Network


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

Hotspot


Dive into the research topics where Pascal Timshel is active.

Publication


Featured researches published by Pascal Timshel.


Bioinformatics | 2015

SNPsnap: a Web-based tool for identification and annotation of matched SNPs

Tune H. Pers; Pascal Timshel; Joel N. Hirschhorn

SUMMARY An important computational step following genome-wide association studies (GWAS) is to assess whether disease or trait-associated single-nucleotide polymorphisms (SNPs) enrich for particular biological annotations. SNP-based enrichment analysis needs to account for biases such as co-localization of GWAS signals to gene-dense and high linkage disequilibrium (LD) regions, and correlations of gene size, location and function. The SNPsnap Web server enables SNP-based enrichment analysis by providing matched sets of SNPs that can be used to calibrate background expectations. Specifically, SNPsnap efficiently identifies sets of randomly drawn SNPs that are matched to a set of query SNPs based on allele frequency, number of SNPs in LD, distance to nearest gene and gene density. AVAILABILITY AND IMPLEMENTATION SNPsnap server is available at http://www.broadinstitute.org/mpg/snpsnap/. CONTACT [email protected] SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.


Human Molecular Genetics | 2016

Comprehensive analysis of schizophrenia-associated loci highlights ion channel pathways and biologically plausible candidate causal genes

Tune H. Pers; Pascal Timshel; Stephan Ripke; Samantha Lent; Patrick F. Sullivan; Michael Conlon O'Donovan; Lude Franke; Joel N. Hirschhorn

Over 100 associated genetic loci have been robustly associated with schizophrenia. Gene prioritization and pathway analysis have focused on a priori hypotheses and thus may have been unduly influenced by prior assumptions and missed important causal genes and pathways. Using a data-driven approach, we show that genes in associated loci: (1) are highly expressed in cortical brain areas; (2) are enriched for ion channel pathways (false discovery rates <0.05); and (3) contain 62 genes that are functionally related to each other and hence represent promising candidates for experimental follow up. We validate the relevance of the prioritized genes by showing that they are enriched for rare disruptive variants and de novo variants from schizophrenia sequencing studies (odds ratio 1.67, P = 0.039), and are enriched for genes encoding members of mouse and human postsynaptic density proteomes (odds ratio 4.56, P = 5.00 × 10(-4); odds ratio 2.60, P = 0.049).The authors wish it to be known that, in their opinion, the first 2 authors should be regarded as joint First Author.


Nature Genetics | 2018

Gene discovery and polygenic prediction from a genome-wide association study of educational attainment in 1.1 million individuals.

James J. Lee; Robbee Wedow; Aysu Okbay; Edward Kong; Omeed Maghzian; Meghan Zacher; Tuan Anh Nguyen-Viet; Peter Bowers; Julia Sidorenko; Richard Karlsson Linner; Mark Alan Fontana; Tushar Kundu; Chanwook Lee; Hui Li; Ruoxi Li; Rebecca Royer; Pascal Timshel; Raymond K. Walters; Emily Willoughby; Loic Yengo; Maris Alver; Yanchun Bao; David W. Clark; Felix R. Day; Nicholas A. Furlotte; Peter K. Joshi; Kathryn E. Kemper; Aaron Kleinman; Claudia Langenberg; Reedik Mägi

Here we conducted a large-scale genetic association analysis of educational attainment in a sample of approximately 1.1 million individuals and identify 1,271 independent genome-wide-significant SNPs. For the SNPs taken together, we found evidence of heterogeneous effects across environments. The SNPs implicate genes involved in brain-development processes and neuron-to-neuron communication. In a separate analysis of the X chromosome, we identify 10 independent genome-wide-significant SNPs and estimate a SNP heritability of around 0.3% in both men and women, consistent with partial dosage compensation. A joint (multi-phenotype) analysis of educational attainment and three related cognitive phenotypes generates polygenic scores that explain 11–13% of the variance in educational attainment and 7–10% of the variance in cognitive performance. This prediction accuracy substantially increases the utility of polygenic scores as tools in research.Gene discovery and polygenic predictions from a genome-wide association study of educational attainment in 1.1 million individuals.


Molecular metabolism | 2018

Profiling of G Protein-Coupled Receptors in Vagal Afferents Reveals Novel Gut-to-Brain Sensing Mechanisms

Kristoffer L. Egerod; Natalia Petersen; Pascal Timshel; Jens C. Rekling; Yibing Wang; Qinghua Liu; Thue W. Schwartz; Laurent Gautron

Objectives G protein-coupled receptors (GPCRs) act as transmembrane molecular sensors of neurotransmitters, hormones, nutrients, and metabolites. Because unmyelinated vagal afferents richly innervate the gastrointestinal mucosa, gut-derived molecules may directly modulate the activity of vagal afferents through GPCRs. However, the types of GPCRs expressed in vagal afferents are largely unknown. Here, we determined the expression profile of all GPCRs expressed in vagal afferents of the mouse, with a special emphasis on those innervating the gastrointestinal tract. Methods Using a combination of high-throughput quantitative PCR, RNA sequencing, and in situ hybridization, we systematically quantified GPCRs expressed in vagal unmyelinated Nav1.8-expressing afferents. Results GPCRs for gut hormones that were the most enriched in Nav1.8-expressing vagal unmyelinated afferents included NTSR1, NPY2R, CCK1R, and to a lesser extent, GLP1R, but not GHSR and GIPR. Interestingly, both GLP1R and NPY2R were coexpressed with CCK1R. In contrast, NTSR1 was coexpressed with GPR65, a marker preferentially enriched in intestinal mucosal afferents. Only few microbiome-derived metabolite sensors such as GPR35 and, to a lesser extent, GPR119 and CaSR were identified in the Nav1.8-expressing vagal afferents. GPCRs involved in lipid sensing and inflammation (e.g. CB1R, CYSLTR2, PTGER4), and neurotransmitters signaling (CHRM4, DRD2, CRHR2) were also highly enriched in Nav1.8-expressing neurons. Finally, we identified 21 orphan GPCRs with unknown functions in vagal afferents. Conclusion Overall, this study provides a comprehensive description of GPCR-dependent sensing mechanisms in vagal afferents, including novel coexpression patterns, and conceivably coaction of key receptors for gut-derived molecules involved in gut-brain communication.


bioRxiv | 2018

scVAE: Variational auto-encoders for single-cell gene expression data

Christopher Heje Grønbech; Maximillian Fornitz Vording; Pascal Timshel; Casper Kaae Sønderby; Tune H. Pers; Ole Winther

We propose a novel variational auto-encoder-based method for analysis of single-cell RNA sequencing (scRNA-seq) data. It avoids data preprocessing by using raw count data as input and can robustly estimate the expected gene expression levels and a latent representation for each cell. We show for several scRNA-seq data sets that our method outperforms recently proposed scRNA-seq methods in clustering cells. Our software tool scVAE has support for several count likelihood functions and a variant of the variational auto-encoder has a priori clustering in the latent space.


bioRxiv | 2018

Genome-wide study identifies 611 loci associated with risk tolerance and risky behaviors

Richard Karlsson Linner; Pietro Biroli; Edward Kong; S. Fleur W. Meddens; Robbee Wedow; Mark Alan Fontana; Mael Lebreton; Abdel Abdellaoui; Anke R. Hammerschlag; Michel G. Nivard; Aysu Okbay; Cornelius A. Rietveld; Pascal Timshel; Stephen P Tino; Maciej Trzaskowski; Ronald de Vlaming; Christian L Zünd; Yanchun Bao; Laura Buzdugan; Ann H Caplin; Chia-Yen Chen; Peter Eibich; Pierre Fontanillas; Juan R. González; Peter K. Joshi; Ville Karhunen; Aaron Kleinman; Remy Z Levin; Christina M. Lill; Gerardus A. Meddens

Humans vary substantially in their willingness to take risks. In a combined sample of over one million individuals, we conducted genome-wide association studies (GWAS) of general risk tolerance, adventurousness, and risky behaviors in the driving, drinking, smoking, and sexual domains. We identified 611 approximately independent genetic loci associated with at least one of our phenotypes, including 124 with general risk tolerance. We report evidence of substantial shared genetic influences across general risk tolerance and risky behaviors: 72 of the 124 general risk tolerance loci contain a lead SNP for at least one of our other GWAS, and general risk tolerance is moderately to strongly genetically correlated ( to 0.50) with a range of risky behaviors. Bioinformatics analyses imply that genes near general-risk-tolerance-associated SNPs are highly expressed in brain tissues and point to a role for glutamatergic and GABAergic neurotransmission. We find no evidence of enrichment for genes previously hypothesized to relate to risk tolerance.Humans vary substantially in their willingness to take risks. In a combined sample of over one million individuals, we conducted genome-wide association studies (GWAS) of general risk tolerance, adventurousness, and risky behaviors in the driving, drinking, smoking, and sexual domains. We identified 611 approximately independent genetic loci associated with at least one of our phenotypes, including 124 with general risk tolerance. We report evidence of substantial shared genetic influences across general risk tolerance and risky behaviors: 72 of the 124 general risk tolerance loci contain a lead SNP for at least one of our other GWAS, and general risk tolerance is moderately to strongly genetically correlated (|rˆ g | ~ 0.25 to 0.50) with a range of risky behaviors. Bioinformatics analyses imply that genes near general-risk-tolerance-associated SNPs are highly expressed in brain tissues and point to a role for glutamatergic and GABAergic neurotransmission. We find no evidence of enrichment for genes previously hypothesized to relate to risk tolerance.


Oncotarget | 2018

Analysis of a gene panel for targeted sequencing of colorectal cancer samples

Klaus Højgaard Jensen; Jose M. G. Izarzugaza; Agnieszka Sierakowska Juncker; Rasmus Borup Hansen; Torben Hansen; Pascal Timshel; Thorarinn Blondal; Thomas Skøt Jensen; Eske Rygaard-Hjalsted; Peter Mouritzen; Michael Thorsen; Rasmus Wernersson; Henrik Bjørn Nielsen; Anders Jakobsen; Søren Brunak; Flemming Brandt Sørensen

Colorectal cancer (CRC) is a leading cause of death worldwide. Surgical intervention is a successful treatment for stage I patients, whereas other more advanced cases may require adjuvant chemotherapy. The selection of effective adjuvant treatments remains, however, challenging. Accurate patient stratification is necessary for the identification of the subset of patients likely responding to treatment, while sparing others from pernicious treatment. Targeted sequencing approaches may help in this regard, enabling rapid genetic investigation, and at the same time easily applicable in routine diagnosis. We propose a set of guidelines for the identification, including variant calling and filtering, of somatic mutations driving tumorigenesis in the absence of matched healthy tissue. We also discuss the inclusion criteria for the generation of our gene panel. Furthermore, we evaluate the prognostic impact of individual genes, using Cox regression models in the context of overall survival and disease-free survival. These analyses confirmed the role of commonly used biomarkers, and shed light on controversial genes such as CYP2C8. Applying those guidelines, we created a novel gene panel to investigate the onset and progression of CRC in 273 patients. Our comprehensive biomarker set includes 266 genes that may play a role in the progression through the different stages of the disease. Tracing the developmental state of the tumour, and its resistances, is instrumental in patient stratification and reliable decision making in precision clinical practice.


Endocrinology | 2016

GPR119, a Major Enteroendocrine Sensor of Dietary Triglyceride Metabolites Coacting in Synergy With FFA1 (GPR40).

Jeppe Hvidtfeldt Ekberg; Maria Hauge; Line Vildbrad Kristensen; Andreas N. Madsen; Maja S. Engelstoft; Anna-Sofie Husted; Rasmus Sichlau; Kristoffer L. Egerod; Pascal Timshel; Timothy Kowalski; Fiona M. Gribble; Frank Reiman; Harald S. Hansen; Andrew D. Howard; Birgitte Holst; Thue W. Schwartz


Molecular and Cellular Endocrinology | 2017

Gq and Gs signaling acting in synergy to control GLP-1 secretion

Maria Hauge; Jeppe Pio Ekberg; Maja S. Engelstoft; Pascal Timshel; Andreas N. Madsen; Thue W. Schwartz


Nature Communications | 2018

Re-analysis of public genetic data reveals a rare X-chromosomal variant associated with type 2 diabetes

Sílvia Bonàs-Guarch; Marta Guindo-Martínez; Irene Miguel-Escalada; Niels Grarup; David Sebastián; Elias Rodríguez-Fos; Friman Sánchez; Mercè Planas-Fèlix; Paula Cortes-Sánchez; Santi González; Pascal Timshel; Tune H. Pers; Claire C. Morgan; Ignasi Moran; Goutham Atla; Juan R. González; Montserrat Puiggròs; Jonathan Martí; Ehm A. Andersson; Carlos Díaz; Rosa M. Badia; Miriam S. Udler; Aaron Leong; Varindepal Kaur; Jason Flannick; Torben Jørgensen; Allan Linneberg; Marit E. Jørgensen; Daniel R. Witte; Cramer Christensen

Collaboration


Dive into the Pascal Timshel's collaboration.

Top Co-Authors

Avatar

Tune H. Pers

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cramer Christensen

University of Southern Denmark

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge