Network


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

Hotspot


Dive into the research topics where Oliver M. Dovey is active.

Publication


Featured researches published by Oliver M. Dovey.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Histone deacetylase 1 (HDAC1), but not HDAC2, controls embryonic stem cell differentiation

Oliver M. Dovey; Charles T. Foster; Shaun M. Cowley

Histone deacetylases (HDAC) 1 and 2 are highly similar enzymes that help regulate chromatin structure as the core catalytic components of corepressor complexes. Although tissue-specific deletion of HDAC1 and HDAC2 has demonstrated functional redundancy, germ-line deletion of HDAC1 in the mouse causes early embryonic lethality, whereas HDAC2 does not. To address the unique requirement for HDAC1 in early embryogenesis we have generated conditional knockout embryonic stem (ES) cells in which HDAC1 or HDAC2 genes can be inactivated. Deletion of HDAC1, but not HDAC2, causes a significant reduction in the HDAC activity of Sin3A, NuRD, and CoREST corepressor complexes. This reduced corepressor activity results in a specific 1.6-fold increase in histone H3 K56 acetylation (H3K56Ac), thus providing genetic evidence that H3K56Ac is a substrate of HDAC1. In culture, ES cell proliferation was unaffected by loss of either HDAC1 or HDAC2. Rather, we find that loss of HDAC1 affects ES cell differentiation. ES cells lacking either HDAC1 or HDAC2 were capable of forming embryoid bodies (EBs), which stimulates differentiation into the three primary germ layers. However, HDAC1-deficient EBs were significantly smaller, showed spontaneous rhythmic contraction, and increased expression of both cardiomyocyte and neuronal markers. In summary, our genetic study of HDAC1 and HDAC2 in ES cells, which mimic the embryonic epiblast, has identified a unique requirement for HDAC1 in the optimal activity of HDAC1/2 corepressor complexes and cell fate determination during differentiation.


Molecular and Cellular Biology | 2010

Lysine-Specific Demethylase 1 Regulates the Embryonic Transcriptome and CoREST Stability

Charles T. Foster; Oliver M. Dovey; Larissa Lezina; Jin Li Luo; Timothy W. Gant; Nick Barlev; Allan Bradley; Shaun M. Cowley

ABSTRACT Lysine-specific demethylase 1 (LSD1), which demethylates mono- and dimethylated histone H3-Lys4 as part of a complex including CoREST and histone deacetylases (HDACs), is essential for embryonic development in the mouse beyond embryonic day 6.5 (e6.5). To determine the role of LSD1 during this early period of embryogenesis, we have generated loss-of-function gene trap mice and conditional knockout embryonic stem (ES) cells. Analysis of postimplantation gene trap embryos revealed that LSD1 expression, and therefore function, is restricted to the epiblast. Conditional deletion of LSD1 in mouse ES cells, the in vitro counterpart of the epiblast, revealed a reduction in CoREST protein and associated HDAC activity, resulting in a global increase in histone H3-Lys56 acetylation, but not H3-Lys4 methylation. Despite this biochemical perturbation, ES cells with LSD1 deleted proliferate normally and retain stem cell characteristics. Loss of LSD1 causes the aberrant expression of 588 genes, including those coding for transcription factors with roles in anterior/posterior patterning and limb development, such as brachyury, Hoxb7, Hoxd8, and retinoic acid receptor γ (RARγ). The gene coding for brachyury, a key regulator of mesodermal differentiation, is a direct target gene of LSD1 and is overexpressed in e6.5 Lsd1 gene trap embryos. Thus, LSD1 regulates the expression and appropriate timing of key developmental regulators, as part of the LSD1/CoREST/HDAC complex, during early embryonic development.


The Journal of Pathology | 2006

Array CGH profiling of favourable histology Wilms tumours reveals novel gains and losses associated with relapse

Rachael Natrajan; Richard D. Williams; Sandra Hing; Alan Mackay; Jorge S. Reis-Filho; Kerry Fenwick; Marjan Iravani; Haukur Valgeirsson; Anita Grigoriadis; Cordelia Langford; Oliver M. Dovey; Simon G. Gregory; Barbara L. Weber; Alan Ashworth; Paul E. Grundy; Kathy Pritchard-Jones; Chris Jones

Despite the excellent survival of Wilms tumour patients treated with multimodality therapy, approximately 15% will suffer from tumour relapse, where response rates are markedly reduced. We have carried out microarray‐based comparative genomic hybridisation on a series of 76 Wilms tumour samples, enriched for cases which recurred, to identify changes in DNA copy number associated with clinical outcome. Using 1Mb‐spaced genome‐wide BAC arrays, the most significantly different genomic changes between favourable histology tumours that did (n = 37), and did not (n = 39), subsequently relapse were gains on 1q, and novel deletions at 12q24 and 18q21. Further relapse‐associated loci included losses at 1q32.1, 2q36.3‐2q37.1, and gain at 13q31. 1q gains correlated strongly with loss of 1p and/or 16q. In 3 of 11 cases with concurrent 1p−/1q+, a breakpoint was identified at 1p13. Multiple low‐level sub‐megabase gains along the length of 1q were identified using chromosome 1 tiling‐path arrays. One such recurrent region at 1q22‐q23.1 included candidate genes RAB25, NES, CRABP2, HDGF and NTRK1, which were screened for mRNA expression using quantitative RT‐PCR. These data provide a high‐resolution catalogue of genomic copy number changes in relapsing favourable histology Wilms tumours. Copyright


Cell Reports | 2016

A CRISPR Dropout Screen Identifies Genetic Vulnerabilities and Therapeutic Targets in Acute Myeloid Leukemia

Konstantinos Tzelepis; Hiroko Koike-Yusa; Etienne De Braekeleer; Yilong Li; Emmanouil Metzakopian; Oliver M. Dovey; Annalisa Mupo; Vera Grinkevich; Meng Li; Milena Mazan; Malgorzata Gozdecka; Shuhei Ohnishi; Jonathan L. Cooper; Miten Patel; Thomas McKerrell; Bin Chen; Ana Filipa Domingues; Paolo Gallipoli; Sarah A. Teichmann; Hannes Ponstingl; Ultan McDermott; Julio Saez-Rodriguez; Brian J. P. Huntly; Francesco Iorio; Cristina Pina; George S. Vassiliou; Kosuke Yusa

Summary Acute myeloid leukemia (AML) is an aggressive cancer with a poor prognosis, for which mainstream treatments have not changed for decades. To identify additional therapeutic targets in AML, we optimize a genome-wide clustered regularly interspaced short palindromic repeats (CRISPR) screening platform and use it to identify genetic vulnerabilities in AML cells. We identify 492 AML-specific cell-essential genes, including several established therapeutic targets such as DOT1L, BCL2, and MEN1, and many other genes including clinically actionable candidates. We validate selected genes using genetic and pharmacological inhibition, and chose KAT2A as a candidate for downstream study. KAT2A inhibition demonstrated anti-AML activity by inducing myeloid differentiation and apoptosis, and suppressed the growth of primary human AMLs of diverse genotypes while sparing normal hemopoietic stem-progenitor cells. Our results propose that KAT2A inhibition should be investigated as a therapeutic strategy in AML and provide a large number of genetic vulnerabilities of this leukemia that can be pursued in downstream studies.


Blood | 2013

Histone deacetylase 1 and 2 are essential for normal T-cell development and genomic stability in mice

Oliver M. Dovey; Charles T. Foster; Nathalie Conte; Sally A. Edwards; Jennifer M. Edwards; Rajinder Singh; George S. Vassiliou; Allan Bradley; Shaun M. Cowley

Histone deacetylase 1 and 2 (HDAC1/2) regulate chromatin structure as the catalytic core of the Sin3A, NuRD and CoREST co-repressor complexes. To better understand the key pathways regulated by HDAC1/2 in the adaptive immune system and inform their exploitation as drug targets, we have generated mice with a T-cell specific deletion. Loss of either HDAC1 or HDAC2 alone has little effect, while dual inactivation results in a 5-fold reduction in thymocyte cellularity, accompanied by developmental arrest at the double-negative to double-positive transition. Transcriptome analysis revealed 892 misregulated genes in Hdac1/2 knock-out thymocytes, including down-regulation of LAT, Themis and Itk, key components of the T-cell receptor (TCR) signaling pathway. Down-regulation of these genes suggests a model in which HDAC1/2 deficiency results in defective propagation of TCR signaling, thus blocking development. Furthermore, mice with reduced HDAC1/2 activity (Hdac1 deleted and a single Hdac2 allele) develop a lethal pathology by 3-months of age, caused by neoplastic transformation of immature T cells in the thymus. Tumor cells become aneuploid, express increased levels of c-Myc and show elevated levels of the DNA damage marker, γH2AX. These data demonstrate a crucial role for HDAC1/2 in T-cell development and the maintenance of genomic stability.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Histone deacetylase (HDAC) 1 and 2 are essential for accurate cell division and the pluripotency of embryonic stem cells

Shereen Jamaladdin; Richard D.W. Kelly; Laura O’Regan; Oliver M. Dovey; Grace E. Hodson; Christopher J. Millard; Nicola Portolano; Andrew M. Fry; John W. R. Schwabe; Shaun M. Cowley

Significance Histone deacetylase 1 and 2 (HDAC1/2) are sister proteins that regulate access to DNA by modulating chromatin. We have generated the first double knockout (DKO) of Hdac1/2 in embryonic stem (ES) cells and find that gene inactivation causes a loss of cell viability, which is associated with increased abnormal mitotic spindles and chromosome segregation defects. Transcriptome analysis revealed that almost 2,000 genes are deregulated in DKO cells. Significantly for the self-renewal properties of ES cells, this includes down-regulation of the core pluripotent factors, Oct4, Nanog, and Rex1. Furthermore, using the rescue of Hdac1/2-null cells as a model system to monitor HDAC1/2 activity, we have also shown that mutations that abolish inositol tetraphosphate binding reduce the activity of HDAC1 in vivo. Histone deacetylases 1 and 2 (HDAC1/2) form the core catalytic components of corepressor complexes that modulate gene expression. In most cell types, deletion of both Hdac1 and Hdac2 is required to generate a discernible phenotype, suggesting their activity is largely redundant. We have therefore generated an ES cell line in which Hdac1 and Hdac2 can be inactivated simultaneously. Loss of HDAC1/2 resulted in a 60% reduction in total HDAC activity and a loss of cell viability. Cell death is dependent upon cell cycle progression, because differentiated, nonproliferating cells retain their viability. Furthermore, we observe increased mitotic defects, chromatin bridges, and micronuclei, suggesting HDAC1/2 are necessary for accurate chromosome segregation. Consistent with a critical role in the regulation of gene expression, microarray analysis of Hdac1/2-deleted cells reveals 1,708 differentially expressed genes. Significantly for the maintenance of stem cell self-renewal, we detected a reduction in the expression of the pluripotent transcription factors, Oct4, Nanog, Esrrb, and Rex1. HDAC1/2 activity is regulated through binding of an inositol tetraphosphate molecule (IP4) sandwiched between the HDAC and its cognate corepressor. This raises the important question of whether IP4 regulates the activity of the complex in cells. By rescuing the viability of double-knockout cells, we demonstrate for the first time (to our knowledge) that mutations that abolish IP4 binding reduce the activity of HDAC1/2 in vivo. Our data indicate that HDAC1/2 have essential and pleiotropic roles in cellular proliferation and regulate stem cell self-renewal by maintaining expression of key pluripotent transcription factors.


Leukemia | 2013

A powerful molecular synergy between mutant Nucleophosmin and Flt3-ITD drives acute myeloid leukemia in mice

Annalisa Mupo; L Celani; Oliver M. Dovey; Jonathan L. Cooper; C Grove; Roland Rad; Paolo Sportoletti; Brunangelo Falini; Allan Bradley; George S. Vassiliou

Acute myeloid leukemia (AML) is the commonest myeloid malignancy, yet there has been little therapeutic progress for this disease in decades, and only 25–30% of patients survive long term.1 This reflects its pathogenetic complexity and the fact that the molecular basis of its largest cytogenetic subgroup, AML with a normal karyotype (AML-NK), was obscure until recently. Recent advances in DNA sequencing have revealed that AML-NK is molecularly heterogeneous with >30 genes recurrently targeted by somatic mutations in this disease.2 What is also evident is that each individual case of AML-NK appears to harbor only a small number of coding driver mutations, often as few as three and usually no more than five.2, 3 Furthermore, it is manifest that the precise combination of driver mutations in the genome of each AML impacts on its salient features, including responsiveness to treatments and prognosis.3


Oncogene | 2008

Loss of Rassf1a cooperates with Apc(Min) to accelerate intestinal tumourigenesis

L van der Weyden; M. J. Arends; Oliver M. Dovey; H L Harrison; Gregory C. Lefebvre; Nathalie Conte; Fanni Gergely; Allan Bradley; David J. Adams

Promoter methylation of the RAS-association domain family 1, isoform A gene (RASSF1A) is one of the most frequent events found in human tumours. In this study we set out to test the hypothesis that loss of Rassf1a can cooperate with inactivation of the adenomatous polyposis coli (Apc) gene to accelerate intestinal tumourigenesis using the Apc-Min (ApcMin/+) mouse model, as mutational or deletional inactivation of APC is a frequent early event in the genesis of intestinal cancer. Further, loss of RASSF1A has also been reported to occur in premalignant adenomas of the bowel. RASSF1A has been implicated in an array of pivotal cellular processes, including regulation of the cell cycle, apoptosis, microtubule stability and most recently in the β-catenin signalling pathway. By interbreeding isoform specific Rassf1a knockout mice with Apc+/Min mice, we showed that loss of Rassf1a results in a significant increase in adenomas of the small intestine and accelerated intestinal tumourigenesis leading to the earlier death of adenocarcinoma-bearing mice and decreased overall survival. Comparative genomic hybridization of adenomas from Rassf1a−/−; Apc+/Min mice revealed no evidence of aneuploidy or gross chromosomal instability (no difference to adenomas from Rassf1a+/+; Apc+/Min mice). Immunohistochemical analysis of adenomas revealed increased nuclear β-catenin accumulation in adenomas from Rassf1a−/−; Apc+/Min mice, compared to those from Rassf1a+/+; Apc+/Min mice, but no differences in proliferation marker (Ki67) staining patterns. Collectively these data demonstrate cooperation between inactivation of Rassf1a and Apc resulting in accelerated intestinal tumourigenesis, with adenomas showing increased nuclear accumulation of β-catenin, supporting a mechanistic link via loss of the known interaction of Rassf1 with β-TrCP that usually mediates degradation of β-catenin.


Cell Cycle | 2010

Emphasizing the positive: A role for histone deacetylases in transcriptional activation

Oliver M. Dovey; Charles T. Foster; Shaun M. Cowley

Comment on: Wang Z, et al. Cell 2009; 138:1019-1031.


Clinical Epigenetics | 2013

Histone deacetylase (HDAC) 1 and 2 are essential for normal T cell development and genomic stability in mice

Oliver M. Dovey; George Vasilliou; Allan Bradley; Shaun M. Cowley

The highly related enzymes, histone deacetylase 1 and 2 (HDAC1/2), regulate chromatin structure as the catalytic core of the Sin3A, NuRD and CoREST co-repressor complexes. To better understand their role in the adaptive immune system and inform their exploitation as drug targets, we have generated mice with a T-cell specific inactivation of both Hdac1/2 genes. Loss of either HDAC1 or HDAC2 alone has little effect, while dual inactivation results in a 5-fold reduction in thymocyte cellularity, accompanied by developmental arrest at the double-negative to double-positive transition. Mice with reduced HDAC1/2 activity (Hdac1 deleted and a single Hdac2 allele) develop a lethal pathology by 3-months of age, caused by neoplastic transformation of immature T cells in the thymus. Tumor cells become aneuploid, express increased levels of c-Myc and show elevated levels of the DNA damage marker, γH2AX. Intriguingly, recent data has shown that these same hypomorphic tumour cells show increased levels of cell death in response to treatment with the HDAC inhibitor, SAHA. Therefore, although a partial reduction in HDAC1/2 activity (>60% of WT) could potentially encourage tumour growth, it may also be an Achilles heel for the treatment of cancer cells with standard HDAC inhibitor therapy.

Collaboration


Dive into the Oliver M. Dovey's collaboration.

Top Co-Authors

Avatar

George S. Vassiliou

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar

Allan Bradley

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar

Jonathan L. Cooper

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar

Cordelia Langford

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Annalisa Mupo

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar

Konstantinos Tzelepis

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emmanouil Metzakopian

Wellcome Trust Sanger Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge