Network


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

Hotspot


Dive into the research topics where Marc Jan Bonder is active.

Publication


Featured researches published by Marc Jan Bonder.


Science | 2016

Population-level analysis of gut microbiome variation

Gwen Falony; Marie Joossens; Sara Vieira-Silva; Jun Wang; Youssef Darzi; Karoline Faust; Alexander Kurilshikov; Marc Jan Bonder; Mireia Valles-Colomer; Doris Vandeputte; Raul Y. Tito; Samuel Chaffron; Leen Rymenans; Chloë Verspecht; Lise De Sutter; Gipsi Lima-Mendez; Kevin D’hoe; Karl Jonckheere; Daniel Homola; Roberto Garcia; Ettje F. Tigchelaar; Linda Eeckhaudt; Jingyuan Fu; Liesbet Henckaerts; Alexandra Zhernakova; Cisca Wijmenga; Jeroen Raes

“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. Fecal microbiome variation in the average, healthy population has remained under-investigated. Here, we analyzed two independent, extensively phenotyped cohorts: the Belgian Flemish Gut Flora Project (FGFP; discovery cohort; N = 1106) and the Dutch LifeLines-DEEP study (LLDeep; replication; N = 1135). Integration with global data sets (N combined = 3948) revealed a 14-genera core microbiota, but the 664 identified genera still underexplore total gut diversity. Sixty-nine clinical and questionnaire-based covariates were found associated to microbiota compositional variation with a 92% replication rate. Stool consistency showed the largest effect size, whereas medication explained largest total variance and interacted with other covariate-microbiota associations. Early-life events such as birth mode were not reflected in adult microbiota composition. Finally, we found that proposed disease marker genera associated to host covariates, urging inclusion of the latter in study design.


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.


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.


Circulation Research | 2015

The Gut Microbiome Contributes to a Substantial Proportion of the Variation in Blood Lipids

Jingyuan Fu; Marc Jan Bonder; Maria Carmen Cenit; Ettje F. Tigchelaar; Astrid Maatman; Jackie A.M. Dekens; Eelke Brandsma; Joanna Marczynska; Floris Imhann; Rinse K. Weersma; Lude Franke; Tiffany W. Poon; Ramnik J. Xavier; Dirk Gevers; Marten H. Hofker; Cisca Wijmenga; Alexandra Zhernakova

Supplemental Digital Content is available in the text.


Nature Genetics | 2016

The effect of host genetics on the gut microbiome

Marc Jan Bonder; Alexander Kurilshikov; Ettje F. Tigchelaar; Zlatan Mujagic; Floris Imhann; Arnau Vich Vila; Patrick Deelen; Tommi Vatanen; Melanie Schirmer; Sanne P. Smeekens; Daria V. Zhernakova; Soesma A. Jankipersadsing; Martin Jaeger; Marije Oosting; Maria Carmen Cenit; Ad Masclee; Morris A. Swertz; Yang Li; Vinod Kumar; Leo A. B. Joosten; Hermie J. M. Harmsen; Rinse K. Weersma; Lude Franke; Marten H. Hofker; Ramnik J. Xavier; Daisy Jonkers; Mihai G. Netea; Cisca Wijmenga; Jingyuan Fu; Alexandra Zhernakova

The gut microbiome is affected by multiple factors, including genetics. In this study, we assessed the influence of host genetics on microbial species, pathways and gene ontology categories, on the basis of metagenomic sequencing in 1,514 subjects. In a genome-wide analysis, we identified associations of 9 loci with microbial taxonomies and 33 loci with microbial pathways and gene ontology terms at P < 5 × 10−8. Additionally, in a targeted analysis of regions involved in complex diseases, innate and adaptive immunity, or food preferences, 32 loci were identified at the suggestive level of P < 5 × 10−6. Most of our reported associations are new, including genome-wide significance for the C-type lectin molecules CLEC4F–CD207 at 2p13.3 and CLEC4A–FAM90A1 at 12p13. We also identified association of a functional LCT SNP with the Bifidobacterium genus (P = 3.45 × 10−8) and provide evidence of a gene–diet interaction in the regulation of Bifidobacterium abundance. Our results demonstrate the importance of understanding host–microbe interactions to gain better insight into human health.


Cell | 2016

Linking the Human Gut Microbiome to Inflammatory Cytokine Production Capacity

Melanie Schirmer; Sanne P. Smeekens; Hera Vlamakis; Martin Jaeger; Marije Oosting; Eric A. Franzosa; Rob ter Horst; Trees Jansen; Liesbeth Jacobs; Marc Jan Bonder; Alexander Kurilshikov; Jingyuan Fu; Leo A. B. Joosten; Alexandra Zhernakova; Curtis Huttenhower; Cisca Wijmenga; Mihai G. Netea; Ramnik J. Xavier

Gut microbial dysbioses are linked to aberrant immune responses, which are often accompanied by abnormal production of inflammatory cytokines. As part of the Human Functional Genomics Project (HFGP), we investigate how differences in composition and function of gut microbial communities may contribute to inter-individual variation in cytokine responses to microbial stimulations in healthy humans. We observe microbiome-cytokine interaction patterns that are stimulus specific, cytokine specific, and cytokine and stimulus specific. Validation of two predicted host-microbial interactions reveal that TNFα and IFNγ production are associated with specific microbial metabolic pathways: palmitoleic acid metabolism and tryptophan degradation to tryptophol. Besides providing a resource of predicted microbially derived mediators that influence immune phenotypes in response to common microorganisms, these data can help to define principles for understanding disease susceptibility. The three HFGP studies presented in this issue lay the groundwork for further studies aimed at understanding the interplay between microbial, genetic, and environmental factors in the regulation of the immune response in humans. PAPERCLIP.


Nature Genetics | 2017

Disease variants alter transcription factor levels and methylation of their binding sites

Marc Jan Bonder; René Luijk; Daria V. Zhernakova; Matthijs Moed; Patrick Deelen; Martijn Vermaat; Maarten van Iterson; Freerk van Dijk; Michiel van Galen; Jan Bot; Roderick C. Slieker; P. Mila Jhamai; Michael Verbiest; H. Eka D. Suchiman; Marijn Verkerk; Ruud van der Breggen; Jeroen van Rooij; N. Lakenberg; Wibowo Arindrarto; Szymon M. Kielbasa; Iris Jonkers; Peter van ‘t Hof; Irene Nooren; Marian Beekman; Joris Deelen; Diana van Heemst; Alexandra Zhernakova; Ettje F. Tigchelaar; Morris A. Swertz; Albert Hofman

Most disease-associated genetic variants are noncoding, making it challenging to design experiments to understand their functional consequences. Identification of expression quantitative trait loci (eQTLs) has been a powerful approach to infer the downstream effects of disease-associated variants, but most of these variants remain unexplained. The analysis of DNA methylation, a key component of the epigenome, offers highly complementary data on the regulatory potential of genomic regions. Here we show that disease-associated variants have widespread effects on DNA methylation in trans that likely reflect differential occupancy of trans binding sites by cis-regulated transcription factors. Using multiple omics data sets from 3,841 Dutch individuals, we identified 1,907 established trait-associated SNPs that affect the methylation levels of 10,141 different CpG sites in trans (false discovery rate (FDR) < 0.05). These included SNPs that affect both the expression of a nearby transcription factor (such as NFKB1, CTCF and NKX2-3) and methylation of its respective binding site across the genome. Trans methylation QTLs effectively expose the downstream effects of disease-associated variants.


PLOS ONE | 2012

The Relation between Oral Candida Load and Bacterial Microbiome Profiles in Dutch Older Adults

Eefje A. Kraneveld; Mark J. Buijs; Marc Jan Bonder; Marjolein Visser; Bart J. F. Keijser; Wim Crielaard; Egija Zaura

Currently there are no evidence-based ecological measures for prevention of overgrowth and subsequent infection by fungi in the oral cavity. The aim of this study was to increase our knowledge on fungal–bacterial ecological interactions. Salivary Candida abundance of 82 Dutch adults aged 58–80 years was established relative to the bacterial load by quantitative PCR analysis of the Internal Transcribed (ITS) region (Candida) and 16S rDNA gene (bacteria). The salivary microbiome was assessed using barcoded pyrosequencing of the bacterial hypervariable regions V5–V7 of 16S rDNA. Sequencing data was preprocessed by denoising and chimera removal, clustered in Operational Taxonomic Units (OTUs) and assigned to taxonomy. Both OTU-based (PCA, diversity statistics) and phylogeny-based analyses (UniFrac, PCoA) were performed. Saliva of Dutch older adults contained 0–4 × 108 CFU/mL Candida with a median Candida load of 0.06%. With increased Candida load the diversity of the salivary microbiome decreased significantly (p<0.001). Increase in the Candida load correlated positively with class Bacilli, and negatively with class Fusobacteria, Flavobacteria, and Bacteroidia. Microbiomes with high Candida load were less diverse and had a distinct microbial composition towards dominance by saccharolytic and acidogenic bacteria - streptococci. The control of the acidification of the oral environment may be a potential preventive measure for Candida outgrowth that should be evaluated in longitudinal clinical intervention trials.


Gut | 2018

Interplay of host genetics and gut microbiota underlying the onset and clinical presentation of inflammatory bowel disease

Floris Imhann; Arnau Vich Vila; Marc Jan Bonder; Jingyuan Fu; Dirk Gevers; Marijn C. Visschedijk; Lieke M. Spekhorst; Rudi Alberts; Lude Franke; Hendrik M. van Dullemen; Rinze W F Ter Steege; Curtis Huttenhower; Gerard Dijkstra; Ramnik J. Xavier; Eleonora A. Festen; Cisca Wijmenga; Alexandra Zhernakova; Rinse K. Weersma

Objective Patients with IBD display substantial heterogeneity in clinical characteristics. We hypothesise that individual differences in the complex interaction of the host genome and the gut microbiota can explain the onset and the heterogeneous presentation of IBD. Therefore, we performed a case–control analysis of the gut microbiota, the host genome and the clinical phenotypes of IBD. Design Stool samples, peripheral blood and extensive phenotype data were collected from 313 patients with IBD and 582 truly healthy controls, selected from a population cohort. The gut microbiota composition was assessed by tag-sequencing the 16S rRNA gene. All participants were genotyped. We composed genetic risk scores from 11 functional genetic variants proven to be associated with IBD in genes that are directly involved in the bacterial handling in the gut: NOD2, CARD9, ATG16L1, IRGM and FUT2. Results Strikingly, we observed significant alterations of the gut microbiota of healthy individuals with a high genetic risk for IBD: the IBD genetic risk score was significantly associated with a decrease in the genus Roseburia in healthy controls (false discovery rate 0.017). Moreover, disease location was a major determinant of the gut microbiota: the gut microbiota of patients with colonic Crohns disease (CD) is different from that of patients with ileal CD, with a decrease in alpha diversity associated to ileal disease (p=3.28×10−13). Conclusions We show for the first time that genetic risk variants associated with IBD influence the gut microbiota in healthy individuals. Roseburia spp are acetate-to-butyrate converters, and a decrease has already been observed in patients with IBD.


Nature Genetics | 2017

Identification of context-dependent expression quantitative trait loci in whole blood

Daria V. Zhernakova; Patrick Deelen; Martijn Vermaat; Maarten van Iterson; Michiel van Galen; Wibowo Arindrarto; Peter van ‘t Hof; Hailiang Mei; Freerk van Dijk; Harm-Jan Westra; Marc Jan Bonder; Jeroen van Rooij; Marijn Verkerk; P. Mila Jhamai; Matthijs Moed; Szymon M. Kielbasa; Jan Bot; Irene Nooren; René Pool; Jenny van Dongen; Jouke J. Hottenga; Coen D. A. Stehouwer; Carla J.H. van der Kallen; Casper G. Schalkwijk; Alexandra Zhernakova; Yang Li; Ettje F. Tigchelaar; Niek de Klein; Marian Beekman; Joris Deelen

Genetic risk factors often localize to noncoding regions of the genome with unknown effects on disease etiology. Expression quantitative trait loci (eQTLs) help to explain the regulatory mechanisms underlying these genetic associations. Knowledge of the context that determines the nature and strength of eQTLs may help identify cell types relevant to pathophysiology and the regulatory networks underlying disease. Here we generated peripheral blood RNA–seq data from 2,116 unrelated individuals and systematically identified context-dependent eQTLs using a hypothesis-free strategy that does not require previous knowledge of the identity of the modifiers. Of the 23,060 significant cis-regulated genes (false discovery rate (FDR) ≤ 0.05), 2,743 (12%) showed context-dependent eQTL effects. The majority of these effects were influenced by cell type composition. A set of 145 cis-eQTLs depended on type I interferon signaling. Others were modulated by specific transcription factors binding to the eQTL SNPs.

Collaboration


Dive into the Marc Jan Bonder's collaboration.

Top Co-Authors

Avatar

Alexandra Zhernakova

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Cisca Wijmenga

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Lude Franke

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Jingyuan Fu

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Ettje F. Tigchelaar

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Patrick Deelen

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alexander Kurilshikov

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Daria V. Zhernakova

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Marten H. Hofker

University Medical Center Groningen

View shared research outputs
Researchain Logo
Decentralizing Knowledge