Network


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

Hotspot


Dive into the research topics where Kaur Alasoo is active.

Publication


Featured researches published by Kaur Alasoo.


Nature | 2017

Common genetic variation drives molecular heterogeneity in human iPSCs

Helena Kilpinen; Angela Goncalves; Andreas Leha; Vackar Afzal; Kaur Alasoo; Sofie Ashford; Sendu Bala; Dalila Bensaddek; Francesco Paolo Casale; Oliver J. Culley; Petr Danecek; Adam Faulconbridge; Peter W. Harrison; Annie Kathuria; Davis J. McCarthy; Shane McCarthy; Ruta Meleckyte; Yasin Memari; Nathalie Moens; Filipa Soares; Alice L. Mann; Ian Streeter; Chukwuma A. Agu; Alex Alderton; Rachel Nelson; Sarah Harper; Minal Patel; Alistair White; Sharad R Patel; Laura Clarke

Technology utilizing human induced pluripotent stem cells (iPS cells) has enormous potential to provide improved cellular models of human disease. However, variable genetic and phenotypic characterization of many existing iPS cell lines limits their potential use for research and therapy. Here we describe the systematic generation, genotyping and phenotyping of 711 iPS cell lines derived from 301 healthy individuals by the Human Induced Pluripotent Stem Cells Initiative. Our study outlines the major sources of genetic and phenotypic variation in iPS cells and establishes their suitability as models of complex human traits and cancer. Through genome-wide profiling we find that 5–46% of the variation in different iPS cell phenotypes, including differentiation capacity and cellular morphology, arises from differences between individuals. Additionally, we assess the phenotypic consequences of genomic copy-number alterations that are repeatedly observed in iPS cells. In addition, we present a comprehensive map of common regulatory variants affecting the transcriptome of human pluripotent cells.


Scientific Reports | 2015

Transcriptional profiling of macrophages derived from monocytes and iPS cells identifies a conserved response to LPS and novel alternative transcription.

Kaur Alasoo; Fernando O. Martinez; Christine Hale; Siamon Gordon; Fiona Powrie; Gordon Dougan; Subhankar Mukhopadhyay; Daniel J. Gaffney

Macrophages differentiated from human induced pluripotent stem cells (IPSDMs) are a potentially valuable new tool for linking genotype to phenotype in functional studies. However, at a genome-wide level these cells have remained largely uncharacterised. Here, we compared the transcriptomes of naïve and lipopolysaccharide (LPS) stimulated monocyte-derived macrophages (MDMs) and IPSDMs using RNA-Seq. The IPSDM and MDM transcriptomes were broadly similar and exhibited a highly conserved response to LPS. However, there were also significant differences in the expression of genes associated with antigen presentation and tissue remodelling. Furthermore, genes coding for multiple chemokines involved in neutrophil recruitment were more highly expressed in IPSDMs upon LPS stimulation. Additionally, analysing individual transcript expression identified hundreds of genes undergoing alternative promoter and 3′ untranslated region usage following LPS treatment representing a previously under-appreciated level of regulation in the LPS response.


Nature Genetics | 2018

Molecular and functional variation in iPSC-derived sensory neurons

Jeremy Schwartzentruber; Stefanie Foskolou; Helena Kilpinen; Julia Rodrigues; Kaur Alasoo; Andrew J. Knights; Minal Patel; Angela Goncalves; Rita Ferreira; Caroline L. Benn; Anna Wilbrey; Magda Bictash; Emma Impey; Lishuang Cao; Sergio Lainez; Alexandre J C Loucif; Paul Whiting; Alex Gutteridge; Daniel J. Gaffney

Induced pluripotent stem cells (iPSCs), and cells derived from them, have become key tools for modeling biological processes, particularly in cell types that are difficult to obtain from living donors. Here we present a map of regulatory variants in iPSC-derived neurons, based on 123 differentiations of iPSCs to a sensory neuronal fate. Gene expression was more variable across cultures than in primary dorsal root ganglion, particularly for genes related to nervous system development. Using single-cell RNA-sequencing, we found that the number of neuronal versus contaminating cells was influenced by iPSC culture conditions before differentiation. Despite high differentiation-induced variability, our allele-specific method detected thousands of quantitative trait loci (QTLs) that influenced gene expression, chromatin accessibility, and RNA splicing. On the basis of these detected QTLs, we estimate that recall-by-genotype studies that use iPSC-derived cells will require cells from at least 20–80 individuals to detect the effects of regulatory variants with moderately large effect sizes.This study identifies regulatory variants in sensory neurons derived from induced pluripotent stem cells. Despite differentiation-induced variability, an allele-specific method allowed detection of loci influencing gene expression, chromatin accessibility and RNA splicing.


PLOS ONE | 2015

Induced pluripotent stem cell derived macrophages as a cellular system to study salmonella and other pathogens.

Christine Hale; Amy T. Y. Yeung; David Goulding; Derek Pickard; Kaur Alasoo; Fiona Powrie; Gordon Dougan; Subhankar Mukhopadhyay

A number of pathogens, including several human-restricted organisms, persist and replicate within macrophages (Mφs) as a key step in pathogenesis. The mechanisms underpinning such host-restricted intracellular adaptations are poorly understood, in part, due to a lack of appropriate model systems. Here we explore the potential of human induced pluripotent stem cell derived macrophages (iPSDMs) to study such pathogen interactions. We show iPSDMs express a panel of established Mφ-specific markers, produce cytokines, and polarise into classical and alternative activation states in response to IFN-γ and IL-4 stimulation, respectively. iPSDMs also efficiently phagocytosed inactivated bacterial particles as well as live Salmonella Typhi and S. Typhimurium and were able to kill these pathogens. We conclude that iPSDMs can support productive Salmonella infection and propose this as a flexible system to study host/pathogen interactions. Furthermore, iPSDMs can provide a flexible and practical cellular platform for assessing host responses in multiple genetic backgrounds.


bioRxiv | 2017

Shared genetic effects on chromatin and gene expression reveal widespread enhancer priming in immune response

Kaur Alasoo; Julia Rodrigues; Subhankar Mukhopadhyay; Andrew J. Knights; Alice L. Mann; Kousik Kundu; Christine Hale; Gordon Dougan; Daniel J. Gaffney

Noncoding regulatory variants play an important role in the genetics of complex traits. Although quantitative trait locus (QTL) mapping is a powerful approach to identify these variants, many genetic effects may remain unobserved when cells are sampled in only one of a large number of possible environments. Using a novel induced pluripotent stem cell-derived system, we mapped QTLs regulating chromatin accessibility and gene expression in macrophages in four conditions mimicking the interplay between interferon-gamma response and Salmonella infection. We found that approximately 50% of condition-specific effects on gene expression altered chromatin accessibility prior to stimulation. Furthermore, 6% of the chromatin accessibility QTLs regulated multiple neighbouring regions and these interactions were modulated by stimulation, occasionally producing condition-specific changes in gene expression. Profiling additional states also doubled the number of expression QTLs that could be confidently colocalised with disease associations. Thus, a substantial fraction of disease-associated variants may affect ‘primed’ regulatory elements in naive cells.Noncoding regulatory variants are often highly context-specific, modulating gene expression in a small subset of possible cellular states. Although these genetic effects are likely to play important roles in disease, the molecular mechanisms underlying context-specificity are not well understood. Here, we identify shared quantitative trait loci (QTLs) for chromatin accessibility and gene expression (eQTLs) and show that a large fraction (~60%) of eQTLs that appear following macrophage immune stimulation alter chromatin accessibility in unstimulated cells, suggesting they perturb enhancer priming. We show that such variants are likely to influence the binding of cell type specific transcription factors (TFs), such as PU.1, which then indirectly alter the binding of stimulus-specific TFs, such as NF-κB or STAT2. Our results imply that, although chromatin accessibility assays are powerful for fine mapping causal noncoding variants, detecting their downstream impact on gene expression will be challenging, requiring profiling of large numbers of stimulated cellular states and timepoints.


Emerging Topics in Life Sciences | 2017

Computational biology: deep learning

William Jones; Kaur Alasoo; Dmytro Fishman; Leopold Parts

Deep learning is the trendiest tool in a computational biologists toolbox. This exciting class of methods, based on artificial neural networks, quickly became popular due to its competitive performance in prediction problems. In pioneering early work, applying simple network architectures to abundant data already provided gains over traditional counterparts in functional genomics, image analysis, and medical diagnostics. Now, ideas for constructing and training networks and even off-the-shelf models have been adapted from the rapidly developing machine learning subfield to improve performance in a range of computational biology tasks. Here, we review some of these advances in the last 2 years.


Nature Genetics | 2018

Shared genetic effects on chromatin and gene expression indicate a role for enhancer priming in immune response

Kaur Alasoo; Julia Rodrigues; Subhankar Mukhopadhyay; Andrew J. Knights; Alice L. Mann; Kousik Kundu; Christine Hale; Gordon Dougan; Daniel J. Gaffney

Regulatory variants are often context specific, modulating gene expression in a subset of possible cellular states. Although these genetic effects can play important roles in disease, the molecular mechanisms underlying context specificity are poorly understood. Here, we identified shared quantitative trait loci (QTLs) for chromatin accessibility and gene expression in human macrophages exposed to IFNγ, Salmonella and IFNγ plus Salmonella. We observed that ~60% of stimulus-specific expression QTLs with a detectable effect on chromatin altered the chromatin accessibility in naive cells, thus suggesting that they perturb enhancer priming. Such variants probably influence binding of cell-type-specific transcription factors, such as PU.1, which can then indirectly alter the binding of stimulus-specific transcription factors, such as NF-κB or STAT2. Thus, although chromatin accessibility assays are powerful for fine-mapping causal regulatory variants, detecting their downstream effects on gene expression will be challenging, requiring profiling of large numbers of stimulated cellular states and time points.Analysis of chromatin accessibility and expression quantitative trait loci in stimulated or naïve macrophages identifies loci that constitutively alter chromatin but affect expression only after stimulation, thus indicating an effect on enhancer priming.


Nature | 2017

Corrigendum: Common genetic variation drives molecular heterogeneity in human iPSCs

Helena Kilpinen; Angela Goncalves; Andreas Leha; Vackar Afzal; Kaur Alasoo; Sofie Ashford; Sendu Bala; Dalila Bensaddek; Francesco Paolo Casale; Oliver J. Culley; Petr Danecek; Adam Faulconbridge; Peter W. Harrison; Annie Kathuria; Davis J. McCarthy; Shane McCarthy; Ruta Meleckyte; Yasin Memari; Nathalie Moens; Filipa Soares; Alice L. Mann; Ian Streeter; Chukwuma A. Agu; Alex Alderton; Rachel Nelson; Sarah Harper; Minal Patel; Alistair White; Sharad R Patel; Laura Clarke

This corrects the article DOI: 10.1038/nature22403.


bioRxiv | 2018

Genetic effects on promoter usage are highly context-specific and contribute to complex traits

Kaur Alasoo; Julia Rodrigues; John Danesh; Daniel F. Freitag; Dirk S. Paul; Daniel J. Gaffney

Genetic variants regulating RNA splicing and transcript usage have been implicated in both common and rare diseases. However, identifying specific transcriptional effects of these variants remains challenging, partly because reference transcriptomes are incomplete and contain many truncated transcripts that lack annotated 3’ or 5’ ends. We developed a novel analytical approach to overcome these limitations by stratifying transcript annotations into three separate components: promoters, internal exons and 3’ ends. We apply our method to genotype and RNA-seq data from human macrophages exposed to a range of inflammatory stimuli (IFNγ, Salmonella, IFNγ + Salmonella) and a metabolic stimulus (acetylated LDL), obtained from up to 84 individuals. We found that over half of the quantitative trait loci (QTLs) colocalising with complex traits were identified only at the transcript level with no detectable effect on total gene expression. Furthermore, 55% of the transcript-level associations regulated either promoter or 3’ end usage, many of which are missed by methods that only quantify exon-exon junctions. Finally, we demonstrate that promoter-usage QTLs have distinct genetic architecture and are 50% more likely to be context-specific than alternatively spliced internal exons. In summary, we highlight how different RNA-seq quantification approaches capture distinct aspects of transcription and that characterizing the full spectrum of transcriptional consequences of genetic variation requires a combination of analytical strategies.


Trends in Cell Biology | 2018

AP-1 Takes Centre Stage in Enhancer Chromatin Dynamics

Pedro Madrigal; Kaur Alasoo

Recent studies involving induced pluripotent stem cell (iPSC) reprogramming in mice and monocyte-to-macrophage differentiation in humans have revealed a role for the transcription factor (TF) activator protein 1 (AP-1) in chromatin accessibility. Enhancer selection may be determined not only by cell type-specific sets of TFs but also by broadly expressed ones like AP-1.

Collaboration


Dive into the Kaur Alasoo's collaboration.

Top Co-Authors

Avatar

Alice L. Mann

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar

Daniel J. Gaffney

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar

Helena Kilpinen

Swiss Institute of Bioinformatics

View shared research outputs
Top Co-Authors

Avatar

Angela Goncalves

European Bioinformatics Institute

View shared research outputs
Top Co-Authors

Avatar

Christine Hale

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar

Gordon Dougan

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar

Julia Rodrigues

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Subhankar Mukhopadhyay

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar

Adam Faulconbridge

European Bioinformatics Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge