Network


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

Hotspot


Dive into the research topics where Rinse K. Weersma is active.

Publication


Featured researches published by Rinse K. Weersma.


Nature Genetics | 2010

Genome-wide meta-analysis increases to 71 the number of confirmed Crohn's disease susceptibility loci

Andre Franke; Dermot McGovern; Jeffrey C. Barrett; Kai Wang; Graham L. Radford-Smith; Tariq Ahmad; Charlie W. Lees; Tobias Balschun; James C. Lee; Rebecca L. Roberts; Carl A. Anderson; Joshua C. Bis; Suzanne Bumpstead; David Ellinghaus; Eleonora M. Festen; Michel Georges; Todd Green; Talin Haritunians; Luke Jostins; Anna Latiano; Christopher G. Mathew; Grant W. Montgomery; Natalie J. Prescott; Soumya Raychaudhuri; Jerome I. Rotter; Philip Schumm; Yashoda Sharma; Lisa A. Simms; Kent D. Taylor; David C. Whiteman

We undertook a meta-analysis of six Crohns disease genome-wide association studies (GWAS) comprising 6,333 affected individuals (cases) and 15,056 controls and followed up the top association signals in 15,694 cases, 14,026 controls and 414 parent-offspring trios. We identified 30 new susceptibility loci meeting genome-wide significance (P < 5 × 10−8). A series of in silico analyses highlighted particular genes within these loci and, together with manual curation, implicated functionally interesting candidate genes including SMAD3, ERAP2, IL10, IL2RA, TYK2, FUT2, DNMT3A, DENND1B, BACH2 and TAGAP. Combined with previously confirmed loci, these results identify 71 distinct loci with genome-wide significant evidence for association with Crohns disease.


Nature Genetics | 2015

Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations

Jimmy Z. Liu; Suzanne van Sommeren; Hailiang Huang; Siew C. Ng; Rudi Alberts; Atsushi Takahashi; Stephan Ripke; James C. Lee; Luke Jostins; Tejas Shah; Shifteh Abedian; Jae Hee Cheon; Judy H. Cho; Naser E Daryani; Lude Franke; Yuta Fuyuno; Ailsa Hart; Ramesh C. Juyal; Garima Juyal; Won Ho Kim; Andrew P. Morris; Hossein Poustchi; William G. Newman; Vandana Midha; Timothy R. Orchard; Homayon Vahedi; Ajit Sood; Joseph J.Y. Sung; Reza Malekzadeh; Harm-Jan Westra

Ulcerative colitis and Crohns disease are the two main forms of inflammatory bowel disease (IBD). Here we report the first trans-ancestry association study of IBD, with genome-wide or Immunochip genotype data from an extended cohort of 86,640 European individuals and Immunochip data from 9,846 individuals of East Asian, Indian or Iranian descent. We implicate 38 loci in IBD risk for the first time. For the majority of the IBD risk loci, the direction and magnitude of effect are consistent in European and non-European cohorts. Nevertheless, we observe genetic heterogeneity between divergent populations at several established risk loci driven by differences in allele frequency (NOD2) or effect size (TNFSF15 and ATG16L1) or a combination of these factors (IL23R and IRGM). Our results provide biological insights into the pathogenesis of IBD and demonstrate the usefulness of trans-ancestry association studies for mapping loci associated with complex diseases and understanding genetic architecture across diverse populations.


Nature Genetics | 2008

Sequence variants in IL10, ARPC2 and multiple other loci contribute to ulcerative colitis susceptibility

Andre Franke; Tobias Balschun; Tom H. Karlsen; Jurgita Sventoraityte; Susanna Nikolaus; Gabriele Mayr; Francisco S. Domingues; Mario Albrecht; Michael Nothnagel; David Ellinghaus; Christian Sina; Clive M. Onnie; Rinse K. Weersma; Pieter Stokkers; Cisca Wijmenga; Maria Gazouli; David P. Strachan; Wendy L. McArdle; Severine Vermeire; Paul Rutgeerts; Philip Rosenstiel; Michael Krawczak; Morten H. Vatn; Christopher G. Mathew; Stefan Schreiber

Inflammatory bowel disease (IBD) typically manifests as either ulcerative colitis (UC) or Crohns disease (CD). Systematic identification of susceptibility genes for IBD has thus far focused mainly on CD, and little is known about the genetic architecture of UC. Here we report a genome-wide association study with 440,794 SNPs genotyped in 1,167 individuals with UC and 777 healthy controls. Twenty of the most significantly associated SNPs were tested for replication in three independent European case-control panels comprising a total of 1,855 individuals with UC and 3,091 controls. Among the four consistently replicated markers, SNP rs3024505 immediately flanking the IL10 (interleukin 10) gene on chromosome 1q32.1 showed the most significant association in the combined verification samples (P = 1.35 × 10−12; OR = 1.46 (1.31–1.62)). The other markers were located in ARPC2 and in the HLA-BTNL2 region. Association between rs3024505 and CD (1,848 cases, 1,804 controls) was weak (P = 0.013; OR = 1.17 (1.01–1.34)). IL10 is an immunosuppressive cytokine that has long been proposed to influence IBD pathophysiology. Our findings strongly suggest that defective IL10 function is central to the pathogenesis of the UC subtype of IBD.


Nature Genetics | 2010

Genome-wide association identifies multiple ulcerative colitis susceptibility loci

Dermot McGovern; Agnès Gardet; Leif Törkvist; Philippe Goyette; Jonah Essers; Kent D. Taylor; Benjamin M. Neale; Rick Twee-Hee Ong; Caroline Lagacé; Chun Li; Todd Green; Christine Stevens; Claudine Beauchamp; Phillip Fleshner; Marie Carlson; Mauro D'Amato; Jonas Halfvarson; Martin L. Hibberd; Mikael Lördal; Leonid Padyukov; Angelo Andriulli; E. Colombo; Anna Latiano; Orazio Palmieri; Edmond Jean Bernard; Colette Deslandres; Daan W. Hommes; Dirk J. de Jong; Pieter Stokkers; Rinse K. Weersma

Ulcerative colitis is a chronic, relapsing inflammatory condition of the gastrointestinal tract with a complex genetic and environmental etiology. In an effort to identify genetic variation underlying ulcerative colitis risk, we present two distinct genome-wide association studies of ulcerative colitis and their joint analysis with a previously published scan, comprising, in aggregate, 2,693 individuals with ulcerative colitis and 6,791 control subjects. Fifty-nine SNPs from 14 independent loci attained an association significance of P < 10−5. Seven of these loci exceeded genome-wide significance (P < 5 × 10−8). After testing an independent cohort of 2,009 cases of ulcerative colitis and 1,580 controls, we identified 13 loci that were significantly associated with ulcerative colitis (P < 5 × 10−8), including the immunoglobulin receptor gene FCGR2A, 5p15, 2p16 and ORMDL3 (orosomucoid1-like 3). We confirmed association with 14 previously identified ulcerative colitis susceptibility loci, and an analysis of acknowledged Crohns disease loci showed that roughly half of the known Crohns disease associations are shared with ulcerative colitis. These data implicate approximately 30 loci in ulcerative colitis, thereby providing insight into disease pathogenesis.


Science | 2016

Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity.

Alexandra Zhernakova; Alexander Kurilshikov; Marc Jan Bonder; Ettje F. Tigchelaar; Melanie Schirmer; Tommi Vatanen; Zlatan Mujagic; Arnau Vich Vila; Gwen Falony; Sara Vieira-Silva; Jun Wang; Floris Imhann; Eelke Brandsma; Soesma A. Jankipersadsing; Marie Joossens; Maria Carmen Cenit; Patrick Deelen; Morris A. Swertz; Rinse K. Weersma; Edith J. M. Feskens; Mihai G. Netea; Dirk Gevers; Daisy Jonkers; Lude Franke; Yurii S. Aulchenko; Curtis Huttenhower; Jeroen Raes; Marten H. Hofker; Ramnik J. Xavier; Cisca Wijmenga

“Normal” for the gut microbiota For the benefit of future clinical studies, it is critical to establish what constitutes a “normal” gut microbiome, if it exists at all. Through fecal samples and questionnaires, Falony et al. and Zhernakova et al. targeted general populations in Belgium and the Netherlands, respectively. Gut microbiota composition correlated with a range of factors including diet, use of medication, red blood cell counts, fecal chromogranin A, and stool consistency. The data give some hints for possible biomarkers of normal gut communities. Science, this issue pp. 560 and 565 Two large-scale studies in Western Europe establish environment-diet-microbe-host interactions. Deep sequencing of the gut microbiomes of 1135 participants from a Dutch population-based cohort shows relations between the microbiome and 126 exogenous and intrinsic host factors, including 31 intrinsic factors, 12 diseases, 19 drug groups, 4 smoking categories, and 60 dietary factors. These factors collectively explain 18.7% of the variation seen in the interindividual distance of microbial composition. We could associate 110 factors to 125 species and observed that fecal chromogranin A (CgA), a protein secreted by enteroendocrine cells, was exclusively associated with 61 microbial species whose abundance collectively accounted for 53% of microbial composition. Low CgA concentrations were seen in individuals with a more diverse microbiome. These results are an important step toward a better understanding of environment-diet-microbe-host interactions.


Gastroenterology | 2010

Genome-Wide Association Analysis in Primary Sclerosing Cholangitis

Tom H. Karlsen; Andre Franke; Espen Melum; Arthur Kaser; Johannes R. Hov; Tobias Balschun; Benedicte A. Lie; Annika Bergquist; Christoph Schramm; Tobias J. Weismüller; Daniel Gotthardt; Christian Rust; Eva Philipp; Teresa Fritz; Liesbet Henckaerts; Rinse K. Weersma; Pieter Stokkers; Cyriel Y. Ponsioen; Cisca Wijmenga; Martina Sterneck; Michael Nothnagel; Jochen Hampe; Andreas Teufel; Heiko Runz; Philip Rosenstiel; Adolf Stiehl; Severine Vermeire; Ulrich Beuers; Michael P. Manns; Erik Schrumpf

BACKGROUND & AIMS We aimed to characterize the genetic susceptibility to primary sclerosing cholangitis (PSC) by means of a genome-wide association analysis of single nucleotide polymorphism (SNP) markers. METHODS A total of 443,816 SNPs on the Affymetrix SNP Array 5.0 (Affymetrix, Santa Clara, CA) were genotyped in 285 Norwegian PSC patients and 298 healthy controls. Associations detected in this discovery panel were re-examined in independent case-control panels from Scandinavia (137 PSC cases and 368 controls), Belgium/The Netherlands (229 PSC cases and 735 controls), and Germany (400 cases and 1832 controls). RESULTS The strongest associations were detected near HLA-B at chromosome 6p21 (rs3099844: odds ratio [OR], 4.8; 95% confidence interval [CI], 3.6-6.5; P = 2.6 x 10(-26); and rs2844559: OR, 4.7; 95% CI, 3.5-6.4; P = 4.2 x 10(-26) in the discovery panel). Outside the HLA complex, rs9524260 at chromosome 13q31 showed significant associations in 3 of 4 study panels. Lentiviral silencing of glypican 6, encoded at this locus, led to the up-regulation of proinflammatory markers in a cholangiocyte cell line. Of 15 established ulcerative colitis susceptibility loci, significant replication was obtained at chromosomes 2q35 and 3p21 (rs12612347: OR, 1.26; 95% CI, 1.06-1.50; and rs3197999: OR, 1.22; 95% CI, 1.02-1.47, respectively), with circumstantial evidence supporting the G-protein-coupled bile acid receptor 1 and macrophage-stimulating 1, respectively, as the likely disease genes. CONCLUSIONS Strong HLA associations and a subset of genes involved in bile homeostasis and other inflammatory conditions constitute key components of the genetic architecture of PSC.


PLOS Genetics | 2011

Trans-eQTLs Reveal That Independent Genetic Variants Associated with a Complex Phenotype Converge on Intermediate Genes, with a Major Role for the HLA

Rudolf S. N. Fehrmann; Ritsert C. Jansen; Jan H. Veldink; Harm-Jan Westra; Danny Arends; Marc Jan Bonder; Jingyuan Fu; Patrick Deelen; Harry J.M. Groen; Asia Smolonska; Rinse K. Weersma; Robert M. W. Hofstra; Wim A. Buurman; Sander S. Rensen; Marcel G. M. Wolfs; Mathieu Platteel; Alexandra Zhernakova; Clara C. Elbers; Eleanora M. Festen; Gosia Trynka; Marten H. Hofker; Christiaan G.J. Saris; Roel A. Ophoff; Leonard H. van den Berg; David A. van Heel; Cisca Wijmenga; Gerard J. te Meerman; Lude Franke

For many complex traits, genetic variants have been found associated. However, it is still mostly unclear through which downstream mechanism these variants cause these phenotypes. Knowledge of these intermediate steps is crucial to understand pathogenesis, while also providing leads for potential pharmacological intervention. Here we relied upon natural human genetic variation to identify effects of these variants on trans-gene expression (expression quantitative trait locus mapping, eQTL) in whole peripheral blood from 1,469 unrelated individuals. We looked at 1,167 published trait- or disease-associated SNPs and observed trans-eQTL effects on 113 different genes, of which we replicated 46 in monocytes of 1,490 different individuals and 18 in a smaller dataset that comprised subcutaneous adipose, visceral adipose, liver tissue, and muscle tissue. HLA single-nucleotide polymorphisms (SNPs) were 10-fold enriched for trans-eQTLs: 48% of the trans-acting SNPs map within the HLA, including ulcerative colitis susceptibility variants that affect plausible candidate genes AOAH and TRBV18 in trans. We identified 18 pairs of unlinked SNPs associated with the same phenotype and affecting expression of the same trans-gene (21 times more than expected, P<10−16). This was particularly pronounced for mean platelet volume (MPV): Two independent SNPs significantly affect the well-known blood coagulation genes GP9 and F13A1 but also C19orf33, SAMD14, VCL, and GNG11. Several of these SNPs have a substantially higher effect on the downstream trans-genes than on the eventual phenotypes, supporting the concept that the effects of these SNPs on expression seems to be much less multifactorial. Therefore, these trans-eQTLs could well represent some of the intermediate genes that connect genetic variants with their eventual complex phenotypic outcomes.


Hepatology | 2013

Population-based epidemiology, malignancy risk, and outcome of primary sclerosing cholangitis.

Kirsten Boonstra; Rinse K. Weersma; Karel J. van Erpecum; Erik A. J. Rauws; B.W. Marcel Spanier; Alexander C. Poen; Karin M.J. van Nieuwkerk; Joost P. H. Drenth; Ben J. Witteman; Hans Tuynman; Anton H. Naber; Paul J. Kingma; Henk R. van Buuren; Bart van Hoek; Frank P. Vleggaar; Nan van Geloven; Ulrich Beuers; Cyriel Y. Ponsioen

Extensive population‐based studies are much needed to accurately establish epidemiology and disease course in patients with primary sclerosing cholangitis (PSC). We aimed to obtain population‐based prevalence and incidence figures, insight in disease course with regard to survival, liver transplantation (LT), and occurrence of malignancies, as well as risk factors thereof. Four independent hospital databases were searched in 44 hospitals in a large geographically defined area of the Netherlands, comprising 50% of the population. In addition, all PSC patients in the three Dutch liver transplant centers and all inflammatory bowel disease (IBD) patients in the adherence area of a large district hospital were identified. All medical records were reviewed on‐site, verifying diagnosis. Five hundred and ninety PSC patients were identified, resulting in an incidence of 0.5 and a point prevalence of 6.0 per 100,000. Median follow up was 92 months. Estimated median survival from diagnosis until LT or PSC‐related death in the entire cohort was 21.3 years, as opposed to 13.2 years in the combined transplant centers cohort (n = 422; P < 0.0001). Colorectal carcinoma (CRC) risk was 10‐fold increased, as compared to ulcerative colitis controls, and developed at a much younger age (39 years; range, 26‐64), compared to IBD controls (59 years; range, 34‐73; P = 0.019). Colonoscopic surveillance was associated with significantly better outcome. Conclusion: This study exemplifies that, for relatively rare diseases, it is paramount to collect observational data from large, population‐based cohorts, because incidence and prevalence rates of PSC are markedly lower and survival much longer than previously reported. The selection of a bias‐free, population‐based cohort showed a significantly longer survival, compared to the tertiary referral cohort. CRC can develop at an early age, warranting surveillance from time of PSC diagnosis. (Hepatology 2013; 58:2045–2055)


The Lancet | 2016

Inherited determinants of Crohn's disease and ulcerative colitis phenotypes: a genetic association study

Isabelle Cleynen; Gabrielle Boucher; Luke Jostins; L. Philip Schumm; Sebastian Zeissig; Tariq Ahmad; Vibeke Andersen; Jane M. Andrews; Vito Annese; Stephan Brand; Steven R. Brant; Judy H. Cho; Mark J. Daly; Marla Dubinsky; Richard H. Duerr; Lynnette R. Ferguson; Andre Franke; Richard B. Gearry; Philippe Goyette; Hakon Hakonarson; Jonas Halfvarson; Johannes R. Hov; Hailang Huang; Nicholas A. Kennedy; Ian C. Lawrance; James C. Lee; Jack Satsangi; Stephan Schreiber; Emilie Théâtre; Andrea E. van der Meulen-de Jong

Summary Background Crohns disease and ulcerative colitis are the two major forms of inflammatory bowel disease; treatment strategies have historically been determined by this binary categorisation. Genetic studies have identified 163 susceptibility loci for inflammatory bowel disease, mostly shared between Crohns disease and ulcerative colitis. We undertook the largest genotype association study, to date, in widely used clinical subphenotypes of inflammatory bowel disease with the goal of further understanding the biological relations between diseases. Methods This study included patients from 49 centres in 16 countries in Europe, North America, and Australasia. We applied the Montreal classification system of inflammatory bowel disease subphenotypes to 34 819 patients (19 713 with Crohns disease, 14 683 with ulcerative colitis) genotyped on the Immunochip array. We tested for genotype–phenotype associations across 156 154 genetic variants. We generated genetic risk scores by combining information from all known inflammatory bowel disease associations to summarise the total load of genetic risk for a particular phenotype. We used these risk scores to test the hypothesis that colonic Crohns disease, ileal Crohns disease, and ulcerative colitis are all genetically distinct from each other, and to attempt to identify patients with a mismatch between clinical diagnosis and genetic risk profile. Findings After quality control, the primary analysis included 29 838 patients (16 902 with Crohns disease, 12 597 with ulcerative colitis). Three loci (NOD2, MHC, and MST1 3p21) were associated with subphenotypes of inflammatory bowel disease, mainly disease location (essentially fixed over time; median follow-up of 10·5 years). Little or no genetic association with disease behaviour (which changed dramatically over time) remained after conditioning on disease location and age at onset. The genetic risk score representing all known risk alleles for inflammatory bowel disease showed strong association with disease subphenotype (p=1·65 × 10−78), even after exclusion of NOD2, MHC, and 3p21 (p=9·23 × 10−18). Predictive models based on the genetic risk score strongly distinguished colonic from ileal Crohns disease. Our genetic risk score could also identify a small number of patients with discrepant genetic risk profiles who were significantly more likely to have a revised diagnosis after follow-up (p=6·8 × 10−4). Interpretation Our data support a continuum of disorders within inflammatory bowel disease, much better explained by three groups (ileal Crohns disease, colonic Crohns disease, and ulcerative colitis) than by Crohns disease and ulcerative colitis as currently defined. Disease location is an intrinsic aspect of a patients disease, in part genetically determined, and the major driver to changes in disease behaviour over time. Funding International Inflammatory Bowel Disease Genetics Consortium members funding sources (see Acknowledgments for full list).


American Journal of Human Genetics | 2008

Genetic analysis of innate immunity in Crohn's disease and ulcerative colitis identifies two susceptibility loci harboring CARD9 and IL18RAP.

Alexandra Zhernakova; Eleanora M. Festen; Lude Franke; Gosia Trynka; Cleo C. van Diemen; Alienke J. Monsuur; Marianna Bevova; Rian M. Nijmeijer; Ruben van 't Slot; Roel Heijmans; H. Marike Boezen; David A. van Heel; Adriaan A. van Bodegraven; Pieter Stokkers; Cisca Wijmenga; J. Bart A. Crusius; Rinse K. Weersma

The two main phenotypes of inflammatory bowel disease (IBD)--Crohns disease (CD) and ulcerative colitis (UC)--are chronic intestinal inflammatory disorders with a complex genetic background. Using a three-stage design, we performed a functional candidate-gene analysis of innate immune pathway in IBD. In phase I, we typed 354 SNPs from 85 innate immunity genes in 520 Dutch IBD patients (284 CD, 236 UC) and 808 controls. In phase II, ten autosomal SNPs showing association at p < 0.006 in phase I were replicated in a second cohort of 545 IBD patients (326 CD, 219 UC) and 360 controls. In phase III, four SNPs with p < 0.01 in the combined phase I and phase II analysis were genotyped in an additional 786 IBD samples (452 CD, 334 UC) and 768 independent controls. Joint analysis of 1851 IBD patients (1062 CD, 789 UC) and 1936 controls demonstrated strong association to the IL18RAP rs917997 SNP for both CD and UC (p(IBD) 1.9 x 10(-8); OR 1.35). Association in CD is independently supported by the Crohns disease dataset of the Wellcome Trust Case Control Consortium (imputed SNP rs917997, p = 9.19 x 10(-4)). In addition, an association of the CARD9 rs10870077 SNP to CD and UC was observed (p(IBD) = 3.25 x 10(-5); OR 1.21). Both genes are located in extended haplotype blocks on 2q11-2q12 and 9q34.3, respectively. Our results indicate two IBD loci and further support the importance of the innate immune system in the predisposition to both CD and UC.

Collaboration


Dive into the Rinse K. Weersma's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gerard Dijkstra

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lude Franke

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Floris Imhann

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Jan J. Koornstra

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Marijn C. Visschedijk

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Alexandra Zhernakova

University Medical Center Groningen

View shared research outputs
Researchain Logo
Decentralizing Knowledge