Network


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

Hotspot


Dive into the research topics where Scott Newman is active.

Publication


Featured researches published by Scott Newman.


Molecular Cell | 2012

Independence of Repressive Histone Marks and Chromatin Compaction during Senescent Heterochromatic Layer Formation

Tamir Chandra; Kristina Kirschner; Jean Yves Thuret; Benjamin D. Pope; Tyrone Ryba; Scott Newman; Kashif Ahmed; Shamith Samarajiwa; Rafik Salama; Thomas Carroll; Rory Stark; Rekin’s Janky; Masako Narita; Lixiang Xue; Agustin Chicas; Sabrina Nũnez; Ralf Janknecht; Yoko Hayashi-Takanaka; Michael D. Wilson; Aileen Marshall; Duncan T. Odom; M. Madan Babu; David P. Bazett-Jones; Simon Tavaré; Paul A.W. Edwards; Scott W. Lowe; Hiroshi Kimura; David M. Gilbert; Masashi Narita

The expansion of repressive epigenetic marks has been implicated in heterochromatin formation during embryonic development, but the general applicability of this mechanism is unclear. Here we show that nuclear rearrangement of repressive histone marks H3K9me3 and H3K27me3 into nonoverlapping structural layers characterizes senescence-associated heterochromatic foci (SAHF) formation in human fibroblasts. However, the global landscape of these repressive marks remains unchanged upon SAHF formation, suggesting that in somatic cells, heterochromatin can be formed through the spatial repositioning of pre-existing repressively marked histones. This model is reinforced by the correlation of presenescent replication timing with both the subsequent layered structure of SAHFs and the global landscape of the repressive marks, allowing us to integrate microscopic and genomic information. Furthermore, modulation of SAHF structure does not affect the occupancy of these repressive marks, nor vice versa. These experiments reveal that high-order heterochromatin formation and epigenetic remodeling of the genome can be discrete events.


Nature | 2011

Aberrant chromosome morphology in human cells defective for Holliday junction resolution

Thomas Wechsler; Scott Newman; Stephen C. West

In somatic cells, Holliday junctions can be formed between sister chromatids during the recombinational repair of DNA breaks or after replication fork demise. A variety of processes act upon Holliday junctions to remove them from DNA, in events that are critical for proper chromosome segregation. In human cells, the BLM protein, inactivated in individuals with Bloom’s syndrome, acts in combination with topoisomerase IIIα, RMI1 and RMI2 (BTR complex) to promote the dissolution of double Holliday junctions. Cells defective for BLM exhibit elevated levels of sister chromatid exchanges (SCEs) and patients with Bloom’s syndrome develop a broad spectrum of early-onset cancers caused by chromosome instability. MUS81–EME1 (refs 4–7), SLX1–SLX4 (refs 8–11) and GEN1 (refs 12, 13) also process Holliday junctions but, in contrast to the BTR complex, do so by endonucleolytic cleavage. Here we deplete these nucleases from Bloom’s syndrome cells to analyse human cells compromised for the known Holliday junction dissolution/resolution pathways. We show that depletion of MUS81 and GEN1, or SLX4 and GEN1, from Bloom’s syndrome cells results in severe chromosome abnormalities, such that sister chromatids remain interlinked in a side-by-side arrangement and the chromosomes are elongated and segmented. Our results indicate that normally replicating human cells require Holliday junction processing activities to prevent sister chromatid entanglements and thereby ensure accurate chromosome condensation. This phenotype was not apparent when both MUS81 and SLX4 were depleted from Bloom’s syndrome cells, suggesting that GEN1 can compensate for their absence. Additionally, we show that depletion of MUS81 or SLX4 reduces the high frequency of SCEs in Bloom’s syndrome cells, indicating that MUS81 and SLX4 promote SCE formation, in events that may ultimately drive the chromosome instabilities that underpin early-onset cancers associated with Bloom’s syndrome.


Science Signaling | 2011

Amplification of the Driving Oncogene, KRAS or BRAF, Underpins Acquired Resistance to MEK1/2 Inhibitors in Colorectal Cancer Cells

Annette S. Little; Kathryn Balmanno; Matthew J. Sale; Scott Newman; Jonathan R. Dry; Mark Hampson; Paul A.W. Edwards; Paul D. Smith; Simon J. Cook

Resistance to cancer therapeutics targeting the second kinase in a three-kinase cascade involves amplification of the upstream kinase, not the inhibited kinase. Driving Resistance The promise of using targeted small-molecule kinase inhibitors in treating cancer has been shadowed by the development of resistance to these drugs. Here, Little et al. used colorectal cancer cell lines with oncogenic mutations in either KRAS or BRAF—both of which lead to increased signaling through the ERK (extracellular signal–regulated kinase) signaling pathway—to investigate the mechanisms whereby cells developed resistance to AZD6244, a MEK1/2 (mitogen-activated or extracellular signal–regulated protein kinase kinases 1 and 2) inhibitor now in clinical trials. Rather than developing mutations in MEK1/2, cancer cells became resistant to MEK1/2 through amplification of the driving oncogene (oncogenic KRAS or BRAF) and a consequent increase in signaling through the ERK pathway. These observations have implications for the use of MEK1/2 inhibitors—and possibly other inhibitors that target downstream pathway components rather than the driving oncogene itself—in combination with other antineoplastic therapies. The acquisition of resistance to protein kinase inhibitors is a growing problem in cancer treatment. We modeled acquired resistance to the MEK1/2 (mitogen-activated or extracellular signal–regulated protein kinase kinases 1 and 2) inhibitor selumetinib (AZD6244) in colorectal cancer cell lines harboring mutations in BRAF (COLO205 and HT29 lines) or KRAS (HCT116 and LoVo lines). AZD6244-resistant derivatives were refractory to AZD6244-induced cell cycle arrest and death and exhibited a marked increase in ERK1/2 (extracellular signal–regulated kinases 1 and 2) pathway signaling and cyclin D1 abundance when assessed in the absence of inhibitor. Genomic sequencing revealed no acquired mutations in MEK1 or MEK2, the primary target of AZD6244. Rather, resistant lines showed a marked up-regulation of their respective driving oncogenes, BRAF600E or KRAS13D, due to intrachromosomal amplification. Inhibition of BRAF reversed resistance to AZD6244 in COLO205 cells, which suggested that combined inhibition of MEK1/2 and BRAF may reduce the likelihood of acquired resistance in tumors with BRAF600E. Knockdown of KRAS reversed AZD6244 resistance in HCT116 cells as well as reduced the activation of ERK1/2 and protein kinase B; however, the combined inhibition of ERK1/2 and phosphatidylinositol 3-kinase signaling had little effect on AZD6244 resistance, suggesting that additional KRAS effector pathways contribute to this process. Microarray analysis identified increased expression of an 18-gene signature previously identified as reflecting MEK1/2 pathway output in resistant cells. Thus, amplification of the driving oncogene (BRAF600E or KRAS13D) can drive acquired resistance to MEK1/2 inhibitors by increasing signaling through the ERK1/2 pathway. However, up-regulation of KRAS13D leads to activation of multiple KRAS effector pathways, underlining the therapeutic challenge posed by KRAS mutations. These results may have implications for the use of combination therapies.


PLOS Medicine | 2015

Spatial and Temporal Heterogeneity in High-Grade Serous Ovarian Cancer: A Phylogenetic Analysis

Roland F. Schwarz; Charlotte K.Y. Ng; Susanna L. Cooke; Scott Newman; Jillian Temple; Anna Piskorz; Davina Gale; Karen Sayal; Muhammed Murtaza; Peter Baldwin; Nitzan Rosenfeld; Helena M. Earl; Evis Sala; Mercedes Jimenez-Linan; Christine Parkinson; Florian Markowetz; James D. Brenton

Background The major clinical challenge in the treatment of high-grade serous ovarian cancer (HGSOC) is the development of progressive resistance to platinum-based chemotherapy. The objective of this study was to determine whether intra-tumour genetic heterogeneity resulting from clonal evolution and the emergence of subclonal tumour populations in HGSOC was associated with the development of resistant disease. Methods and Findings Evolutionary inference and phylogenetic quantification of heterogeneity was performed using the MEDICC algorithm on high-resolution whole genome copy number profiles and selected genome-wide sequencing of 135 spatially and temporally separated samples from 14 patients with HGSOC who received platinum-based chemotherapy. Samples were obtained from the clinical CTCR-OV03/04 studies, and patients were enrolled between 20 July 2007 and 22 October 2009. Median follow-up of the cohort was 31 mo (interquartile range 22–46 mo), censored after 26 October 2013. Outcome measures were overall survival (OS) and progression-free survival (PFS). There were marked differences in the degree of clonal expansion (CE) between patients (median 0.74, interquartile range 0.66–1.15), and dichotimization by median CE showed worse survival in CE-high cases (PFS 12.7 versus 10.1 mo, p = 0.009; OS 42.6 versus 23.5 mo, p = 0.003). Bootstrap analysis with resampling showed that the 95% confidence intervals for the hazard ratios for PFS and OS in the CE-high group were greater than 1.0. These data support a relationship between heterogeneity and survival but do not precisely determine its effect size. Relapsed tissue was available for two patients in the CE-high group, and phylogenetic analysis showed that the prevalent clonal population at clinical recurrence arose from early divergence events. A subclonal population marked by a NF1 deletion showed a progressive increase in tumour allele fraction during chemotherapy. Conclusions This study demonstrates that quantitative measures of intra-tumour heterogeneity may have predictive value for survival after chemotherapy treatment in HGSOC. Subclonal tumour populations are present in pre-treatment biopsies in HGSOC and can undergo expansion during chemotherapy, causing clinical relapse.


Genome Research | 2012

Estimation of rearrangement phylogeny for cancer genomes

Christopher Greenman; Erin Pleasance; Scott Newman; Fengtang Yang; Beiyuan Fu; Serena Nik-Zainal; David Jones; King Wai Lau; Nigel P. Carter; Paul A.W. Edwards; P. Andrew Futreal; Michael R. Stratton; Peter J. Campbell

Cancer genomes are complex, carrying thousands of somatic mutations including base substitutions, insertions and deletions, rearrangements, and copy number changes that have been acquired over decades. Recently, technologies have been introduced that allow generation of high-resolution, comprehensive catalogs of somatic alterations in cancer genomes. However, analyses of these data sets generally do not indicate the order in which mutations have occurred, or the resulting karyotype. Here, we introduce a mathematical framework that begins to address this problem. By using samples with accurate data sets, we can reconstruct relatively complex temporal sequences of rearrangements and provide an assembly of genomic segments into digital karyotypes. For cancer genes mutated in rearranged regions, this information can provide a chronological examination of the selective events that have taken place.


The Journal of Pathology | 2012

The role of tandem duplicator phenotype in tumour evolution in high-grade serous ovarian cancer

Charlotte K.Y. Ng; Susanna L. Cooke; Kevin L. Howe; Scott Newman; Jian Xian; Jillian Temple; Elizabeth M. Batty; Jessica C.M. Pole; Simon P. Langdon; Paul A.W. Edwards; James D. Brenton

High‐grade serous ovarian carcinoma (HGSOC) is characterized by genomic instability, ubiquitous TP53 loss, and frequent development of platinum resistance. Loss of homologous recombination (HR) is a mutator phenotype present in 50% of HGSOCs and confers hypersensitivity to platinum treatment. We asked which other mutator phenotypes are present in HGSOC and how they drive the emergence of platinum resistance. We performed whole‐genome paired‐end sequencing on a model of two HGSOC cases, each consisting of a pair of cell lines established before and after clinical resistance emerged, to describe their structural variants (SVs) and to infer their ancestral genomes as the SVs present within each pair. The first case (PEO1/PEO4), with HR deficiency, acquired translocations and small deletions through its early evolution, but a revertant BRCA2 mutation restoring HR function in the resistant lineage re‐stabilized its genome and reduced platinum sensitivity. The second case (PEO14/PEO23) had 216 tandem duplications and did not show evidence of HR or mismatch repair deficiency. By comparing the cell lines to the tissues from which they originated, we showed that the tandem duplicator mutator phenotype arose early in progression in vivo and persisted throughout evolution in vivo and in vitro, which may have enabled continual evolution. From the analysis of SNP array data from 454 HGSOC cases in The Cancer Genome Atlas series, we estimate that 12.8% of cases show patterns of aberrations similar to the tandem duplicator, and this phenotype is mutually exclusive with BRCA1/2 carrier mutations. Copyright


Genome Research | 2011

Large duplications at reciprocal translocation breakpoints that might be the counterpart of large deletions and could arise from stalled replication bubbles

Karen Howarth; Jessica C.M. Pole; Juliet C. Beavis; Elizabeth M. Batty; Scott Newman; Graham R. Bignell; Paul A.W. Edwards

Reciprocal chromosome translocations are often not exactly reciprocal. Most familiar are deletions at the breakpoints, up to megabases in extent. We describe here the opposite phenomenon-duplication of tens or hundreds of kilobases at the breakpoint junction, so that the same sequence is present on both products of a translocation. When the products of the translocation are mapped on the genome, they overlap. We report several of these overlapping-breakpoint duplications in breast cancer cell lines HCC1187, HCC1806, and DU4475. These lines also had deletions and essentially balanced translocations. In HCC1187 and HCC1806, we identified five cases of duplication ranging between 46 kb and 200 kb, with the partner chromosome showing deletions between 29 bp and 31 Mb. DU4475 had a duplication of at least 200 kb. Breakpoints were mapped using array painting, i.e., hybridization of chromosomes isolated by flow cytometry to custom oligonucleotide microarrays. Duplications were verified by fluorescent in situ hybridization (FISH), PCR on isolated chromosomes, and cloning of breakpoints. We propose that these duplications are the counterpart of deletions and that they are produced at a replication bubble, comprising two replication forks with the duplicated sequence in between. Both copies of the duplicated sequence would go to one daughter cell, on different products of the translocation, while the other daughter cell would show deletion. These duplications may have been overlooked because they may be missed by FISH and array-CGH and may be interpreted as insertions by paired-end sequencing. Such duplications may therefore be quite frequent.


PLOS ONE | 2013

The Relative Timing of Mutations in a Breast Cancer Genome

Scott Newman; Karen Howarth; Christopher Greenman; Graham R. Bignell; Simon Tavaré; Paul A.W. Edwards

Many tumors have highly rearranged genomes, but a major unknown is the relative importance and timing of genome rearrangements compared to sequence-level mutation. Chromosome instability might arise early, be a late event contributing little to cancer development, or happen as a single catastrophic event. Another unknown is which of the point mutations and rearrangements are selected. To address these questions we show, using the breast cancer cell line HCC1187 as a model, that we can reconstruct the likely history of a breast cancer genome. We assembled probably the most complete map to date of a cancer genome, by combining molecular cytogenetic analysis with sequence data. In particular, we assigned most sequence-level mutations to individual chromosomes by sequencing of flow sorted chromosomes. The parent of origin of each chromosome was assigned from SNP arrays. We were then able to classify most of the mutations as earlier or later according to whether they occurred before or after a landmark event in the evolution of the genome, endoreduplication (duplication of its entire genome). Genome rearrangements and sequence-level mutations were fairly evenly divided earlier and later, suggesting that genetic instability was relatively constant throughout the life of this tumor, and chromosome instability was not a late event. Mutations that caused chromosome instability would be in the earlier set. Strikingly, the great majority of inactivating mutations and in-frame gene fusions happened earlier. The non-random timing of some of the mutations may be evidence that they were selected.


Genome Medicine | 2010

High-throughput analysis of chromosome translocations and other genome rearrangements in epithelial cancers

Scott Newman; Paul Anthony Edwards

Genes that are broken or fused by structural changes to the genome are an important class of mutation in the leukemias and sarcomas but have been largely overlooked in the common epithelial cancers. Large-scale sequencing is changing our perceptions of the cancer genome, and it is now being applied to structural changes, using the paired end strategy. This reveals more clearly than before the extent to which many cancer genomes are rearranged and how much these rearrangements contribute to the mutational burden of epithelial tumors. In particular, there are probably many fusion genes, analogous to those found in leukemias, to be found in common cancers, such as breast carcinoma, and some of these will prove to be important in cancer diagnosis and treatment.


Cancer Research | 2010

Abstract 1189: Chromosomal aberrations reveal novel genes with prognostic significance in esophageal adenocarcinoma

Xin Yi C. Goh; Jonathan Re Rees; Suet-Feung Chin; Scott Newman; Maria O'Donovan; Bauke Yistra; Carlos Caldas; Paul A.W. Edwards; Rebecca C. Fitzgerald

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DCnnINTRODUCTION: The incidence of esophageal adenocarcinoma (EAC) has increased 6-fold in the west over the last 30 years, with a dismal survival rate of about 10%. Understanding the genomic aberrations and molecular pathogenesis in this cancer may enhance disease interventions.nnAIMS: To identify novel molecular targets with prognostic significance and to better characterize EAC by analyzing chromosomal aberrations across the whole genome.nnMETHODS: An in-house oligonucleotide array comparative genomic hybridization (CGH) was performed on 89 chemo-naive EAC resection samples with long-term clinical follow-up data to identify regions of copy number gains/losses: good quality data obtained for 56 samples. Illumina Human Cyto Single-nucleotide Polymorphism (SNP)-12 Beadarray was carried out to map genomic boundaries of regions with aberrations. Touch-imprinting fluorescence in situ hybridization (FISH) assays were carried out using bacterial artificial chromosome probes on interphase nuclei of frozen samples to confirm findings. Regions with potential homozygous deletions (HDs) were investigated by multiplex nested PCR using microdissected DNA of frozen samples. The degree and pattern of genomic gains were studied using different FISH probes.nnRESULTS: 19 minimal regions with gains and 12 regions with losses (cut off=2 standard deviations (SD) above/below mean log2 ratios) were identified. Potential HDs (cut off=3 SD) were identified in 4 samples (7.1%) at 9p21. Nested multiplex PCR and FISH (quantitative: 100 nuclei per sample counted) confirmed a p16 deletion in all 4 samples. SNP analysis suggested another region of potential HD that involves a novel gene, ELAVL2. Amplifications (cut off=3 SD) in 6 samples (10.7%) at 8p23.1 were confirmed by analyses from the SNP array, which narrowed down the width of amplicon to 2 regions involving: (a) SOX7/PINX1 (5/6 samples) and (b) GATA4, NEIL2, FDFT1 and CTSB (4/6 samples). FISH assays confirmed amplifications of GATA4, NEIL2, FDFT1, CTSB, SOX7 (6 or more copies/centromere) and gains of PINX1 (3-6 copies/centromere). Unsupervised K means clustering (50 iterations) of the array CGH data generated 5 groups and survival analyses identified a group (32.1% of cohort) with significantly worse prognosis (median survival=500 days, p=0.0149). Modified T test with adjusted Bonferroni correction identified 17 clones with different log2 ratios (p<0.05), implicating 4 regions of gains and including 6 novel genes, between this group and the other 4 combined. The only clinical difference between this group and the others was an increased male:female ratio (Fishers exact test; p=0.01).nnCONCLUSIONS: 9p deletions and 8p amplifications are present in 7-11% of samples. The use of array CGH has identified copy number gains with prognostic significance in EAC. Further work is required to determine whether the genes involved have a causative role.nnNote: This abstract was not presented at the AACR 101st Annual Meeting 2010 because the presenter was unable to attend.nnCitation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 1189.

Collaboration


Dive into the Scott Newman's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Graham R. Bignell

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Susanna L. Cooke

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge