Dirk S. Paul
University of Cambridge
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Dirk S. Paul.
Nature Genetics | 2012
Cornelis A. Albers; Dirk S. Paul; Harald Schulze; Kathleen Freson; Jonathan Stephens; Peter A. Smethurst; Jennifer Jolley; Ana Cvejic; Myrto Kostadima; Paul Bertone; Martijn H. Breuning; Najet Debili; Panos Deloukas; Rémi Favier; Janine Fiedler; Catherine M. Hobbs; Ni Huang; Graham Kiddle; Ingrid P. C. Krapels; Paquita Nurden; Claudia Ruivenkamp; Jennifer Sambrook; Kenneth Smith; Derek L. Stemple; Gabriele Strauss; Chantal Thys; Christel Van Geet; Ruth Newbury-Ecob; Willem H. Ouwehand; Cedric Ghevaert
The exon-junction complex (EJC) performs essential RNA processing tasks. Here, we describe the first human disorder, thrombocytopenia with absent radii (TAR), caused by deficiency in one of the four EJC subunits. Compound inheritance of a rare null allele and one of two low-frequency SNPs in the regulatory regions of RBM8A, encoding the Y14 subunit of EJC, causes TAR. We found that this inheritance mechanism explained 53 of 55 cases (P < 5 × 10−228) of the rare congenital malformation syndrome. Of the 53 cases with this inheritance pattern, 51 carried a submicroscopic deletion of 1q21.1 that has previously been associated with TAR, and two carried a truncation or frameshift null mutation in RBM8A. We show that the two regulatory SNPs result in diminished RBM8A transcription in vitro and that Y14 expression is reduced in platelets from individuals with TAR. Our data implicate Y14 insufficiency and, presumably, an EJC defect as the cause of TAR syndrome.
Cell | 2016
William Astle; Heather Elding; Tao Jiang; Dave Allen; Dace Ruklisa; Alice L. Mann; Daniel Mead; Heleen Bouman; Fernando Riveros-Mckay; Myrto Kostadima; John J. Lambourne; Suthesh Sivapalaratnam; Kate Downes; Kousik Kundu; Lorenzo Bomba; Kim Berentsen; John R. Bradley; Louise C. Daugherty; Olivier Delaneau; Kathleen Freson; Stephen F. Garner; Luigi Grassi; Jose A. Guerrero; Matthias Haimel; Eva M. Janssen-Megens; Anita M. Kaan; Mihir Anant Kamat; Bowon Kim; Amit Mandoli; Jonathan Marchini
Summary Many common variants have been associated with hematological traits, but identification of causal genes and pathways has proven challenging. We performed a genome-wide association analysis in the UK Biobank and INTERVAL studies, testing 29.5 million genetic variants for association with 36 red cell, white cell, and platelet properties in 173,480 European-ancestry participants. This effort yielded hundreds of low frequency (<5%) and rare (<1%) variants with a strong impact on blood cell phenotypes. Our data highlight general properties of the allelic architecture of complex traits, including the proportion of the heritable component of each blood trait explained by the polygenic signal across different genome regulatory domains. Finally, through Mendelian randomization, we provide evidence of shared genetic pathways linking blood cell indices with complex pathologies, including autoimmune diseases, schizophrenia, and coronary heart disease and evidence suggesting previously reported population associations between blood cell indices and cardiovascular disease may be non-causal.
Cell | 2016
Lu Chen; Bing Ge; Francesco Paolo Casale; Louella Vasquez; Tony Kwan; Diego Garrido-Martín; Stephen Watt; Ying Yan; Kousik Kundu; Simone Ecker; Avik Datta; David C. Richardson; Frances Burden; Daniel Mead; Alice L. Mann; José María Fernández; Sophia Rowlston; Steven P. Wilder; Samantha Farrow; Xiaojian Shao; John J. Lambourne; Adriana Redensek; Cornelis A. Albers; Vyacheslav Amstislavskiy; Sofie Ashford; Kim Berentsen; Lorenzo Bomba; Guillaume Bourque; David Bujold; Stephan Busche
Summary Characterizing the multifaceted contribution of genetic and epigenetic factors to disease phenotypes is a major challenge in human genetics and medicine. We carried out high-resolution genetic, epigenetic, and transcriptomic profiling in three major human immune cell types (CD14+ monocytes, CD16+ neutrophils, and naive CD4+ T cells) from up to 197 individuals. We assess, quantitatively, the relative contribution of cis-genetic and epigenetic factors to transcription and evaluate their impact as potential sources of confounding in epigenome-wide association studies. Further, we characterize highly coordinated genetic effects on gene expression, methylation, and histone variation through quantitative trait locus (QTL) mapping and allele-specific (AS) analyses. Finally, we demonstrate colocalization of molecular trait QTLs at 345 unique immune disease loci. This expansive, high-resolution atlas of multi-omics changes yields insights into cell-type-specific correlation between diverse genomic inputs, more generalizable correlations between these inputs, and defines molecular events that may underpin complex disease risk.
Bioinformatics | 2016
James R. Staley; James Blackshaw; Mihir Anant Kamat; Steve Ellis; Praveen Surendran; Benjamin Sun; Dirk S. Paul; Daniel F. Freitag; Stephen Burgess; John Danesh; Robin Young; Adam S. Butterworth
Abstract Summary: PhenoScanner is a curated database of publicly available results from large-scale genetic association studies. This tool aims to facilitate ‘phenome scans’, the cross-referencing of genetic variants with many phenotypes, to help aid understanding of disease pathways and biology. The database currently contains over 350 million association results and over 10 million unique genetic variants, mostly single nucleotide polymorphisms. It is accompanied by a web-based tool that queries the database for associations with user-specified variants, providing results according to the same effect and non-effect alleles for each input variant. The tool provides the option of searching for trait associations with proxies of the input variants, calculated using the European samples from 1000 Genomes and Hapmap. Availability and Implementation: PhenoScanner is available at www.phenoscanner.medschl.cam.ac.uk. Contact: [email protected] Supplementary information: Supplementary data are available at Bioinformatics online.
Nature Genetics | 2017
Joanna M. M. Howson; Wei Zhao; Daniel R. Barnes; Weang Kee Ho; Robin Young; Dirk S. Paul; Lindsay L. Waite; Daniel F. Freitag; Eric Fauman; Elias Salfati; Benjamin B. Sun; John D. Eicher; Andrew D. Johnson; Wayne H-H Sheu; Sune F. Nielsen; Wei-Yu Lin; Praveen Surendran; Anders Mälarstig; Jemma B. Wilk; Anne Tybjærg-Hansen; Katrine L. Rasmussen; Pia R. Kamstrup; Panos Deloukas; Jeanette Erdmann; Sekar Kathiresan; Nilesh J. Samani; Heribert Schunkert; Hugh Watkins; CARDIoGRAMplusC D; Ron Do
Coronary artery disease (CAD) is a leading cause of morbidity and mortality worldwide. Although 58 genomic regions have been associated with CAD thus far, most of the heritability is unexplained, indicating that additional susceptibility loci await identification. An efficient discovery strategy may be larger-scale evaluation of promising associations suggested by genome-wide association studies (GWAS). Hence, we genotyped 56,309 participants using a targeted gene array derived from earlier GWAS results and performed meta-analysis of results with 194,427 participants previously genotyped, totaling 88,192 CAD cases and 162,544 controls. We identified 25 new SNP–CAD associations (P < 5 × 10−8, in fixed-effects meta-analysis) from 15 genomic regions, including SNPs in or near genes involved in cellular adhesion, leukocyte migration and atherosclerosis (PECAM1, rs1867624), coagulation and inflammation (PROCR, rs867186 (p.Ser219Gly)) and vascular smooth muscle cell differentiation (LMOD1, rs2820315). Correlation of these regions with cell-type-specific gene expression and plasma protein levels sheds light on potential disease mechanisms.
Trends in Molecular Medicine | 2014
Dirk S. Paul; Stephan Beck
Epigenome-wide association studies (EWASs) provide a systematic approach to uncovering epigenetic variants underlying common diseases. Discoveries have shed light on novel molecular mechanisms of disease and enabled the application of epigenetic variants as biomarkers. Here, we highlight the recent advances in this emerging line of research and discuss key challenges for current and future studies.
Nature Communications | 2016
Dirk S. Paul; Andrew E. Teschendorff; Mary A N Dang; Robert Lowe; Mohammed I. Hawa; Simone Ecker; Huriya Beyan; Stephanie Cunningham; Alexandra R. Fouts; Anita Ramelius; Frances Burden; Samantha Farrow; Sophia Rowlston; Karola Rehnström; Mattia Frontini; Kate Downes; Stephan Busche; Warren Cheung; Bing Ge; Marie Michelle Simon; David Bujold; Tony Kwan; Guillaume Bourque; Avik Datta; Ernesto Lowy; Laura Clarke; Paul Flicek; Emanuele Libertini; Simon Heath; Marta Gut
The incidence of type 1 diabetes (T1D) has substantially increased over the past decade, suggesting a role for non-genetic factors such as epigenetic mechanisms in disease development. Here we present an epigenome-wide association study across 406,365 CpGs in 52 monozygotic twin pairs discordant for T1D in three immune effector cell types. We observe a substantial enrichment of differentially variable CpG positions (DVPs) in T1D twins when compared with their healthy co-twins and when compared with healthy, unrelated individuals. These T1D-associated DVPs are found to be temporally stable and enriched at gene regulatory elements. Integration with cell type-specific gene regulatory circuits highlight pathways involved in immune cell metabolism and the cell cycle, including mTOR signalling. Evidence from cord blood of newborns who progress to overt T1D suggests that the DVPs likely emerge after birth. Our findings, based on 772 methylomes, implicate epigenetic changes that could contribute to disease pathogenesis in T1D.
PLOS Genetics | 2011
Dirk S. Paul; James Nisbet; Tsun-Po Yang; Stuart Meacham; Augusto Rendon; Katta Hautaviita; Jonna Tallila; Jacqui White; Marloes R. Tijssen; Suthesh Sivapalaratnam; Hanneke Basart; Mieke D. Trip; Berthold Göttgens; Nicole Soranzo; Willem H. Ouwehand; Panos Deloukas
Turning genetic discoveries identified in genome-wide association (GWA) studies into biological mechanisms is an important challenge in human genetics. Many GWA signals map outside exons, suggesting that the associated variants may lie within regulatory regions. We applied the formaldehyde-assisted isolation of regulatory elements (FAIRE) method in a megakaryocytic and an erythroblastoid cell line to map active regulatory elements at known loci associated with hematological quantitative traits, coronary artery disease, and myocardial infarction. We showed that the two cell types exhibit distinct patterns of open chromatin and that cell-specific open chromatin can guide the finding of functional variants. We identified an open chromatin region at chromosome 7q22.3 in megakaryocytes but not erythroblasts, which harbors the common non-coding sequence variant rs342293 known to be associated with platelet volume and function. Resequencing of this open chromatin region in 643 individuals provided strong evidence that rs342293 is the only putative causative variant in this region. We demonstrated that the C- and G-alleles differentially bind the transcription factor EVI1 affecting PIK3CG gene expression in platelets and macrophages. A protein–protein interaction network including up- and down-regulated genes in Pik3cg knockout mice indicated that PIK3CG is associated with gene pathways with an established role in platelet membrane biogenesis and thrombus formation. Thus, rs342293 is the functional common variant at this locus; to the best of our knowledge this is the first such variant to be elucidated among the known platelet quantitative trait loci (QTLs). Our data suggested a molecular mechanism by which a non-coding GWA index SNP modulates platelet phenotype.
Genome Research | 2013
Dirk S. Paul; Cornelis A. Albers; Augusto Rendon; Katrin Voss; Jonathan Stephens; Pim van der Harst; John Chambers; Nicole Soranzo; Willem H. Ouwehand; Panos Deloukas
Nearly three-quarters of the 143 genetic signals associated with platelet and erythrocyte phenotypes identified by meta-analyses of genome-wide association (GWA) studies are located at non-protein-coding regions. Here, we assessed the role of candidate regulatory variants associated with cell type-restricted, closely related hematological quantitative traits in biologically relevant hematopoietic cell types. We used formaldehyde-assisted isolation of regulatory elements followed by next-generation sequencing (FAIRE-seq) to map regions of open chromatin in three primary human blood cells of the myeloid lineage. In the precursors of platelets and erythrocytes, as well as in monocytes, we found that open chromatin signatures reflect the corresponding hematopoietic lineages of the studied cell types and associate with the cell type-specific gene expression patterns. Dependent on their signal strength, open chromatin regions showed correlation with promoter and enhancer histone marks, distance to the transcription start site, and ontology classes of nearby genes. Cell type-restricted regions of open chromatin were enriched in sequence variants associated with hematological indices. The majority (63.6%) of such candidate functional variants at platelet quantitative trait loci (QTLs) coincided with binding sites of five transcription factors key in regulating megakaryopoiesis. We experimentally tested 13 candidate regulatory variants at 10 platelet QTLs and found that 10 (76.9%) affected protein binding, suggesting that this is a frequent mechanism by which regulatory variants influence quantitative trait levels. Our findings demonstrate that combining large-scale GWA data with open chromatin profiles of relevant cell types can be a powerful means of dissecting the genetic architecture of closely related quantitative traits.
BioEssays | 2014
Dirk S. Paul; Nicole Soranzo; Stephan Beck
Understanding the functional mechanisms underlying genetic signals associated with complex traits and common diseases, such as cancer, diabetes and Alzheimers disease, is a formidable challenge. Many genetic signals discovered through genome‐wide association studies map to non‐protein coding sequences, where their molecular consequences are difficult to evaluate. This article summarizes concepts for the systematic interpretation of non‐coding genetic signals using genome annotation data sets in different cellular systems. We outline strategies for the global analysis of multiple association intervals and the in‐depth molecular investigation of individual intervals. We highlight experimental techniques to validate candidate (potential causal) regulatory variants, with a focus on novel genome‐editing techniques including CRISPR/Cas9. These approaches are also applicable to low‐frequency and rare variants, which have become increasingly important in genomic studies of complex traits and diseases. There is a pressing need to translate genetic signals into biological mechanisms, leading to prognostic, diagnostic and therapeutic advances.