Network


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

Hotspot


Dive into the research topics where Julia Steinberg is active.

Publication


Featured researches published by Julia Steinberg.


American Journal of Human Genetics | 2014

Gene Age Predicts the Strength of Purifying Selection Acting on Gene Expression Variation in Humans

Konstantin Popadin; Maria Gutierrez-Arcelus; Tuuli Lappalainen; Alfonso Buil; Julia Steinberg; Sergey Igorievich Nikolaev; Samuel W. Lukowski; Georgii A. Bazykin; Vladimir B. Seplyarskiy; Panagiotis Ioannidis; Evgeny M. Zdobnov; Emmanouil T. Dermitzakis

Gene expression levels can be subject to selection. We hypothesized that the age of gene origin is associated with expression constraints, given that it affects the level of gene integration into the functional cellular environment. By studying the genetic variation affecting gene expression levels (cis expression quantitative trait loci [cis-eQTLs]) and protein levels (cis protein QTLs [cis-pQTLs]), we determined that young, primate-specific genes are enriched in cis-eQTLs and cis-pQTLs. Compared to cis-eQTLs of old genes originating before the zebrafish divergence, cis-eQTLs of young genes have a higher effect size, are located closer to the transcription start site, are more significant, and tend to influence genes in multiple tissues and populations. These results suggest that the expression constraint of each gene increases throughout its lifespan. We also detected a positive correlation between expression constraints (approximated by cis-eQTL properties) and coding constraints (approximated by Ka/Ks) and observed that this correlation might be driven by gene age. To uncover factors associated with the increase in gene-age-related expression constraints, we demonstrated that gene connectivity, gene involvement in complex regulatory networks, gene haploinsufficiency, and the strength of posttranscriptional regulation increase with gene age. We also observed an increase in heritability of gene expression levels with age, implying a reduction of the environmental component. In summary, we show that gene age shapes key gene properties during evolution and is therefore an important component of genome function.


Journal of Orthopaedic Research | 2016

Functional genomics in osteoarthritis: Past, present, and future

Julia Steinberg; Eleftheria Zeggini

Osteoarthritis (OA) is a common complex disease of high public health burden. OA is characterized by the degeneration of affected joints leading to pain and reduced mobility. Over the last few years, several studies have focused on the genomic changes underpinning OA. Here, we provide a comprehensive overview of genome‐wide, non‐hypothesis‐driven functional genomics (methylation, gene, and protein expression) studies of knee and hip OA in humans. Individual studies have generally been limited in sample size and hence power, and have differed in their approaches; nonetheless, some common themes have started to emerge, notably the role played by biological processes related to the extracellular matrix, immune response, the WNT pathway, angiogenesis, and skeletal development. Larger‐scale studies and streamlined, robust methodologies will be needed to further elucidate the biological etiology of OA going forward.


Scientific Reports | 2017

Integrative epigenomics, transcriptomics and proteomics of patient chondrocytes reveal genes and pathways involved in osteoarthritis.

Julia Steinberg; Graham R. S. Ritchie; Theodoros Roumeliotis; Raveen L. Jayasuriya; Matthew J. Clark; Roger A. Brooks; Abbie L.A. Binch; Karan M. Shah; Rachael Coyle; Mercedes Pardo; Christine L. Le Maitre; Y.F. Ramos; Rob G. H. H. Nelissen; Ingrid Meulenbelt; A. W. McCaskie; Jyoti S. Choudhary; J. Mark Wilkinson; Eleftheria Zeggini

Osteoarthritis (OA) is a common disease characterized by cartilage degeneration and joint remodeling. The underlying molecular changes underpinning disease progression are incompletely understood. We investigated genes and pathways that mark OA progression in isolated primary chondrocytes taken from paired intact versus degraded articular cartilage samples across 38 patients undergoing joint replacement surgery (discovery cohort: 12 knee OA, replication cohorts: 17 knee OA, 9 hip OA patients). We combined genome-wide DNA methylation, RNA sequencing, and quantitative proteomics data. We identified 49 genes differentially regulated between intact and degraded cartilage in at least two –omics levels, 16 of which have not previously been implicated in OA progression. Integrated pathway analysis implicated the involvement of extracellular matrix degradation, collagen catabolism and angiogenesis in disease progression. Using independent replication datasets, we showed that the direction of change is consistent for over 90% of differentially expressed genes and differentially methylated CpG probes. AQP1, COL1A1 and CLEC3B were significantly differentially regulated across all three –omics levels, confirming their differential expression in human disease. Through integration of genome-wide methylation, gene and protein expression data in human primary chondrocytes, we identified consistent molecular players in OA progression that replicated across independent datasets and that have translational potential.


Nucleic Acids Research | 2015

Haploinsufficiency predictions without study bias

Julia Steinberg; Frantisek Honti; Stephen Meader; Caleb Webber

Any given human individual carries multiple genetic variants that disrupt protein-coding genes, through structural variation, as well as nucleotide variants and indels. Predicting the phenotypic consequences of a gene disruption remains a significant challenge. Current approaches employ information from a range of biological networks to predict which human genes are haploinsufficient (meaning two copies are required for normal function) or essential (meaning at least one copy is required for viability). Using recently available study gene sets, we show that these approaches are strongly biased towards providing accurate predictions for well-studied genes. By contrast, we derive a haploinsufficiency score from a combination of unbiased large-scale high-throughput datasets, including gene co-expression and genetic variation in over 6000 human exomes. Our approach provides a haploinsufficiency prediction for over twice as many genes currently unassociated with papers listed in Pubmed as three commonly-used approaches, and outperforms these approaches for predicting haploinsufficiency for less-studied genes. We also show that fine-tuning the predictor on a set of well-studied ‘gold standard’ haploinsufficient genes does not improve the prediction for less-studied genes. This new score can readily be used to prioritize gene disruptions resulting from any genetic variant, including copy number variants, indels and single-nucleotide variants.


Nature Genetics | 2018

Genome-wide analyses using UK Biobank data provide insights into the genetic architecture of osteoarthritis.

Eleni Zengini; Konstantinos Hatzikotoulas; Ioanna Tachmazidou; Julia Steinberg; Fernando Pires Hartwig; Lorraine Southam; Sophie Hackinger; C.G. Boer; Unnur Styrkarsdottir; Arthur Gilly; Daniel Suveges; Britt Killian; Thorvaldur Ingvarsson; Helgi Jonsson; George C. Babis; Andrew McCaskie; André G. Uitterlinden; Joyce B. J. van Meurs; Unnur Thorsteinsdottir; Kari Stefansson; George Davey Smith; J.M. Wilkinson; Eleftheria Zeggini

Osteoarthritis is a common complex disease imposing a large public-health burden. Here, we performed a genome-wide association study for osteoarthritis, using data across 16.5 million variants from the UK Biobank resource. After performing replication and meta-analysis in up to 30,727 cases and 297,191 controls, we identified nine new osteoarthritis loci, in all of which the most likely causal variant was noncoding. For three loci, we detected association with biologically relevant radiographic endophenotypes, and in five signals we identified genes that were differentially expressed in degraded compared with intact articular cartilage from patients with osteoarthritis. We established causal effects on osteoarthritis for higher body mass index but not for triglyceride levels or genetic predisposition to type 2 diabetes.Genome-wide association study for osteoarthritis using data from UK Biobank identifies loci for knee- and hip-specific disease. Functional analyses of chondrocytes provide further insight into candidate causal genes.


Human Molecular Genetics | 2017

Evaluation of shared genetic aetiology between osteoarthritis and bone mineral density identifies SMAD3 as a novel osteoarthritis risk locus

Sophie Hackinger; Katerina Trajanoska; Unnur Styrkarsdottir; Eleni Zengini; Julia Steinberg; Graham R. S. Ritchie; Konstantinos Hatzikotoulas; Arthur Gilly; Evangelos Evangelou; John P. Kemp; David Evans; Thorvaldur Ingvarsson; Helgi Jonsson; Unnur Thorsteinsdottir; Kari Stefansson; A. W. McCaskie; Roger A. Brooks; J.M. Wilkinson; Fernando Rivadeneira; Eleftheria Zeggini

Abstract Osteoarthritis (OA) is a common complex disease with high public health burden and no curative therapy. High bone mineral density (BMD) is associated with an increased risk of developing OA, suggesting a shared underlying biology. Here, we performed the first systematic overlap analysis of OA and BMD on a genome wide scale. We used summary statistics from the GEFOS consortium for lumbar spine (n = 31,800) and femoral neck (n = 32,961) BMD, and from the arcOGEN consortium for three OA phenotypes (hip, ncases=3,498; knee, ncases=3,266; hip and/or knee, ncases=7,410; ncontrols=11,009). Performing LD score regression we found a significant genetic correlation between the combined OA phenotype (hip and/or knee) and lumbar spine BMD (rg=0.18, P = 2.23 × 10−2), which may be driven by the presence of spinal osteophytes. We identified 143 variants with evidence for cross-phenotype association which we took forward for replication in independent large-scale OA datasets, and subsequent meta-analysis with arcOGEN for a total sample size of up to 23,425 cases and 236,814 controls. We found robustly replicating evidence for association with OA at rs12901071 (OR 1.08 95% CI 1.05–1.11, Pmeta=3.12 × 10−10), an intronic variant in the SMAD3 gene, which is known to play a role in bone remodeling and cartilage maintenance. We were able to confirm expression of SMAD3 in intact and degraded cartilage of the knee and hip. Our findings provide the first systematic evaluation of pleiotropy between OA and BMD, highlight genes with biological relevance to both traits, and establish a robust new OA genetic risk locus at SMAD3.


Bioinformatics | 2015

GeneNet Toolbox for MATLAB: a flexible platform for the analysis of gene connectivity in biological networks

Avigail Taylor; Julia Steinberg; Tallulah Andrews; Caleb Webber

Summary: We present GeneNet Toolbox for MATLAB (also available as a set of standalone applications for Linux). The toolbox, available as command-line or with a graphical user interface, enables biologists to assess connectivity among a set of genes of interest (‘seed-genes’) within a biological network of their choosing. Two methods are implemented for calculating the significance of connectivity among seed-genes: ‘seed randomization’ and ‘network permutation’. Options include restricting analyses to a specified subnetwork of the primary biological network, and calculating connectivity from the seed-genes to a second set of interesting genes. Pre-analysis tools help the user choose the best connectivity-analysis algorithm for their network. The toolbox also enables visualization of the connections among seed-genes. GeneNet Toolbox functions execute in reasonable time for very large networks (∼10 million edges) on a desktop computer. Availability and implementation: GeneNet Toolbox is open source and freely available from http://avigailtaylor.github.io/gntat14. Supplementary information: Supplementary data are available at Bioinformatics online. Contact: [email protected]


PLOS Genetics | 2015

Gene Networks Underlying Convergent and Pleiotropic Phenotypes in a Large and Systematically-Phenotyped Cohort with Heterogeneous Developmental Disorders

Tallulah Andrews; Stephen Meader; Anneke T. Vulto-van Silfhout; Avigail Taylor; Julia Steinberg; Jayne Y. Hehir-Kwa; Rolph Pfundt; Nicole de Leeuw; Bert B.A. de Vries; Caleb Webber

Readily-accessible and standardised capture of genotypic variation has revolutionised our understanding of the genetic contribution to disease. Unfortunately, the corresponding systematic capture of patient phenotypic variation needed to fully interpret the impact of genetic variation has lagged far behind. Exploiting deep and systematic phenotyping of a cohort of 197 patients presenting with heterogeneous developmental disorders and whose genomes harbour de novo CNVs, we systematically applied a range of commonly-used functional genomics approaches to identify the underlying molecular perturbations and their phenotypic impact. Grouping patients into 408 non-exclusive patient-phenotype groups, we identified a functional association amongst the genes disrupted in 209 (51%) groups. We find evidence for a significant number of molecular interactions amongst the association-contributing genes, including a single highly-interconnected network disrupted in 20% of patients with intellectual disability, and show using microcephaly how these molecular networks can be used as baits to identify additional members whose genes are variant in other patients with the same phenotype. Exploiting the systematic phenotyping of this cohort, we observe phenotypic concordance amongst patients whose variant genes contribute to the same functional association but note that (i) this relationship shows significant variation across the different approaches used to infer a commonly perturbed molecular pathway, and (ii) that the phenotypic similarities detected amongst patients who share the same inferred pathway perturbation result from these patients sharing many distinct phenotypes, rather than sharing a more specific phenotype, inferring that these pathways are best characterized by their pleiotropic effects.


Annals of the Rheumatic Diseases | 2018

A novel variant in GLIS3 is associated with osteoarthritis

Elisabetta Casalone; Ioanna Tachmazidou; Eleni Zengini; Konstantinos Hatzikotoulas; Sophie Hackinger; Daniel Suveges; Julia Steinberg; Nigel W. Rayner; J.M. Wilkinson; Kalliope Panoutsopoulou; Eleftheria Zeggini

Objectives Osteoarthritis (OA) is a complex disease, but its genetic aetiology remains poorly characterised. To identify novel susceptibility loci for OA, we carried out a genome-wide association study (GWAS) in individuals from the largest UK-based OA collections to date. Methods We carried out a discovery GWAS in 5414 OA individuals with knee and/or hip total joint replacement (TJR) and 9939 population-based controls. We followed-up prioritised variants in OA subjects from the interim release of the UK Biobank resource (up to 12 658 cases and 50 898 controls) and our lead finding in operated OA subjects from the full release of UK Biobank (17 894 cases and 89 470 controls). We investigated its functional implications in methylation, gene expression and proteomics data in primary chondrocytes from 12 pairs of intact and degraded cartilage samples from patients undergoing TJR. Results We detect a genome-wide significant association at rs10116772 with TJR (P=3.7×10−8; for allele A: OR (95% CI) 0.97 (0.96 to 0.98)), an intronic variant in GLIS3, which is expressed in cartilage. Variants in strong correlation with rs10116772 have been associated with elevated plasma glucose levels and diabetes. Conclusions We identify a novel susceptibility locus for OA that has been previously implicated in diabetes and glycaemic traits.


bioRxiv | 2017

The genetic architecture of osteoarthritis: insights from UK Biobank

Eleni Zengini; Konstantinos Hatzikotoulas; Ioanna Tachmazidou; Julia Steinberg; Fernando Pires Hartwig; Lorraine Southam; Sophie Hackinger; C.G. Boer; Unnur Styrkarsdottir; Daniel Suveges; Britt Kilian; Arthur Gilly; Thorvaldur Ingvarsson; Helgi Jonsson; George C. Babis; A. W. McCaskie; André G. Uitterlinden; Joyce B. J. van Meurs; Unnur Thorsteinsdottir; Kari Stefansson; George Davey Smith; J.M. Wilkinson; Eleftheria Zeggini

Osteoarthritis is a common complex disease with huge public health burden. Here we perform a genome-wide association study for osteoarthritis using data across 16.5 million variants from the UK Biobank resource. Following replication and meta-analysis in up to 30,727 cases and 297,191 controls, we report 9 new osteoarthritis loci, in all of which the most likely causal variant is non-coding. For three loci, we detect association with biologically-relevant radiographic endophenotypes, and in five signals we identify genes that are differentially expressed in degraded compared to intact articular cartilage from osteoarthritis patients. We establish causal effects for higher body mass index, but not for triglyceride levels or type 2 diabetes liability, on osteoarthritis.

Collaboration


Dive into the Julia Steinberg's collaboration.

Top Co-Authors

Avatar

Eleftheria Zeggini

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lorraine Southam

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar

Graham R. S. Ritchie

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar

Ioanna Tachmazidou

Wellcome Trust Sanger Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge