagiotopoulos, Shyam Kolvekar, Mary Falzon, Elaine Borg, Teresa Marafioti, Celia Simeon, Gemma Hector, Amy Smith, Marie Aranda, Marco Novelli, Dahmane Oukrif, Sam M. Janes, Ricky Thakrar, Martin Forster, Tanya Ahmad, Siow Ming Lee, Dionysis Papadatos-Pastos, Dawn Carnell, Ruheena Mendes, Jeremy George, Neal Navani, Asia Ahmed, Magali Taylor, Junaid Choudhary, Yvonne Summers, Raffaele Califano, Paul Taylor, Rajesh Shah, Piotr Krysiak, Kendadai Rammohan, Eustace Fontaine, Richard Booton, Matthew Evison, Phil Crosbie, Stuart Moss, Faiza Idries, Leena Joseph, Paul Bishop, Anshuman Chaturved, Anne Marie Quinn, Helen Doran, Angela Leek, Phil Harrison, Katrina Moore, Rachael Waddington, Juliette Novasio, Fiona Blackhall, Jane Rogan, Elaine Smith, Caroline Dive, Jonathan Tugwood, Ged Brady, Dominic G. Rothwell, Francesca Chemi, Jackie Pierce, Sakshi Gulati, Babu Naidu, Gerald Langman, Simon Trotter, Mary Bellamy, Hollie Bancroft, Amy Kerr, Salma Kadiri, Joanne Webb, Gary Middleton, Madava Djearaman, Dean Fennell, Jacqui A. Shaw, John Le Quesne, David Moore, Apostolos Nakas, Sridhar Rathinam, William Monteiro, Hilary Marshall, Louise Nelson, Jonathan Bennett, Joan Riley, Lindsay Primrose, Luke Martinson, Girija Anand, Sajid Khan, Anita Amadi, Marianne Nicolson, Keith Kerr, Shirley Palmer, Hardy Remmen, Joy Miller, Keith Buchan, Mahendran Chetty, Lesley Gomersall, Jason Lester, Alison Edwards, Fiona Morgan, Haydn Adams, Helen Davies, Malgorzata Kornaszewska, Richard Attanoos, Sara Lock, Azmina Verjee, Mairead MacKenzie, Maggie Wilcox, Harriet Bell, Allan Hackshaw, Yenting Ngai, Sean Smith, Nicole Gower, Christian Ottensmeier, Serena Chee, Benjamin Johnson, Aiman Alzetani, Emily Shaw, Eric Lim, Paulo De Sousa, Monica Tavares Barbosa, Alex Bowman, Simon Jordan, Alexandra Rice, Hilgardt Raubenheimer, Chiara Proli, Maria Elena Cufari, John Carlo Ronquillo, Angela Kwayie, Harshil Bhayani, Morag Hamilton, Yusura Bakar, Natalie Mensah, Lyn Ambrose, Anand Devaraj, Silviu Buderi, Jonathan Finch, Leire Azcarate, Hema Chavan, Sophie Green, Hillaria Mashinga, Andrew G. Nicholson, Kelvin Lau, Michael Sheaff, Peter Schmid, John Conibear, Veni Ezhil, Babikir Ismail, Melanie Irvin-sellers, Vineet Prakash, Peter Russell, Teresa Light, Tracey Horey, Sarah Danson, Jonathan Bury, John Edwards, Jennifer Hill, Sue Matthews, Yota Kitsanta, Kim Suvarna, Patricia Fisher, Allah Dino Keerio, Michael Shackcloth, John Gosney, Pieter Postmus, Sarah Feeney, Julius Asante-Siaw, Hugo J.W.L. Aerts, Stefan Dentro, Christophe Dessimoz, TRACERx Consortium, Swanton, C., Jamal-Hanjani, M., Veeriah, S., Shafi, S., Czyzewska-Khan, J., Johnson, D., Laycock, J., Bosshard-Carter, L., Rosenthal, R., Gorman, P., Hynds, R.E., Wilson, G., Birkbak, N.J., Watkins, TBK, McGranahan, N., Horswell, S., Mitter, R., Escudero, M., Stewart, A., Van Loo, P., Rowan, A., Xu, H., Turajlic, S., Hiley, C., Abbosh, C., Goldman, J., Stone, R.K., Denner, T., Matthews, N., Elgar, G., Ward, S., Costa, M., Begum, S., Phillimore, B., Chambers, T., Nye, E., Graca, S., Al Bakir, M., Joshi, K., Furness, A., Ben Aissa, A., Wong, YNS, Georgiou, A., Quezada, S., Hartley, J.A., Lowe, H.L., Herrero, J., Lawrence, D., Hayward, M., Panagiotopoulos, N., Kolvekar, S., Falzon, M., Borg, E., Marafioti, T., Simeon, C., Hector, G., Smith, A., Aranda, M., Novelli, M., Oukrif, D., Janes, S.M., Thakrar, R., Forster, M., Ahmad, T., Lee, S.M., Papadatos-Pastos, D., Carnell, D., Mendes, R., George, J., Navani, N., Ahmed, A., Taylor, M., Choudhary, J., Summers, Y., Califano, R., Taylor, P., Shah, R., Krysiak, P., Rammohan, K., Fontaine, E., Booton, R., Evison, M., Crosbie, P., Moss, S., Idries, F., Joseph, L., Bishop, P., Chaturved, A., Quinn, A.M., Doran, H., Leek, A., Harrison, P., Moore, K., Waddington, R., Novasio, J., Blackhall, F., Rogan, J., Smith, E., Dive, C., Tugwood, J., Brady, G., Rothwell, D.G., Chemi, F., Pierce, J., Gulati, S., Naidu, B., Langman, G., Trotter, S., Bellamy, M., Bancroft, H., Kerr, A., Kadiri, S., Webb, J., Middleton, G., Djearaman, M., Fennell, D., Shaw, J.A., Le Quesne, J., Moore, D., Nakas, A., Rathinam, S., Monteiro, W., Marshall, H., Nelson, L., Bennett, J., Riley, J., Primrose, L., Martinson, L., Anand, G., Khan, S., Amadi, A., Nicolson, M., Kerr, K., Palmer, S., Remmen, H., Miller, J., Buchan, K., Chetty, M., Gomersall, L., Lester, J., Edwards, A., Morgan, F., Adams, H., Davies, H., Kornaszewska, M., Attanoos, R., Lock, S., Verjee, A., MacKenzie, M., Wilcox, M., Bell, H., Hackshaw, A., Ngai, Y., Smith, S., Gower, N., Ottensmeier, C., Chee, S., Johnson, B., Alzetani, A., Shaw, E., Lim, E., De Sousa, P., Barbosa, M.T., Bowman, A., Jordan, S., Rice, A., Raubenheimer, H., Proli, C., Cufari, M.E., Ronquillo, J.C., Kwayie, A., Bhayani, H., Hamilton, M., Bakar, Y., Mensah, N., Ambrose, L., Devaraj, A., Buderi, S., Finch, J., Azcarate, L., Chavan, H., Green, S., Mashinga, H., Nicholson, A.G., Lau, K., Sheaff, M., Schmid, P., Conibear, J., Ezhil, V., Ismail, B., Irvin-Sellers, M., Prakash, V., Russell, P., Light, T., Horey, T., Danson, S., Bury, J., Edwards, J., Hill, J., Matthews, S., Kitsanta, Y., Suvarna, K., Fisher, P., Keerio, A.D., Shackcloth, M., Gosney, J., Postmus, P., Feeney, S., Asante-Siaw, J., Aerts, HJWL, Dentro, S., and Dessimoz, C.
- Subjects
Male ,immune-editing ,0301 basic medicine ,DOWN-REGULATION ,immune-escape ,Lung Neoplasms ,Loss of Heterozygosity ,Cohort Studies ,Loss of heterozygosity ,HLA Antigens ,Carcinoma, Non-Small-Cell Lung ,Chromosome instability ,MUTATIONAL PROCESSES ,11 Medical and Health Sciences ,Aged, 80 and over ,Antigen Presentation ,cancer evolution ,Manchester Cancer Research Centre ,bioinformatics ,Middle Aged ,3. Good health ,Female ,loss of heterozygosity ,SENSITIVITY ,Life Sciences & Biomedicine ,Adult ,Biochemistry & Molecular Biology ,chromosomal instability ,Antigen presentation ,Locus (genetics) ,NEOANTIGENS ,Human leukocyte antigen ,Biology ,Polymorphism, Single Nucleotide ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Immune system ,copy number ,medicine ,Humans ,Lung cancer ,Aged ,Science & Technology ,ResearchInstitutes_Networks_Beacons/mcrc ,CTLA-4 BLOCKADE ,Cell Biology ,06 Biological Sciences ,medicine.disease ,PD-1 BLOCKADE ,neoantigen ,lung cancer ,030104 developmental biology ,Carcinoma, Non-Small-Cell Lung/genetics ,Carcinoma, Non-Small-Cell Lung/immunology ,Carcinoma, Non-Small-Cell Lung/pathology ,Carcinoma, Non-Small-Cell Lung/therapy ,HLA Antigens/genetics ,HLA Antigens/immunology ,Lung Neoplasms/genetics ,Lung Neoplasms/immunology ,Lung Neoplasms/pathology ,Lung Neoplasms/therapy ,Mutation ,Tumor Escape ,heterogeneity ,TRACERx Consortium ,DISCOVERY ,CELLS ,Immunology ,RESISTANCE ,Developmental Biology - Abstract
Immune evasion is a hallmark of cancer. Losing the ability to present neoantigens through human leukocyte antigen (HLA) loss may facilitate immune evasion. However, the polymorphic nature of the locus has precluded accurate HLA copy-number analysis. Here, we present loss of heterozygosity in human leukocyte antigen (LOHHLA), a computational tool to determine HLA allele-specific copy number from sequencing data. Using LOHHLA, we find that HLA LOH occurs in 40% of non-small-cell lung cancers (NSCLCs) and is associated with a high subclonal neoantigen burden, APOBEC-mediated mutagenesis, upregulation of cytolytic activity, and PD-L1 positivity. The focal nature of HLA LOH alterations, their subclonal frequencies, enrichment in metastatic sites, and occurrence as parallel events suggests that HLA LOH is an immune escape mechanism that is subject to strong microenvironmental selection pressures later in tumor evolution. Characterizing HLA LOH with LOHHLA refines neoantigen prediction and may have implications for our understanding of resistance mechanisms and immunotherapeutic approaches targeting neoantigens. VIDEO ABSTRACT.
- Published
- 2017
- Full Text
- View/download PDF
21. BCL9L dysfunction impairs caspase-2 expression permitting aneuploidy tolerance in colorectal cancer
- Author
-
Charles Swanton, Nicholas Matthews, Eva Grönroos, Sebastijan Hobor, Nicholas McGranahan, Andrew Rowan, Nicolai Juul Birkbak, Sharmin Begum, Michal Kovac, Benjamin Phillimore, Gordon Stamp, Dahmane Oukrif, Enric Domingo, Stuart Horswell, Håvard E. Danielsen, Aengus Stewart, Marco Novelli, Laurent Sansregret, Rebecca A. Burrell, Ian Tomlinson, Bradley Spencer-Dene, Carlos López-García, and Francesco Favero
- Subjects
0301 basic medicine ,Genome instability ,caspase-2 ,p53 ,Cancer Research ,chromosome segregation errors ,Colorectal cancer ,Aneuploidy ,colorectal cancer evolution ,Loss of heterozygosity ,Mice ,Chromosome instability ,Chromosome Segregation ,Genetics ,Aged, 80 and over ,aneuploidy tolerance ,Wnt signaling pathway ,Caspase 2 ,Proto-Oncogene Proteins c-mdm2 ,Middle Aged ,3. Good health ,DNA-Binding Proteins ,Cysteine Endopeptidases ,Oncology ,mitotic checkpoint ,Colorectal Neoplasms ,BH3 Interacting Domain Death Agonist Protein ,chromosomal instability ,intratumor heterogeneity ,Biology ,Article ,BID ,03 medical and health sciences ,SDG 3 - Good Health and Well-being ,medicine ,Animals ,Humans ,BCL9L ,neoplasms ,Aged ,Genetic heterogeneity ,Microsatellite instability ,Cell Biology ,medicine.disease ,HCT116 Cells ,030104 developmental biology ,Mutation ,Tumor Suppressor Protein p53 ,Transcription Factors - Abstract
Summary Chromosomal instability (CIN) contributes to cancer evolution, intratumor heterogeneity, and drug resistance. CIN is driven by chromosome segregation errors and a tolerance phenotype that permits the propagation of aneuploid genomes. Through genomic analysis of colorectal cancers and cell lines, we find frequent loss of heterozygosity and mutations in BCL9L in aneuploid tumors. BCL9L deficiency promoted tolerance of chromosome missegregation events, propagation of aneuploidy, and genetic heterogeneity in xenograft models likely through modulation of Wnt signaling. We find that BCL9L dysfunction contributes to aneuploidy tolerance in both TP53-WT and mutant cells by reducing basal caspase-2 levels and preventing cleavage of MDM2 and BID. Efforts to exploit aneuploidy tolerance mechanisms and the BCL9L/caspase-2/BID axis may limit cancer diversity and evolution., Graphical Abstract, Highlights • Loss-of-function alterations in BCL9L are frequent in aneuploid CRC • BCL9L dysfunction drives aneuploidy tolerance by reducing levels of caspase-2 • Caspase-2 activation following aneuploidy results in MDM2 and BID cleavage • p53 stabilization after chromosome missegregation is regulated by caspase-2, López-García et al. find that BCL9L is often genetically inactivated in human colorectal cancers with chromosomal instability. BCL9L dysfunction promotes aneuploidy tolerance by reducing basal caspase-2 levels and preventing cleavage of MDM2 and BID independent of TP53 mutation status.
- Published
- 2017
- Full Text
- View/download PDF
22. ERK-Induced Activation of TCF Family of SRF Cofactors Initiates a Chromatin Modification Cascade Associated with Transcription
- Author
-
Richard Treisman, Stuart Horswell, Aengus Stewart, Gavin Kelly, Phil East, Francesco Gualdrini, Nik Matthews, Cyril Esnault, Lincoln's Inn Fields Laboratories, and Cancer Research UK
- Subjects
0301 basic medicine ,MESH: Signal Transduction ,Serum Response Factor ,Transcription, Genetic ,[SDV]Life Sciences [q-bio] ,Transcription coregulator ,MESH: Mice, Knockout ,Histones ,Mice ,Elk-1 ,Histone methylation ,MESH: Early Growth Response Protein 1 ,Histone code ,histone modification ,MESH: Animals ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,MESH: Extracellular Signal-Regulated MAP Kinases ,Mice, Knockout ,MESH: Histones ,General transcription factor ,biology ,Activating transcription factor 2 ,ternary complex factor ,3. Good health ,ERK ,MESH: Serum Response Factor ,H3 phosphorylation ,embryonic structures ,Tetradecanoylphorbol Acetate ,RNA Interference ,SRF ,MESH: Transcription Initiation Site ,Transcription Initiation Site ,TCF Transcription Factors ,transcription ,Signal Transduction ,MESH: Enzyme Activation ,MESH: Mutation ,MESH: RNA Interference ,Transfection ,immediate-early genes ,Article ,Cell Line ,MESH: Chromatin ,03 medical and health sciences ,Sp3 transcription factor ,Animals ,MESH: ets-Domain Protein Elk-1 ,Transcription factor ,Molecular Biology ,MESH: Mice ,MESH: Tetradecanoylphorbol Acetate ,Early Growth Response Protein 1 ,ets-Domain Protein Elk-1 ,MESH: Phosphorylation ,MESH: Transcription, Genetic ,MESH: Transfection ,Pioneer factor ,MESH: Chromatin Assembly and Disassembly ,Cell Biology ,Chromatin Assembly and Disassembly ,Molecular biology ,MESH: Cell Line ,Enzyme Activation ,030104 developmental biology ,Mutation ,biology.protein ,chromatin ,MESH: TCF Transcription Factors - Abstract
Summary We investigated the relationship among ERK signaling, histone modifications, and transcription factor activity, focusing on the ERK-regulated ternary complex factor family of SRF partner proteins. In MEFs, activation of ERK by TPA stimulation induced a common pattern of H3K9acS10ph, H4K16ac, H3K27ac, H3K9acK14ac, and H3K4me3 at hundreds of transcription start site (TSS) regions and remote regulatory sites. The magnitude of the increase in histone modification correlated well with changes in transcription. H3K9acS10ph preceded the other modifications. Most induced changes were TCF dependent, but TCF-independent TSSs exhibited the same hierarchy, indicating that it reflects gene activation per se. Studies with TCF Elk-1 mutants showed that TCF-dependent ERK-induced histone modifications required Elk-1 to be phosphorylated and competent to activate transcription. Analysis of direct TCF-SRF target genes and chromatin modifiers confirmed this and showed that H3S10ph required only Elk-1 phosphorylation. Induction of histone modifications following ERK stimulation is thus directed by transcription factor activation and transcription., Graphical Abstract, Highlights • TPA-driven ERK activation induces a common pattern of histone modifications at TSSs • Most ERK-induced histone modifications are dependent on TCF family of SRF cofactors • At these TSSs, induced histone modifications require ERK-dependent TCF phosphorylation • TCF-dependent histone modifications require TCF to recruit the transcription machinery, In MEFs, TPA-induced ERK activation induces a common pattern of histone modifications at more than 2,000 TSSs. Modifications at most TSSs are dependent on the TCF family of ERK-regulated SRF cofactors. At these TSSs, induced modifications are dependent both on phosphorylation of TCF and on its ability to recruit the transcriptional machinery.
- Published
- 2017
- Full Text
- View/download PDF
23. UV Irradiation Induces a Non-coding RNA that Functionally Opposes the Protein Encoded by the Same Gene
- Author
-
Richard Mitter, Gavin Kelly, Jesper Q. Svejstrup, Anna Lobley, Marco Saponaro, Michael Howell, Laura Williamson, Theodoros Kantidakis, Bradley Spencer-Dene, Philip East, Stefan Boeing, Jane Walker, and Aengus Stewart
- Subjects
0301 basic medicine ,Gene isoform ,RNA, Untranslated ,Transcription Elongation, Genetic ,Transcription, Genetic ,Ultraviolet Rays ,non-coding RNA ,RNA polymerase II ,DNA damage response ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell Line ,ASCC3 ,03 medical and health sciences ,Exon ,lncRNA ,Transcription (biology) ,Humans ,RNA, Messenger ,Gene ,alternative last exon splicing ,Transcription Initiation, Genetic ,Messenger RNA ,biology ,Biochemistry, Genetics and Molecular Biology(all) ,Alternative splicing ,DNA Helicases ,RNA ,Exons ,Molecular biology ,Alternative Splicing ,030104 developmental biology ,biology.protein ,transcript elongation ,UV-irradiation - Abstract
Summary The transcription-related DNA damage response was analyzed on a genome-wide scale with great spatial and temporal resolution. Upon UV irradiation, a slowdown of transcript elongation and restriction of gene activity to the promoter-proximal ∼25 kb is observed. This is associated with a shift from expression of long mRNAs to shorter isoforms, incorporating alternative last exons (ALEs) that are more proximal to the transcription start site. Notably, this includes a shift from a protein-coding ASCC3 mRNA to a shorter ALE isoform of which the RNA, rather than an encoded protein, is critical for the eventual recovery of transcription. The non-coding ASCC3 isoform counteracts the function of the protein-coding isoform, indicating crosstalk between them. Thus, the ASCC3 gene expresses both coding and non-coding transcript isoforms with opposite effects on transcription recovery after UV-induced DNA damage., Graphical Abstract, Highlights • UV elicits elongation slowdown and restricts transcription to the 5′ end of genes • UV induces a switch from long to short alternative last exon (ALE) transcript isoforms • ASCC3 short and long ALE isoforms have antagonistic functions in the UV response • The UV-induced ASCC3 short isoform functions as a long non-coding RNA, UV damage generates a functional non-coding RNA through alternative pre-mRNA processing of a damage response factor transcript, identifying a pathway for repurposing protein coding genes under selective conditions.
- Published
- 2017
24. Distinct modes of SMAD2 chromatin binding and remodeling shape the transcriptional response to NODAL/Activin signaling
- Author
-
Philip East, Daniel S. J. Miller, Harshil Patel, Nik Matthews, Caroline S. Hill, Anna Lobley, Davide M Coda, Aengus Stewart, and Tessa Gaarenstroom
- Subjects
0301 basic medicine ,TBX1 ,animal structures ,Mouse ,Transcription, Genetic ,QH301-705.5 ,Nodal Protein ,Science ,Nodal signaling ,Smad2 Protein ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Chromatin remodeling ,SMAD2 ,chromatin remodeling ,03 medical and health sciences ,Mice ,NODAL/Activin ,Animals ,Biology (General) ,Transcription factor ,Activin type 2 receptors ,Genetics ,General Immunology and Microbiology ,General Neuroscience ,Chromatin binding ,Gene Expression Regulation, Developmental ,General Medicine ,Chromatin ,Cell biology ,Activins ,030104 developmental biology ,Genes and Chromosomes ,Medicine ,signaling dynamics ,transcription ,Research Article ,Protein Binding ,Signal Transduction - Abstract
NODAL/Activin signaling orchestrates key processes during embryonic development via SMAD2. How SMAD2 activates programs of gene expression that are modulated over time however, is not known. Here we delineate the sequence of events that occur from SMAD2 binding to transcriptional activation, and the mechanisms underlying them. NODAL/Activin signaling induces dramatic chromatin landscape changes, and a dynamic transcriptional network regulated by SMAD2, acting via multiple mechanisms. Crucially we have discovered two modes of SMAD2 binding. SMAD2 can bind pre-acetylated nucleosome-depleted sites. However, it also binds to unacetylated, closed chromatin, independently of pioneer factors, where it induces nucleosome displacement and histone acetylation. For a subset of genes, this requires SMARCA4. We find that long term modulation of the transcriptional responses requires continued NODAL/Activin signaling. Thus SMAD2 binding does not linearly equate with transcriptional kinetics, and our data suggest that SMAD2 recruits multiple co-factors during sustained signaling to shape the downstream transcriptional program. DOI: http://dx.doi.org/10.7554/eLife.22474.001, eLife digest To allow a complex animal to develop from a small bundle of cells, the cells need to be able to communicate with each other to coordinate their activities. Furthermore, this communication needs to continue in adulthood to keep the body in balance and to prevent diseases such as cancer. The cells communicate by releasing signals that influence the behavior of their neighbors by activating proteins called transcription factors. These proteins then change the activity of particular genes in the nucleus by binding to specific places on a structure called chromatin (the structure in which the genes are packaged). One group of signaling molecules is known as the transforming growth factor beta superfamily, which is crucial for embryos to develop correctly. Failure to control these signals can also promote the growth of tumors. However, it is not clear how the detection of these signals at the surface of the cell leads to changes in the activity of genes inside the nucleus. Two transforming growth factor beta signals called Activin and NODAL cause a transcription factor known as SMAD2 to move into the nucleus where it can alter gene activity. Here Coda, Gaarenstroom et al. investigated how SMAD2 transmits the Activin/Nodal signal in mouse cancer cells. The experiments showed that SMAD2 can change the activities of genes in multiple ways. SMAD2 can bind to places in the chromatin that are either easy to access (which typically contain genes that are already “switched on”) as well as areas that are difficult to access (which generally contain genes that are “switched off”). As a result, SMAD2 increases the activity of genes that were already active, but also switches on on genes that were previously inactive. Coda, Gaarenstroom et al. also found evidence that SMAD2 remained bound to chromatin after long periods of Activin/NODAL signaling. For some genes, this resulted in high gene activity, but in other cases this decreased the gene’s activity. Therefore, future experiments will investigate which other proteins help SMAD2 to change gene activity at later times. DOI: http://dx.doi.org/10.7554/eLife.22474.002
- Published
- 2017
25. Author response: Distinct modes of SMAD2 chromatin binding and remodeling shape the transcriptional response to NODAL/Activin signaling
- Author
-
Aengus Stewart, Daniel S. J. Miller, Anna Lobley, Philip East, Davide M Coda, Nik Matthews, Caroline S. Hill, Tessa Gaarenstroom, and Harshil Patel
- Subjects
Chemistry ,Chromatin binding ,Transcriptional response ,NODAL ,Cell biology - Published
- 2017
- Full Text
- View/download PDF
26. Dysregulation of gene expression as a cause of Cockayne syndrome neurological disease
- Author
-
Gavin Kelly, Philip J. Brooks, Yuming Wang, Aengus Stewart, Jesper Q. Svejstrup, Giampietro Schiavo, Probir Chakravarty, Michael Ranes, and Edward G. Neilan
- Subjects
musculoskeletal diseases ,Genome instability ,DNA repair ,Blotting, Western ,Biology ,Bioinformatics ,Cockayne syndrome ,Cell Line ,Mice ,Cell Line, Tumor ,Gene expression ,medicine ,Animals ,Humans ,Gene Regulatory Networks ,Cockayne Syndrome ,Poly-ADP-Ribose Binding Proteins ,Gene ,Cells, Cultured ,Oligonucleotide Array Sequence Analysis ,Neurons ,Regulation of gene expression ,Multidisciplinary ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,DNA Helicases ,Fibroblasts ,Biological Sciences ,medicine.disease ,Gene expression profiling ,DNA Repair Enzymes ,Gene Ontology ,HEK293 Cells ,Gene Expression Regulation ,Microscopy, Fluorescence ,Cell Transdifferentiation ,Cancer research ,RNA Interference ,Nucleotide excision repair - Abstract
Cockayne syndrome (CS) is a multisystem disorder with severe neurological symptoms. The majority of CS patients carry mutations in Cockayne syndrome group B (CSB), best known for its role in transcription-coupled nucleotide excision repair. Indeed, because various repair pathways are compromised in patient cells, CS is widely considered a genome instability syndrome. Here, we investigate the connection between the neuropathology of CS and dysregulation of gene expression. Transcriptome analysis of human fibroblasts revealed that even in the absence of DNA damage, CSB affects the expression of thousands of genes, many of which are neuronal genes. CSB is present in a significant subset of these genes, suggesting that regulation is direct, at the level of transcription. Importantly, reprogramming of CS fibroblasts to neuron-like cells is defective unless an exogenous CSB gene is introduced. Moreover, neuroblastoma cells from which CSB is depleted show defects in gene expression programs required for neuronal differentiation, and fail to differentiate and extend neurites. Likewise, neuron-like cells cannot be maintained without CSB. Finally, a number of disease symptoms may be explained by marked gene expression changes in the brain of patients with CS. Together, these data point to dysregulation of gene regulatory networks as a cause of the neurological symptoms in CS.
- Published
- 2014
- Full Text
- View/download PDF
27. The Scc2–Scc4 complex acts in sister chromatid cohesion and transcriptional regulation by maintaining nucleosome-free regions
- Author
-
Harshil Patel, Lidia Lopez-Serra, Gavin Kelly, Aengus Stewart, and Frank Uhlmann
- Subjects
Saccharomyces cerevisiae Proteins ,Chromosomal Proteins, Non-Histone ,Micrognathism ,Chromatids ,Article ,Chromatin remodeling ,03 medical and health sciences ,0302 clinical medicine ,De Lange Syndrome ,Gene Expression Regulation, Fungal ,Intellectual Disability ,Genetics ,Humans ,Nucleosome ,Abnormalities, Multiple ,RSC complex ,Chromatin structure remodeling (RSC) complex ,Promoter Regions, Genetic ,Oligonucleotide Array Sequence Analysis ,030304 developmental biology ,Cohesin loading ,0303 health sciences ,Binding Sites ,biology ,Cohesin ,Gene Expression Profiling ,Chromatin ,Nucleosomes ,Establishment of sister chromatid cohesion ,Face ,biology.protein ,biological phenomena, cell phenomena, and immunity ,Chromosomes, Fungal ,Transcription Initiation Site ,Separase ,Hand Deformities, Congenital ,Neck ,030217 neurology & neurosurgery ,Protein Binding - Abstract
The cohesin complex is at the heart of many chromosomal activities, including sister chromatid cohesion and transcriptional regulation. Cohesin loading onto chromosomes depends on the Scc2-Scc4 cohesin loader complex, but the chromatin features that form cohesin loading sites remain poorly understood. Here we show that the RSC chromatin remodeling complex recruits budding yeast Scc2-Scc4 to broad nucleosome-free regions, which the cohesin loader helps to maintain. Consequently, inactivation of either the cohesin loader or the RSC complex has similar effects on nucleosome positioning, gene expression and sister chromatid cohesion. These results show an intimate link between local chromatin structure and higher-order chromosome architecture. Our findings pertain to the similarities between two severe human disorders, Cornelia de Lange syndrome, which is caused by alterations in the human cohesin loader, and Coffin-Siris syndrome, which results from alterations in human RSC complex components. Both syndromes can arise from gene misregulation due to related changes in the nucleosome landscape.
- Published
- 2014
- Full Text
- View/download PDF
28. Rho-actin signaling to the MRTF coactivators dominates the immediate transcriptional response to serum in fibroblasts
- Author
-
Richard Treisman, Francesco Gualdrini, Cyril Esnault, Stuart Horswell, Phil East, Aengus Stewart, Nik Matthews, Lincoln's Inn Fields Laboratories, and Cancer Research UK
- Subjects
Serum ,MESH: Signal Transduction ,MESH: Period Circadian Proteins ,MESH: Circadian Clocks ,Transcription, Genetic ,genetic structures ,[SDV]Life Sciences [q-bio] ,RNA polymerase II ,Biology ,MESH: Actins ,Rho ,Transcription (biology) ,MRTF ,Gene expression ,Serum response factor ,Genetics ,Animals ,MESH: Animals ,CLOCK Proteins ,MESH: Mice ,Transcription factor ,MESH: Transcription, Genetic ,MESH: Serum ,TCF ,Fibroblasts ,MESH: CLOCK Proteins ,MESH: Mitogen-Activated Protein Kinases ,Molecular biology ,Actins ,Cell biology ,Chromatin ,MESH: Serum Response Factor ,MESH: Fibroblasts ,embryonic structures ,cardiovascular system ,biology.protein ,chromatin ,SRF ,Chromatin immunoprecipitation ,signal transduction ,Research Paper ,Developmental Biology - Abstract
International audience; The transcription factor SRF (serum response factor) recruits two families of coactivators, the MRTFs (myocardin-related transcription factors) and the TCFs (ternary complex factors), to couple gene transcription to growth factor signaling. Here we investigated the role of the SRF network in the immediate transcriptional response of fibroblasts to serum stimulation. SRF recruited its cofactors in a gene-specific manner, and virtually all MRTF binding was directed by SRF. Much of SRF DNA binding was serum-inducible, reflecting a requirement for MRTF-SRF complex formation in nucleosome displacement. We identified 960 serum-responsive SRF target genes, which were mostly MRTF-controlled, as assessed by MRTF chromatin immunoprecipitation (ChIP) combined with deep sequencing (ChIP-seq) and/or sensitivity to MRTF-linked signals. MRTF activation facilitates RNA polymerase II (Pol II) recruitment or promoter escape according to gene context. MRTF targets encode regulators of the cytoskeleton, transcription, and cell growth, underpinning the role of SRF in cytoskeletal dynamics and mechanosensing. Finally, we show that specific activation of either MRTFs or TCFs can reset the circadian clock.
- Published
- 2014
- Full Text
- View/download PDF
29. RECQL5 Controls Transcript Elongation and Suppresses Genome Instability Associated with Transcription Stress
- Author
-
Richard Mitter, Aengus Stewart, Marco Saponaro, Johannes Söding, Theodoros Kantidakis, Gavin Kelly, Hannah Williams, Jesper Q. Svejstrup, and Mark Heron
- Subjects
Genome instability ,Genetics ,Transcription Elongation, Genetic ,biology ,General transcription factor ,RecQ Helicases ,Transcription, Genetic ,Biochemistry, Genetics and Molecular Biology(all) ,Genome, Human ,Chromosomal fragile site ,Helicase ,RNA polymerase II ,General Biochemistry, Genetics and Molecular Biology ,Article ,Genomic Instability ,Elongation factor ,HEK293 Cells ,Transcription (biology) ,biology.protein ,Humans ,RNA Polymerase II ,Gene - Abstract
Summary RECQL5 is the sole member of the RECQ family of helicases associated with RNA polymerase II (RNAPII). We now show that RECQL5 is a general elongation factor that is important for preserving genome stability during transcription. Depletion or overexpression of RECQL5 results in corresponding shifts in the genome-wide RNAPII density profile. Elongation is particularly affected, with RECQL5 depletion causing a striking increase in the average rate, concurrent with increased stalling, pausing, arrest, and/or backtracking (transcription stress). RECQL5 therefore controls the movement of RNAPII across genes. Loss of RECQL5 also results in the loss or gain of genomic regions, with the breakpoints of lost regions located in genes and common fragile sites. The chromosomal breakpoints overlap with areas of elevated transcription stress, suggesting that RECQL5 suppresses such stress and its detrimental effects, and thereby prevents genome instability in the transcribed region of genes., Graphical Abstract, Highlights • RECQL5 is a general RNAPII elongation factor • RECQL5 reduces the elongation rate while decreasing pausing and arrest events • Loss of RECQL5 results in genome instability in genes and at common fragile sites • Incidents of genome instability colocalize with pause and arrest events, Rapid elongation by RNA polymerase II leads to increased transcriptional stress including a high incidence of pausing and arrests, which correlates with sites of genomic instability. RECQL5 modulates the rate of transcription, mitigating both the stress and instability effects.
- Published
- 2014
30. SCAF4 and SCAF8, mRNA Anti-Terminator Proteins
- Author
-
Nick J. Proudfoot, Richard Mitter, Reuven Agami, Jesper Q. Svejstrup, Aengus Stewart, Takayuki Nojima, Alejandro Pineiro Ugalde, and Lea H. Gregersen
- Subjects
Model organisms ,Transcription Elongation, Genetic ,Polyadenylation ,Cell ,Gene Expression ,co-transcriptional mRNA processing ,RNA polymerase II ,Biology ,Biochemistry & Proteomics ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Protein Domains ,Gene expression ,medicine ,Humans ,RNA, Messenger ,polyadenylation ,anti-termination ,Computational & Systems Biology ,030304 developmental biology ,Chemical Biology & High Throughput ,0303 health sciences ,Messenger RNA ,transcriptional termination ,Serine-Arginine Splicing Factors ,C-terminal repeat domain (CTD) ,SCAF8 ,Genome Integrity & Repair ,RNA-Binding Proteins ,Eukaryota ,SCAF4 ,Tumour Biology ,Cell biology ,Elongation factor ,HEK293 Cells ,medicine.anatomical_structure ,Terminator (genetics) ,CTD phosphorylation ,Transcription Termination, Genetic ,biology.protein ,Computer-Aided Design ,Phosphorylation ,Poly A ,Genetics & Genomics ,030217 neurology & neurosurgery ,transcript elongation - Abstract
Summary Accurate regulation of mRNA termination is required for correct gene expression. Here, we describe a role for SCAF4 and SCAF8 as anti-terminators, suppressing the use of early, alternative polyadenylation (polyA) sites. The SCAF4/8 proteins bind the hyper-phosphorylated RNAPII C-terminal repeat domain (CTD) phosphorylated on both Ser2 and Ser5 and are detected at early, alternative polyA sites. Concomitant knockout of human SCAF4 and SCAF8 results in altered polyA selection and subsequent early termination, leading to expression of truncated mRNAs and proteins lacking functional domains and is cell lethal. While SCAF4 and SCAF8 work redundantly to suppress early mRNA termination, they also have independent, non-essential functions. SCAF8 is an RNAPII elongation factor, whereas SCAF4 is required for correct termination at canonical, distal transcription termination sites in the presence of SCAF8. Together, SCAF4 and SCAF8 coordinate the transition between elongation and termination, ensuring correct polyA site selection and RNAPII transcriptional termination in human cells., Graphical Abstract, Highlights • Human SCAF4 and SCAF8 couple RNAPII Ser2P- and Ser5P-binding and RNA processing • SCAF4 and SCAF8 bind nascent RNA upstream of early polyadenylation sites • SCAF4 and SCAF8 prevent early mRNA transcript cleavage and polyadenylation • Lack of SCAF4 and SCAF8 result in truncated protein products and is cell lethal, Eukaryotic anti-terminator proteins suppress usage of early polyadenylation sites to prevent production of truncated proteins.
- Published
- 2019
- Full Text
- View/download PDF
31. A ubiquitylation site in Cockayne syndrome B required for repair of oxidative DNA damage, but not for transcription-coupled nucleotide excision repair
- Author
-
Yuming Wang, Vesela Encheva, Giuseppina Giglia-Mari, Hervé Menoni, Michael Ranes, Jane Walker, Franziska Wienholz, Ambrosius P. Snijders, Jesper Q. Svejstrup, Probir Chakravarty, Pierre-Olivier Mari, Aengus Stewart, Stefan Boeing, Wim Vermeulen, Department of Molecular Genetics [Rotterdam, The Netherlands] (Erasmus MC), Oncode Institute [Rotterdam, The Netherlands]-Cancer Genomics Netherlands [Rotterdam, The Netherlands], Institute for Advanced Biosciences / Institut pour l'Avancée des Biosciences (Grenoble) (IAB), Centre Hospitalier Universitaire [Grenoble] (CHU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Etablissement français du sang - Auvergne-Rhône-Alpes (EFS)-Université Grenoble Alpes [2016-2019] (UGA [2016-2019]), Cancer Research UK, Bioinformatics and Biostatistics Service, The Francis Crick Institute [London], and Molecular Genetics
- Subjects
0301 basic medicine ,Genome instability ,musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,DNA Repair ,Transcription, Genetic ,DNA damage ,DNA repair ,Cell Survival ,Recombinant Fusion Proteins ,[SDV]Life Sciences [q-bio] ,Gene Expression ,Genome Integrity, Repair and Replication ,medicine.disease_cause ,Cockayne syndrome ,Genomic Instability ,Cell Line ,03 medical and health sciences ,Ubiquitin ,Genetics ,medicine ,Cluster Analysis ,Humans ,Amino Acid Sequence ,Cockayne Syndrome ,Gene ,ComputingMilieux_MISCELLANEOUS ,Mutation ,biology ,Gene Expression Profiling ,Cell Cycle ,Ubiquitination ,nutritional and metabolic diseases ,[SDV.BBM.BM]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,medicine.disease ,Molecular biology ,Oxidative Stress ,030104 developmental biology ,biology.protein ,Nucleotide excision repair ,DNA Damage - Abstract
Cockayne syndrome B (CSB), best known for its role in transcription-coupled nucleotide excision repair (TC-NER), contains a ubiquitin-binding domain (UBD), but the functional connection between protein ubiquitylation and this UBD remains unclear. Here, we show that CSB is regulated via site-specific ubiquitylation. Mass spectrometry analysis of CSB identified lysine (K) 991 as a ubiquitylation site. Intriguingly, mutation of this residue (K991R) does not affect CSB's catalytic activity or protein stability, but greatly affects genome stability, even in the absence of induced DNA damage. Moreover, cells expressing CSB K991R are sensitive to oxidative DNA damage, but proficient for TC-NER. K991 becomes ubiquitylated upon oxidative DNA damage, and while CSB K991R is recruited normally to such damage, it fails to dissociate in a timely manner, suggesting a requirement for K991 ubiquitylation in CSB activation. Interestingly, deletion of CSB's UBD gives rise to oxidative damage sensitivity as well, while CSB ΔUBD and CSB K991R affects expression of overlapping groups of genes, further indicating a functional connection. Together, these results shed new light on the regulation of CSB, with K991R representing an important separation-of-function-mutation in this multi-functional protein.
- Published
- 2016
- Full Text
- View/download PDF
32. Mutation of cancer driver MLL2 results in transcription stress and genome instability
- Author
-
Marco Saponaro, A. Francis Stewart, Gavin Kelly, Aengus Stewart, Theodoros Kantidakis, Stuart Horswell, Nik Matthews, Ozan Aygün, Richard Mitter, Jesper Q. Svejstrup, Andrea Kranz, Stefan Boeing, Massachusetts Institute of Technology. Department of Chemistry, and Aygun, Ozan
- Subjects
0301 basic medicine ,Genome instability ,Transcription, Genetic ,RNA polymerase II ,Sister chromatid exchange ,Biology ,medicine.disease_cause ,Genomic Instability ,Cell Line ,03 medical and health sciences ,Mice ,Transcription (biology) ,Genetics ,medicine ,Animals ,Humans ,Gene ,RecQ Helicases ,Chromosomal fragile site ,Histone-Lysine N-Methyltransferase ,3. Good health ,030104 developmental biology ,Histone methyltransferase ,Mutation ,biology.protein ,RNA Polymerase II ,Carcinogenesis ,Myeloid-Lymphoid Leukemia Protein ,Developmental Biology ,DNA Damage ,Research Paper - Abstract
Genome instability is a recurring feature of tumorigenesis. Mutation in MLL2, encoding a histone methyltransferase, is a driver in numerous different cancer types, but the mechanism is unclear. Here, we present evidence that MLL2 mutation results in genome instability. Mouse cells in which MLL2 gene deletion can be induced display elevated levels of sister chromatid exchange, gross chromosomal aberrations, 53BP1 foci, and micronuclei. Human MLL2 knockout cells are characterized by genome instability as well. Interestingly, MLL2 interacts with RNA polymerase II (RNAPII) and RECQL5, and, although MLL2 mutated cells have normal overall H3K4me levels in genes, nucleosomes in the immediate vicinity of RNAPII are hypomethylated. Importantly, MLL2 mutated cells display signs of substantial transcription stress, and the most affected genes overlap with early replicating fragile sites, show elevated levels of γH2AX, and suffer frequent mutation. The requirement for MLL2 in the maintenance of genome stability in genes helps explain its widespread role in cancer and points to transcription stress as a strong driver in tumorigenesis., Francis Crick Institute (grant no. FCI01), Deutsche Krebshilfe (grant), European Research Council (grant), Association for International Cancer Research (grant)
- Published
- 2016
33. Intratumor Heterogeneity and Branched Evolution Revealed by Multiregion Sequencing
- Author
-
Gordon Stamp, Andrew Rowan, Claudio R. Santos, Bradley Spencer-Dene, Aron Charles Eklund, Adam Butler, Patrick S. Tarpey, Lisa Pickering, Nicholas Matthews, Stuart Horswell, Calli Latimer, Julian Downward, James Larkin, Neil Q. McDonald, Aengus Stewart, Zoltan Szallasi, Marco Gerlinger, David Endesfelder, David T. Jones, Martin Gore, Graham Clark, Keiran Raine, P. Andrew Futreal, Sharmin Begum, Eva Grönroos, Ignacio Varela, Charles Swanton, Pierre Martinez, Benjamin Phillimore, and Mahrokh Nohadani
- Subjects
Tumour heterogeneity ,Biopsy ,Kidney ,Polymorphism, Single Nucleotide ,Somatic evolution in cancer ,Chromosome aberration ,Intratumoral Genetic Heterogeneity ,Article ,Evolution, Molecular ,Genetic Heterogeneity ,Biomarkers, Tumor ,Humans ,PTEN ,Exome ,Everolimus ,Neoplasm Metastasis ,Kinase activity ,Carcinoma, Renal Cell ,Phylogeny ,Chromosome Aberrations ,Sirolimus ,Genetics ,Ploidies ,biology ,Genetic heterogeneity ,Sequence Analysis, DNA ,General Medicine ,Kidney Neoplasms ,Phenotype ,Mutation ,biology.protein ,Immunosuppressive Agents - Abstract
Background Intratumor heterogeneity may foster tumor evolution and adaptation and hinder personalized-medicine strategies that depend on results from single tumor-biopsy samples. Methods To examine intratumor heterogeneity, we performed exome sequencing, chromosome aberration analysis, and ploidy profiling on multiple spatially separated samples obtained from primary renal carcinomas and associated metastatic sites. We characterized the consequences of intratumor heterogeneity using immunohistochemical analysis, mutation functional analysis, and profiling of messenger RNA expression. Results Phylogenetic reconstruction revealed branched evolutionary tumor growth, with 63 to 69% of all somatic mutations not detectable across every tumor region. Intratumor heterogeneity was observed for a mutation within an autoinhibitory domain of the mammalian target of rapamycin (mTOR) kinase, correlating with S6 and 4EBP phosphorylation in vivo and constitutive activation of mTOR kinase activity in vitro. Mutational intratumor heterogeneity was seen for multiple tumor-suppressor genes converging on loss of function; SETD2, PTEN, and KDM5C underwent multiple distinct and spatially separated inactivating mutations within a single tumor, suggesting convergent phenotypic evolution. Gene-expression signatures of good and poor prognosis were detected in different regions of the same tumor. Allelic composition and ploidy profiling analysis revealed extensive intratumor heterogeneity, with 26 of 30 tumor samples from four tumors harboring divergent allelic-imbalance profiles and with ploidy heterogeneity in two of four tumors. Conclusions Intratumor heterogeneity can lead to underestimation of the tumor genomics landscape portrayed from single tumor-biopsy samples and may present major challenges to personalized-medicine and biomarker development. Intratumor heterogeneity, associated with heterogeneous protein function, may foster tumor adaptation and therapeutic failure through Darwinian selection. (Funded by the Medical Research Council and others.)
- Published
- 2012
- Full Text
- View/download PDF
34. Tracking Cancer Evolution Reveals Constrained Routes to Metastases: TRACERx Renal
- Author
-
Archana Fernando, George F. Mayhew, Lavinia Spain, Thomas B.K. Watkins, Samantha M. Hill, Aspasia Soultati, Maria F. Becerra, Rosa Guarch, James Larkin, Charles Swanton, Samra Turajlic, David Nicol, Mariam Jamal-Hanjani, Steve Hazell, Simon Chowdhury, Ian Proctor, Mark Stares, Todd Richmond, Stuart Horswell, Sophia Ward, Claudia Eichler-Jonsson, Martin Gore, Aengus Stewart, Ed Reznik, Renzo G. DiNatale, Daniel Burgess, Andrew Rowan, Emma Nye, James J. Hsieh, Tim Chambers, Ben Challacombe, Stacey Stanislaw, Nelson Alexander, José I. López, Faiz Jabbar, Ashish Chandra, Gordon Stamp, Hang Xu, Catherine D. McNally, Kevin Litchfield, Sarah Rudman, Heidi Rosenbaum, Joanna Lynch, Lisa Pickering, Mary Falzon, Tim O'Brien, Lewis Au, Sharanpreet Lall, and Carol Jones
- Subjects
0301 basic medicine ,cell carcinoma ,branched evolution ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Metastasis ,prostate-cancer ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Breast cancer ,tumor thrombus ,Renal cell carcinoma ,Pancreatic cancer ,pancreatic-cancer ,expression ,medicine ,health care economics and organizations ,breast-cancer ,sequencing data ,natural-history ,medicine.disease ,Primary tumor ,3. Good health ,030104 developmental biology ,030220 oncology & carcinogenesis ,Monoclonal ,Cancer research ,Metastasectomy ,cytoreductive nephrectomy - Abstract
Clear-cell renal cell carcinoma (ccRCC) exhibits a broad range of metastatic phenotypes that have not been systematically studied to date. Here, we analyzed 575 primary and 335 metastatic biopsies across 100 patients with metastatic ccRCC, including two cases sampledat post-mortem. Metastatic competence was afforded by chromosome complexity, and we identify 9p loss as a highly selected event driving metastasis and ccRCC-related mortality (p = 0.0014). Distinct patterns of metastatic dissemination were observed, including rapid progression to multiple tissue sites seeded by primary tumors of monoclonal structure. By contrast, we observed attenuated progression in cases characterized by high primary tumor heterogeneity, with metastatic competence acquired gradually and initial progression to solitary metastasis. Finally, we observed early divergence of primitive ancestral clones and protracted latency of up to two decades as a feature of pancreatic metastases. S.T. and H.X. are funded by Cancer Research UK (CRUK) (C50947/A18176). S.T., T.C., J.L., and M.G. are funded by the NIH Research (NIHR) Biomedical Research Centre (BRC) at the Royal Marsden Hospital and Institute of Cancer Research (A109). J.I.L. is funded by the Ministerio de Economia y Competitividad (MINECO, SAF2016-79847-R). M.S., A.S., J.L., R.F., L.A., and L.S. are funded by the Royal Marsden Cancer Charity. K.L. is funded by UK Medical Research Council (MR/P014712/1). N.M. receives funding from CRUK, Rosetrees, and the NIHR BRC at University College London Hospitals. C.S is Royal Society Napier Research Professor. C.S. is funded by Cancer Research UK (TRACERx and CRUK Cancer Immunotherapy Catalyst Network), the CRUK Lung Cancer Centre of Excellence, Stand Up 2 Cancer (SU2C), the Rosetrees and Stoneygate Trusts, NovoNordisk Foundation (ID 16584), the Breast Cancer Research Foundation (BCRF), the European Research Council (THESEUS), Marie Curie Network PloidyNet, the NIHR BRC at University College London Hospitals, and the CRUK University College London Experimental Cancer Medicine Centre. The work presented in this manuscript was funded by Cancer Research UK (grant reference number C50947/A18176), Ventana Medical Systems (grant reference numbers 10467 and 10530), the Kidney Cancer Fund of The Royal Marsden Cancer Charity, NIHR BRC at the Royal Marsden Hospital and Institute of Cancer Research (grant reference number A109), and the Francis Crick Institute, which receives its core funding from Cancer Research UK (FC001202), the UK Medical Research Council (FC001202), and the Wellcome Trust (FC001202). In particular, we acknowledge the support of the Advanced Sequencing Facility and the High-Performance Computing at the Francis Crick Institute. This project was enabled through access to the MRC eMedLab Medical Bioinformatics infrastructure, supported by the Medical Research Council (grant number MR/L016311/1).
- Published
- 2018
- Full Text
- View/download PDF
35. Erratum: Corrigendum: Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution
- Author
-
Martin Hayward, Jason F. Lester, Thomas B.K. Watkins, Selvaraju Veeriah, Joan K. Riley, Phil Crosbie, Tudor Constantin, Matthew Rabinowitz, Wenya Linda Bi, Karl S. Peggs, Nicholas McGranahan, Francisco Zambrana, Marianne Nicolson, Samra Turajlic, Gerald Langman, Raheleh Salari, Rajesh Shah, Simon Trotter, Malgorzata Kornaszewska, Bernhard Zimmermann, Hugo J.W.L. Aerts, Tanya Ahmad, Sridhar Rathinam, Siow Ming Lee, Ayse U. Akarca, Christopher Abbosh, Fiona H Blackhall, Dina Hafez, Nicolai Juul Birkbak, Peter Russell, David Moore, Lesley Gomersall, Nicole Sponer, Apostolos Nakas, Gareth A. Wilson, John A. Hartley, Melanie Irvin-Sellers, Sophia Ward, Veni Ezhil, Fiona M. Fennessy, Aengus Stewart, Anne Marie Quinn, Peter Van Loo, Eser Kirkizlar, Vineet Prakash, Tim Chambers, Zoltan Szallasi, Seema Shafi, Ricky Thakrar, Dahmane Oukrif, Sergio A. Quezada, Sajid A. Khan, Babu Naidu, Harriet Bell, Sam M. Janes, Mariam Jamal-Hanjani, Andrew N. Rowan, Mary Falzon, Asia Ahmed, Eva Grönroos, Justyna Czyzewska-Khan, Stephanie Kareht, Mahendran Chetty, Dean A. Fennell, Ashwini Naik, Helen Lowe, Roland F. Schwarz, Allan Hackshaw, Apratim Ganguly, Joanne Laycock, Crispin T. Hiley, Shyam Kolvekar, Elaine Borg, Yvonne Summers, Diana Johnson, Martin Forster, David Lawrence, Greg Elgar, Richard Attanoos, Keith M. Kerr, Charles Swanton, Rachel Rosenthal, Teresa Marafioti, Maise Al Bakir, Girija Anand, Hardy Remmen, Gary Middleton, Francesco Fraioli, Luke Martinson, Leena Dennis Joseph, C. Jimmy Lin, Natasha Iles, Yenting Ngai, Caroline Dive, Nikolaos Panagiotopoulos, Nik Matthews, Haydn Adams, John Le Quesne, Helen Davies, Jacqui Shaw, Sara Lock, Babikir Ismail, Javier Herrero, and Raymondo Endozo
- Subjects
0301 basic medicine ,Multidisciplinary ,medicine.diagnostic_test ,business.industry ,Volumetric data ,Computed tomography ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Positron emission tomography ,030220 oncology & carcinogenesis ,Medicine ,Stage (cooking) ,business ,Lung cancer ,Nuclear medicine - Abstract
Nature 545, 446–451 (2017); doi:10.1038/nature22364 For 6 of the 96 patients included in this Article (patients CRUK0014, CRUK0030, CRUK0048, CRUK0059, CRUK0096 and CRUK0097) incorrect tumour volumetric data and positron emission tomography (PET) tumour background ratio (TBR) data were analysed. This error occurred because of the incorrect assignment of patient identifiers during the anonymization mandated by the independent review board of pre-operative computed tomography (CT) scans belonging to these patients.
- Published
- 2017
- Full Text
- View/download PDF
36. A simple biophysical model emulates budding yeast chromosome condensation
- Author
-
Raphael A. G. Chaleil, Carmay Lim, Tammy M. K. Cheng, Paul A. Bates, Sebastian Heeger, Aengus Stewart, Nik Matthews, Jon D. Wright, and Frank Uhlmann
- Subjects
Models, Molecular ,Saccharomyces cerevisiae Proteins ,QH301-705.5 ,Science ,Condensin ,Saccharomyces cerevisiae ,S. cerevisiae ,Gene Expression ,macromolecular substances ,Computational biology ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Condensin complex ,Nucleosome ,Biology (General) ,Interphase ,Mathematical Computing ,Mitosis ,mitosis ,Adenosine Triphosphatases ,Genetics ,Stochastic Processes ,Binding Sites ,General Immunology and Microbiology ,biology ,condensin ,General Neuroscience ,Chromosome ,General Medicine ,Chromatin Assembly and Disassembly ,biology.organism_classification ,Nucleosomes ,Chromatin ,DNA-Binding Proteins ,Genes and Chromosomes ,Multiprotein Complexes ,Premature chromosome condensation ,chromosome architecture ,biology.protein ,Medicine ,Chromosomes, Fungal ,Research Article ,Computational and Systems Biology ,Protein Binding - Abstract
Mitotic chromosomes were one of the first cell biological structures to be described, yet their molecular architecture remains poorly understood. We have devised a simple biophysical model of a 300 kb-long nucleosome chain, the size of a budding yeast chromosome, constrained by interactions between binding sites of the chromosomal condensin complex, a key component of interphase and mitotic chromosomes. Comparisons of computational and experimental (4C) interaction maps, and other biophysical features, allow us to predict a mode of condensin action. Stochastic condensin-mediated pairwise interactions along the nucleosome chain generate native-like chromosome features and recapitulate chromosome compaction and individualization during mitotic condensation. Higher order interactions between condensin binding sites explain the data less well. Our results suggest that basic assumptions about chromatin behavior go a long way to explain chromosome architecture and are able to generate a molecular model of what the inside of a chromosome is likely to look like. DOI: http://dx.doi.org/10.7554/eLife.05565.001, eLife digest The genetic material of living things is made up of long strands of DNA. Human cells contain about two meters of DNA split between 46 chromosomes. These chromosomes carry all the instructions to build a human body. To fit all of this information inside each human cell, the DNA is wrapped around hundreds of thousands of proteins such that the chromosomes each resemble a string of beads. Most of the time the chromosomes in a cell are only loosely arranged. But, when a cell prepares to divide into two new cells, its chromosomes become more compacted. This allows the DNA to withstand the physical forces involved when the copies of the chromosomes are pulled into the two daughter cells, and it makes it easier for the cell to handle its genetic material. If a chromosome breaks during cell division, it can result in diseases such as cancer. Several proteins—collectively called condensins—work to compact (or condense) the chromosomes. These proteins are found in a wide range of species, but it remains poorly understood how they cause chromosomes to become more compact. Due to the technical limitations of current imaging methods, it has not been possible to directly visualize the path of the DNA strand within a compacted chromosome. However, Cheng et al. have now overcome this limitation by combining experimental analyses and computational simulations. Cheng et al. used computer modeling to simulate a piece of chromosome that was about the same size as a chromosome from a single-celled microorganism called budding yeast. This model could accurately recreate the behavior of chromosomes as observed in non-dividing cells—and revealed that these chromosomes are in a relaxed state. Cheng et al. then modeled what happens when condensins are introduced. As expected, the chromosomes became more compacted and the model's behavior was then validated using further experiments. This predicted that condensin complexes, bound to regions along the chromosome's length, interact to form pairs that continually separate and form new pairs with other condensins; and that these ‘dynamic pairwise’ interactions compact the chromosome. The current model describes a relatively small chromosome and, in the future, extending the model to larger chromosomes could shed insight. DOI: http://dx.doi.org/10.7554/eLife.05565.002
- Published
- 2015
- Full Text
- View/download PDF
37. Author response: A simple biophysical model emulates budding yeast chromosome condensation
- Author
-
Sebastian Heeger, Frank Uhlmann, Jon D. Wright, Raphael A. G. Chaleil, Paul A. Bates, Aengus Stewart, Carmay Lim, Tammy Mk Cheng, and Nik Matthews
- Subjects
Simple (abstract algebra) ,Premature chromosome condensation ,Biology ,Budding yeast ,Cell biology - Published
- 2015
- Full Text
- View/download PDF
38. SETD2 loss-of-function promotes renal cancer branched evolution through replication stress and impaired DNA repair
- Author
-
Thomas Powles, Charles Swanton, Gareth A. Wilson, Michael R. Stratton, James Larkin, Sakshi Gulati, Claudio R. Santos, Nnennaya Kanu, Harshil Patel, Apolinar Maya-Mendoza, Paul A. Bates, Aengus Stewart, Martin Mistrik, Jiri Bartek, Marco Gerlinger, Nicolai Juul Birkbak, Zoltan Szallasi, Rebecca A. Burrell, Nicholas McGranahan, P East, Andrew Rowan, Eva Grönroos, Tejal Joshi, Pierre Martinez, X. Yi Goh, Jirina Bartkova, Adam Rabinowitz, Ludmil B. Alexandrov, and Nik Matthews
- Subjects
DNA Replication ,Cancer Research ,DNA Repair ,DNA damage ,DNA polymerase ,DNA repair ,Histones ,Genetic Heterogeneity ,Minichromosome maintenance ,SDG 3 - Good Health and Well-being ,Cell Line, Tumor ,Genetics ,Humans ,Molecular Biology ,Carcinoma, Renal Cell ,biology ,DNA replication ,Histone-Lysine N-Methyltransferase ,Kidney Neoplasms ,Chromatin ,Nucleosomes ,Histone ,Mutation ,biology.protein ,Cancer research ,Microsatellite Instability ,Homologous recombination - Abstract
Defining mechanisms that generate intratumour heterogeneity and branched evolution may inspire novel therapeutic approaches to limit tumour diversity and adaptation. SETD2 (Su(var), Enhancer of zeste, Trithorax-domain containing 2) trimethylates histone-3 lysine-36 (H3K36me3) at sites of active transcription and is mutated in diverse tumour types, including clear cell renal carcinomas (ccRCCs). Distinct SETD2 mutations have been identified in spatially separated regions in ccRCC, indicative of intratumour heterogeneity. In this study, we have addressed the consequences of SETD2 loss-of-function through an integrated bioinformatics and functional genomics approach. We find that bi-allelic SETD2 aberrations are not associated with microsatellite instability in ccRCC. SETD2 depletion in ccRCC cells revealed aberrant and reduced nucleosome compaction and chromatin association of the key replication proteins minichromosome maintenance complex component (MCM7) and DNA polymerase δ hindering replication fork progression, and failure to load lens epithelium-derived growth factor and the Rad51 homologous recombination repair factor at DNA breaks. Consistent with these data, we observe chromosomal breakpoint locations are biased away from H3K36me3 sites in SETD2 wild-type ccRCCs relative to tumours with bi-allelic SETD2 aberrations and that H3K36me3-negative ccRCCs display elevated DNA damage in vivo. These data suggest a role for SETD2 in maintaining genome integrity through nucleosome stabilization, suppression of replication stress and the coordination of DNA repair.
- Published
- 2014
- Full Text
- View/download PDF
39. Spatial and temporal diversity in genomic instability processes defines lung cancer evolution
- Author
-
Gordon Stamp, Mariam Jamal-Hanjani, Peter J. Campbell, Lucy R. Yates, Stuart Horswell, Clarence C. Lee, Bradley Spencer-Dene, Peter Van Loo, Nik Matthews, Doris Rassl, Thierry Voet, Martin Forster, Richard Mitter, Nirupa Murugaesu, John Marshall, Adam Rabinowitz, Enock Teefe, Siow Ming Lee, David T. Jones, Zoltan Szallasi, Marco Gerlinger, Seema Shafi, Sam M. Janes, Charles Swanton, Warren Tom, Andrew Rowan, David C. Wedge, Max Salm, Elza C de Bruin, Mary Falzon, Benjamin Phillimore, David Lawrence, Robert C. Rintoul, Timothy T. Harkins, Shann-Ching Chen, Tanya Ahmad, Aengus Stewart, Sharmin Begum, Nicholas McGranahan, Ignacio Varela, Madiha A. Muhammad, Eva Grönroos, Arrigo Capitanio, Rintoul, Robert [0000-0003-3875-3780], Apollo - University of Cambridge Repository, Rosetrees Trust, Medical Research Council (UK), Ministerio de Economía y Competitividad (España), University of Cambridge, Research Foundation - Flanders, European Commission, Cancer Research UK, Prostate Cancer Foundation, Breast Cancer Research Foundation, University College London, National Institute for Health Research (UK), and European Research Council
- Subjects
Genome instability ,APOBEC ,Cytidine deaminase activity ,Lung Neoplasms ,APOBEC-1 Deaminase ,Gene Dosage ,Context (language use) ,Biology ,medicine.disease_cause ,Translocation, Genetic ,Article ,Genomic Instability ,Evolution, Molecular ,Genetic Heterogeneity ,Chromosome instability ,Cytidine Deaminase ,Carcinoma, Non-Small-Cell Lung ,medicine ,Tumor Cells, Cultured ,Humans ,Genetics ,Mutation ,Multidisciplinary ,Genetic heterogeneity ,Smoking ,Cytidine deaminase ,Prognosis ,3. Good health ,respiratory tract diseases ,Carcinogens ,Neoplasm Recurrence, Local - Abstract
PMCID: PMC4636050.-- et al., Spatial and temporal dissection of the genomic changes occurring during the evolution of human non-small cell lung cancer (NSCLC) may help elucidate the basis for its dismal prognosis.We sequenced 25 spatially distinct regions from seven operable NSCLCs and found evidence of branched evolution, with driver mutations arising before and after subclonal diversification. There was pronounced intratumor heterogeneity in copy number alterations, translocations, and mutations associated with APOBEC cytidine deaminase activity. Despite maintained carcinogen exposure, tumors from smokers showed a relative decrease in smoking-related mutations over time, accompanied by an increase in APOBEC-associated mutations. In tumors from former smokers, genome-doubling occurred within a smoking-signature context before subclonal diversification, which suggested that a long period of tumor latency had preceded clinical detection. The regionally separated driver mutations, coupled with the relentless and heterogeneous nature of the genome instability processes, are likely to confound treatment success in NSCLC., E.B. is a Rosetrees Trust fellow; M.J.H. has a Cancer Research UK fellowship; N.Mu. received funding from the Rosetrees Trust; M.G. is funded by the UK Medical Research Council; I.V. is funded by Spanish Ministerio de Economía y Competitividad subprograma Ramón y Cajal; R.C.R. and D.M.R. are partly funded by the Cambridge Biomedical Research Centre and Cancer Research UK Cancer Centre; P.V.L. is a postdoctoral researcher of the Research Foundation—Flanders (FWO); S.M.J. is a Wellcome Senior Fellow in Clinical Science; and C.S. is a senior Cancer Research UK clinical research fellow and is funded by Cancer Research UK, the Rosetrees Trust, European Union Framework Programme 7 (projects PREDICT and RESPONSIFY, ID:259303), the Prostate Cancer Foundation, the European Research Council and the Breast Cancer Research Foundation. This research is supported by the National Institute for Health Research University College London Hospitals Biomedical Research Centre.
- Published
- 2014
40. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing
- Author
-
Charles Swanton, Rosalie Fisher, Sakshi Gulati, Martin Gore, Marco Gerlinger, Claudio R. Santos, Aengus Stewart, Andrew Rowan, James Larkin, Ignacio Varela, Max Salm, Gordon Stamp, Nicholas Matthews, Steven Hazell, Adam Rabinowitz, Nicholas McGranahan, Pierre Martinez, Benjamin Phillimore, Bradley Spencer-Dene, Paul A. Bates, Sharmin Begum, David Nicol, Stuart Horswell, Lisa Pickering, P. Andrew Futreal, Universidad de Cantabria, National Institute for Health Research (UK), Ministerio de Economía y Competitividad (España), Rosetrees Trust, Breast Cancer Research Foundation, Royal Marsden NHS Foundation Trust, University College London, Medical Research Council (UK), Cancer Research UK, European Research Council, and European Commission
- Subjects
DNA Copy Number Variations ,Class I Phosphatidylinositol 3-Kinases ,Genomics ,Biology ,Polymorphism, Single Nucleotide ,PBRM1 ,Evolution, Molecular ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,0302 clinical medicine ,Renal cell carcinoma ,Genetics ,medicine ,Humans ,Exome ,Carcinoma, Renal Cell ,Phylogeny ,Exome sequencing ,030304 developmental biology ,0303 health sciences ,Tumor Suppressor Proteins ,High-Throughput Nucleotide Sequencing ,Nuclear Proteins ,Histone-Lysine N-Methyltransferase ,medicine.disease ,Kidney Neoplasms ,3. Good health ,DNA-Binding Proteins ,CpG site ,Von Hippel-Lindau Tumor Suppressor Protein ,Tumor progression ,030220 oncology & carcinogenesis ,Mutation ,Disease Progression ,Cancer research ,CpG Islands ,Ubiquitin Thiolesterase ,Clear cell ,Transcription Factors - Abstract
PMCID: PMC4636053.-- et al., Clear cell renal carcinomas (ccRCCs) can display intratumor heterogeneity (ITH). We applied multiregion exome sequencing (M-seq) to resolve the genetic architecture and evolutionary histories of ten ccRCCs. Ultra-deep sequencing identified ITH in all cases. We found that 73-75% of identified ccRCC driver aberrations were subclonal confounding estimates of driver mutation prevalence. ITH increased with the number of biopsies analyzed, without evidence of saturation in most tumors. Chromosome 3p loss and VHL aberrations were the only ubiquitous events. The proportion of C>T transitions at CpG sites increased during tumor progression. M-seq permits the temporal resolution of ccRCC evolution and refines mutational signatures occurring during tumor development., C.S. and M. Gerlinger are supported by grants from Cancer Research UK Biomarkers and Imaging Discovery and Development Committee (BIDD), the Medical Research Council and the Seventh European Union Framework Programme, and C.S. is supported by the Breast Cancer Research Foundation and the Rosetrees Trust. We acknowledge the Ramón y Cajal program of the Ministerio de Economía y Competitividad, Spain, and Novartis for funding support for E-PREDICT clinical trials. This study was supported by researchers at the National Institute for Health Research Biomedical Research Centres at University College London Hospitals and at the Royal Marsden Hospital.
- Published
- 2014
41. Incidence of cerebral metastases in patients treated with trastuzumab for metastatic breast cancer
- Author
-
Sarah Danson, A. Clayton, Anthony Howell, B. Magee, S Jolly, W.D.J. Ryder, Peter M Wilkinson, Paul A Burt, R S Welch, Andrew M Wardley, Aengus Stewart, and G. Wilson
- Subjects
Adult ,Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,Receptor, ErbB-2 ,Mammary gland ,Antineoplastic Agents ,Breast Neoplasms ,Antibodies, Monoclonal, Humanized ,Metastasis ,Central Nervous System Neoplasms ,Clinical ,breast cancer ,Breast cancer ,Risk Factors ,Trastuzumab ,brain metastases ,Internal medicine ,Humans ,Medicine ,skin and connective tissue diseases ,Aged ,Retrospective Studies ,Univariate analysis ,business.industry ,Incidence ,Incidence (epidemiology) ,Antibodies, Monoclonal ,Retrospective cohort study ,Middle Aged ,medicine.disease ,Metastatic breast cancer ,medicine.anatomical_structure ,Receptors, Estrogen ,Female ,Receptors, Progesterone ,business ,medicine.drug - Abstract
Trastuzumab is an effective treatment for patients with metastatic breast cancer (MBC) that overexpresses HER-2. A high incidence of brain metastases (BM) has been noted in patients receiving trastuzumab. A retrospective chart review was conducted of 100 patients commencing trastuzumab for metastatic breast cancer from July 1999 to December 2002, at the Christie Hospital. Seven patients were excluded; five patients developed central nervous system metastases prior to starting trastuzumab, and inadequate data were available for two. Out of the remaining 93 patients, 23 (25%) have developed BM to date. In all, 46 patients have died, and of these 18 (39%) have been diagnosed with BM prior to death. Of the 23 patients developing BM, 18 (78%) were hormone receptor negative and 18 (78%) had visceral disease. Univariate analysis showed a significant association between the development of cerebral disease and both hormone receptor status and the presence of visceral disease. In conclusion, a high proportion of patients with MBC treated with trastuzumab develop symptomatic cerebral metastases. HER-2-positive breast cancer may have a predilection for the brain, or trastuzumab therapy may change the disease pattern by prolonging survival. New strategies to address this problem require investigation in this group of patients.
- Published
- 2004
42. Abstract 983: Intratumor heterogeneity in non-small cell lung cancer inferred by multi-region exome sequencing
- Author
-
Charles Swanton, Elza C de Bruin, Nik Matthews, Aengus Stewart, Warren Tom, Mariam Jamal-Hanjani, Stuart Horswell, Richard Mitter, Max Salm, Marco Gerlinger, Peter J. Campbell, Andrew Rowan, Nirupa Murugaesu, Timothy T. Harkins, Lucy R. Yates, Clarence Lee, Chaitali Parikh, Nicholas McGranahan, Ignacio Varela, David C. Wedge, and Seema Shafi
- Subjects
Genetics ,Cancer Research ,Phylogenetic tree ,Cancer ,Biology ,medicine.disease ,Somatic evolution in cancer ,DNA sequencing ,Genetic architecture ,Oncology ,medicine ,Adenocarcinoma ,Lung cancer ,Exome sequencing - Abstract
Lung cancer is the most common cancer worldwide, and a leading cause of cancer-related death. Despite improvements in molecular diagnosis and targeted therapies, the 5-years overall survival remains poor. To obtain a better insight into the genetic architecture of the most common type of lung cancer, non-small lung cancer (NSCLC), we performed multi-region DNA sequencing on 7 resected NSCLC samples. Ultra-deep sequencing of a comprehensive cancer gene panel and whole-exome sequencing revealed intratumor heterogeneity in all samples, with several putative tumor driver mutations present in some but not all regions of a tumor. Phylogenetic tree analyses based on non-synonymous mutations revealed a branched evolution pattern in all tumors. An adenosquamous tumor showed striking intratumor heterogeneity, with only a third of all non-synonymous mutations shared across all tumor regions, and clear separation of adenocarcinoma and squamous cell carcinoma tumor regions in the remaining two-third of the mutations. Our multi-region exome sequencing data also revealed regional differences in DNA copy number alterations. Some tumors, relatively homogeneous in terms of mutations, displayed high levels of intratumor heterogeneity in terms of DNA copy number changes, indicating the presence of distinct patterns of intratumor heterogeneity that might contribute to disease progression in different tumours. Overall, our multi-region deep exome sequencing data revealed intratumor heterogeneity in NSCLC, demonstrating branched evolution, both in terms of non-synoymous mutations and DNA copy number alterations, which has important implications for our understanding of the clonal evolution of NSCLC. Citation Format: Elza De Bruin, Nicholas McGranahan, Lucy Yates, Mariam Jamal-Hanjani, Max Salm, Richard Mitter, Seema Shafi, Nirupa Murugaesu, Andrew Rowan, Marco Gerlinger, David Wedge, Stuart Horswell, Ignacio Varela, Warren Tom, Chaitali Parikh, Timothy Harkins, Clarence Lee, Nik Matthews, Aengus Stewart, Peter Campbell, Charles Swanton. Intratumor heterogeneity in non-small cell lung cancer inferred by multi-region exome sequencing. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 983. doi:10.1158/1538-7445.AM2014-983
- Published
- 2014
- Full Text
- View/download PDF
43. Genome-wide co-localization of Polycomb orthologs and their effects on gene expression in human fibroblasts
- Author
-
Richard D. Palermo, Hollie Chandler, Marc Rodriguez-Niedenführ, Emma Anderton, Julie K. Stock, Sharon Brookes, Nik Matthews, Helen Pemberton, Aengus Stewart, Harshil Patel, Lucas de Breed, Tomas Racek, and Gordon Peters
- Subjects
Polycomb Repressive Complex 1 ,Genetics ,Regulation of gene expression ,Genome, Human ,Research ,Gene Expression Regulation, Developmental ,Polycomb-Group Proteins ,macromolecular substances ,Fibroblasts ,Biology ,Chromatin ,Polycomb-group proteins ,Humans ,Gene silencing ,Cell Lineage ,Human genome ,Gene ,Cell aging ,Chromatin immunoprecipitation ,Cellular Senescence ,Protein Binding - Abstract
Background: Polycomb group proteins form multicomponent complexes that are important for establishing lineage-specific patterns of gene expression. Mammalian cells encode multiple permutations of the prototypic Polycomb repressive complex 1 (PRC1) with little evidence for functional specialization. An aim of this study is to determine whether the multiple orthologs that are co-expressed in human fibroblasts act on different target genes and whether their genomic location changes during cellular senescence. Results: Deep sequencing of chromatin immunoprecipitated with antibodies against CBX6, CBX7, CBX8, RING1 and RING2 reveals that the orthologs co-localize at multiple sites. PCR-based validation at representative loci suggests that a further six PRC1 proteins have similar binding patterns. Importantly, sequential chromatin immunoprecipitation with antibodies against different orthologs implies that multiple variants of PRC1 associate with the same DNA. At many loci, the binding profiles have a distinctive architecture that is preserved in two different types of fibroblast. Conversely, there are several hundred loci at which PRC1 binding is cell type-specific and, contrary to expectations, the presence of PRC1 does not necessarily equate with transcriptional silencing. Interestingly, the PRC1 binding profiles are preserved in senescent cells despite changes in gene expression. Conclusions: The multiple permutations of PRC1 in human fibroblasts congregate at common rather than specific sites in the genome and with overlapping but distinctive binding profiles in different fibroblasts. The data imply that the effects of PRC1 complexes on gene expression are more subtle than simply repressing the loci at which they bind.
- Published
- 2014
- Full Text
- View/download PDF
44. Fugu: the small genome of a puffed-up fish
- Author
-
Aengus Stewart
- Subjects
Genetics ,biology ,Evolutionary biology ,Fugu ,biology.animal ,fungi ,Vertebrate ,%22">Fish ,Fugu rubripes ,Genome ,Human genetics - Abstract
The puffer fish Fugu rubripes is perhaps better known as a Japanese delicacy, but its gene-rich, relatively small genome makes the puffer fish a suitable vertebrate for comparative mapping.
- Published
- 2000
- Full Text
- View/download PDF
45. Xenopus: from tadpole to model organism
- Author
-
Aengus Stewart
- Subjects
animal structures ,biology ,urogenital system ,ved/biology ,Evolutionary biology ,embryonic structures ,ved/biology.organism_classification_rank.species ,Xenopus ,Embryo ,Model organism ,biology.organism_classification ,Tadpole ,Developmental biology - Abstract
Developmental biology, primarily work with the embryos of Xenopus species X. laevis and X. tropicalis, is the focus of this site.
- Published
- 2000
- Full Text
- View/download PDF
46. Abstract 4603: Intratumor heterogeneity in clear cell renal cell carcinoma (ccRCC): Multi-region sequencing redefines the mutational landscape of ccRCCs
- Author
-
James Larkin, Claudio R. Santos, Nicholas Matthews, Aengus Stewart, Zoltan Szallasi, Andrew Rowan, Marco Gerlinger, Charles Swanton, Ignacio Varela, Stuart Horswell, Pierre Martinez, Rosalie Fisher, Max Salm, and P. Andrew Futreal
- Subjects
Genetics ,Cancer Research ,Genetic heterogeneity ,Chromosome ,Biology ,Gene mutation ,medicine.disease ,PBRM1 ,Clear cell renal cell carcinoma ,Oncology ,Chromosome instability ,medicine ,Gene ,Exome sequencing - Abstract
Background: Current efforts to define the genomic landscapes of solid tumours are based on the analysis of single tumour biopsies. We recently demonstrated through multi-region exome sequencing (M-seq) of 2 ccRCC that these cancers are genetically heterogeneous (Gerlinger et al. NEJM, 2012). Thus, single biopsy approaches are likely to underestimate the genetic complexity and the incidence of driver gene mutations. Methods: We applied M-seq and copy number profiling to an average of 7 regions from each of 7 advanced ccRCCs to reveal the genomic landscapes and to reconstructed the life histories of these tumours through phylogenetic analysis. Results: M-seq identified approximately 100 non-synonymous somatic mutations per case on average; twice as many mutations than revealed on average in a single biopsy or the 54 mutation found on average in single biopsies of metastatic ccRCCs analysed by The Cancer Genome Atlas. M-seq revealed that all tumours were heterogeneous with more than 50% of non-synonymous somatic mutations and copy number aberrations not present across all analysed tumour regions. The mutational spectrum and chromosomal instability scores differed between regions of individual tumours. Phylogenetic reconstruction showed branched evolution in all cases. Loss of chromosome 3p and mutations in the VHL and PBRM1 genes were the only ccRCC drivers which were altered ubiquitously throughout all regions in all tumours where they occurred. These were the only known ccRCC driver mutations which were recurrently located on the trunk of the phylogenetic trees, indicating that these truncal genetic events are sufficient for tumour initiation. All other known ccRCC driver mutations and the vast majority of chromosomal driver aberrations were confined to spatially separated subclones, represented as branches on the phylogenetic trees. Thus, genetic alterations of VHL, PBRM1 and chromosome 3p appear to initiate the generation of genetic diversity and the acquisition of additional driver events promoting expansion of spatially separated subclones. The truncal driver mutations were similar across all tumours but clinical outcomes differed significantly between patients, indicating that subclonal drivers may determine clinical outcomes. Conclusions: M-seq revealed intra-tumour heterogeneity and parallel evolution of multiple subclones in each of 7 metastatic tumors, indicating that these are common characteristic of ccRCCs. VHL, PBRM1 and chromosome 3p were the only recurrent truncal driver events identified. All other driver mutations were heterogeneous and these are likely to influence clinical outcome. Thus, M-seq reveals complex heterogeneous genomic landscapes in ccRCCs and demonstrates that the detection of subclonal driver mutations may be highly relevant for personalized cancer medicine approaches. Citation Format: Marco Gerlinger, Stuart Horswell, James Larkin, Andrew J. Rowan, Pierre Martinez, Ignacio Varela, Claudio R. Santos, Rosalie Fisher, Max P. Salm, Zoltan Szallasi, Nicholas Matthews, Aengus Stewart, P Andrew Futreal, Charles Swanton. Intratumor heterogeneity in clear cell renal cell carcinoma (ccRCC): Multi-region sequencing redefines the mutational landscape of ccRCCs. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 4603. doi:10.1158/1538-7445.AM2013-4603
- Published
- 2013
- Full Text
- View/download PDF
47. Abstract 964: Intra-tumor heterogeneity and Darwinian selection revealed by multi-region exome sequencing of renal cell carcinomas
- Author
-
Aengus Stewart, Aron Charles Eklund, Graham Clark, Lisa Pickering, Nicholas Matthews, Keiran Raine, Gordon Stamp, Martin Gore, Patrick S. Tarpey, Stuart Horswell, David Endesfelder, Andrew Rowan, Bradley Spencer-Dene, Marco Gerlinger, Calli Latimer, Eva Grönroos, James Larkin, Adam Butler, Charles Swanton, Zoltan Szallasi, Neil Q. McDonald, P. Andrew Futreal, Claudio R. Santos, Sharmin Begum, Julian Downward, Ignacio Varela, David T. Jones, Pierre Martinez, Benjamin Phillimore, and Mahrokh Nohadani
- Subjects
Genetics ,Cancer Research ,Tumor suppressor gene ,Genetic heterogeneity ,Biology ,medicine.disease ,Primary tumor ,Metastasis ,Oncology ,Tumor progression ,medicine ,biology.protein ,PTEN ,Kinase activity ,Exome sequencing - Abstract
Background: Genetic intra-tumor heterogeneity (ITH) may foster tumor adaptation by providing selectable phenotypes for Darwinian evolution. Extensive genetic heterogeneity may also hinder personalized medicine strategies that rely on the portrayal of the mutational landscape from single tumor biopsies. Methods: To examine ITH, we have subjected multiple biopsies from primary renal cell carcinomas and associated metastatic sites to exome capture sequencing (n=2), SNP-array based chromosomal aberration and ploidy profiling analysis (n=4). Phylogenetic relationships of tumor regions were reconstructed by clonal ordering of non-synonymous somatic mutations. The phenotypic consequences of genetic ITH were characterised by immunohistochemistry, mutation functional analysis and mRNA expression profiling. Results: Phylogenetic reconstruction revealed branched evolutionary tumor growth with 63-69% of somatic mutations identified from single biopsies not detectable across all sequenced tumor regions. Independent tumor suppressor gene loss of function mutations with distinct regional distributions were detected within individual tumors: SETD2 harbored 5 different mutations in 2 tumors and PTEN and KDM5C two different mutations in one tumor, each. Thus, despite genetic divergence during tumor progression, phenotypic convergent evolution occurred, indicating a high degree of early mutational diversity. ITH was observed for a mutation in the kinase domain of mTOR, correlating with S6 and 4EBP phosphorylation specifically in cancer regions carrying the mutation and constitutive activation of mTOR kinase activity in vitro. Expression of a renal cancer-specific prognostic signature differed between tumor regions. Chromosomal aberration analysis revealed extensive ITH with 26/30 tumor biopsies from four tumors harboring divergent allelic imbalance profiles. Ploidy profiling revealed heterogeneity in two out of four tumors and identified an aneuploid tumor cell population in a metastasis that probably evolved from a tetraploid intermediate detectable in the primary tumor. Conclusions: Genetic ITH was present in all tumors and occurs through spatially separated heterogeneous somatic mutations and chromosomal and ploidy aberrations leading to both, phenotypic intra-tumor diversity (mTOR activating mutation) and convergent loss of function(SETD2, PTEN and KDM5C). ITH can lead to underestimation of the tumor genomic landscape portrayed from single biopsies and may present significant challenges to personalized medicine and biomarker development. ITH, associated with heterogeneous protein function, may foster tumor adaptation and therapeutic failure through Darwinian selection. Targeting mutations commonly located on the trunks, rather than the branches of phylogenetic trees, may improve therapeutic outcomes. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 964. doi:1538-7445.AM2012-964
- Published
- 2012
- Full Text
- View/download PDF
48. Analysis of a single institution series of 155 patients treated with adjuvant trastuzumab
- Author
-
G. Wilson, H. Mitchell, Paul A Burt, Carole Farrell, Andrew M Wardley, A. McGurk, V. Lau, Aengus Stewart, J. Loncaster, and B. Magee
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Series (stratigraphy) ,business.industry ,medicine.medical_treatment ,Surgery ,Clinical Practice ,Trastuzumab ,Interim ,Internal medicine ,medicine ,Single institution ,business ,Adjuvant ,medicine.drug - Abstract
11504 Background: Following interim results of the HERA study (2005) trastuzumab (T) was introduced as standard clinical practice three weekly for 18 cycles postchemotherapy (CT). Cardiac dysfuncti...
- Published
- 2008
- Full Text
- View/download PDF
49. Analysis of cardiac events in a single institution series of 155 patients who completed adjuvant Trastuzumab
- Author
-
Paul A Burt, Carole Farrell, Aengus Stewart, H. Lander, H. Mitchell, Andrew M Wardley, A. McGurk, B. Magee, V. Lau, and J. Loncaster
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Series (stratigraphy) ,business.industry ,medicine.medical_treatment ,Trastuzumab ,Internal medicine ,Medicine ,Single institution ,business ,Adjuvant ,medicine.drug - Published
- 2008
- Full Text
- View/download PDF
50. High incidence of cerebral metastases in patients with metastatic breast cancer treated with trastuzumab
- Author
-
Paul A Burt, Peter M Wilkinson, Sarah Danson, Andrew M Wardley, G. Wilson, A. Clayton, B. Magee, D. Ryder, S Jolly, and Aengus Stewart
- Subjects
Oncology ,CA15-3 ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.disease ,Metastatic breast cancer ,Breast cancer ,Trastuzumab ,Internal medicine ,medicine ,In patient ,High incidence ,business ,medicine.drug - Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.