Network


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

Hotspot


Dive into the research topics where Nils Bomer is active.

Publication


Featured researches published by Nils Bomer.


Nature Genetics | 2014

Severe osteoarthritis of the hand associates with common variants within the ALDH1A2 gene and with rare variants at 1p31.

Unnur Styrkarsdottir; Gudmar Thorleifsson; Hafdis T. Helgadottir; Nils Bomer; Sarah Metrustry; Sita M. A. Bierma-Zeinstra; Annelieke M Strijbosch; Evangelos Evangelou; Deborah J. Hart; Marian Beekman; Aslaug Jonasdottir; Asgeir Sigurdsson; Finnur F. Eiriksson; Margret Thorsteinsdottir; Michael L. Frigge; Augustine Kong; Sigurjon A. Gudjonsson; Olafur T. Magnusson; Gisli Masson; Albert Hofman; N K Arden; Thorvaldur Ingvarsson; Stefan Lohmander; Margreet Kloppenburg; Fernando Rivadeneira; Rob G. H. H. Nelissen; Tim D. Spector; André G. Uitterlinden; P. Eline Slagboom; Unnur Thorsteinsdottir

Osteoarthritis is the most common form of arthritis and is a major cause of pain and disability in the elderly. To search for sequence variants that confer risk of osteoarthritis of the hand, we carried out a genome-wide association study (GWAS) in subjects with severe hand osteoarthritis, using variants identified through the whole-genome sequencing of 2,230 Icelanders. We found two significantly associated loci in the Icelandic discovery set: at 15q22 (frequency of 50.7%, odds ratio (OR) = 1.51, P = 3.99 × 10−10) in the ALDH1A2 gene and at 1p31 (frequency of 0.02%, OR = 50.6, P = 9.8 × 10−10). Among the carriers of the variant at 1p31 is a family with several members in whom the risk allele segregates with osteoarthritis. The variants within the ALDH1A2 gene were confirmed in replication sets from The Netherlands and the UK, yielding an overall association of OR = 1.46 and P = 1.1 × 10−11 (rs3204689).


Annals of the Rheumatic Diseases | 2014

Knee and hip articular cartilage have distinct epigenomic landscapes: implications for future cartilage regeneration approaches

W. den Hollander; Y.F. Ramos; S.D. Bos; Nils Bomer; R. van der Breggen; N. Lakenberg; W.J. de Dijcker; B.J. Duijnisveld; P.E. Slagboom; Rob G. H. H. Nelissen; Ingrid Meulenbelt

Objectives To elucidate the functional epigenomic landscape of articular cartilage in osteoarthritis (OA) affected knee and hip joints in relation to gene expression. Methods Using Illumina Infinium HumanMethylation450 BeadChip arrays, genome-wide DNA methylation was measured in 31 preserved and lesioned cartilage sample pairs (14 knees and 17 hips) from patients who underwent a total joint replacement due to primary OA. Using previously published genome-wide expression data of 33 pairs of cartilage samples, of which 13 pairs were overlapping with the current methylation dataset, we assessed gene expression differences in differentially methylated regions (DMRs). Results Principal component analysis of the methylation data revealed distinct clustering of knee and hip samples, irrespective of OA pathophysiology. A total of 6272 CpG dinucleotides were differentially methylated between the two joints, comprising a total of 357 DMRs containing 1817 CpGs and 245 unique genes. Enrichment analysis of genes proximal of the DMRs revealed significant enrichment for developmental pathways and homeobox (HOX) genes. Subsequent transcriptomic analysis of DMR genes exposed distinct knee and hip expression patterns. Conclusions Our findings reveal consistent DMRs between knee and hip articular cartilage that marked transcriptomic differences among HOX genes, which were not reflecting the temporal sequential HOX expression pattern during development. This implies distinct mechanisms for maintaining cartilage integrity in adulthood, thereby contributing to our understanding of cartilage homeostasis and future tissue regeneration approaches.


PLOS ONE | 2014

Genes involved in the osteoarthritis process identified through genome wide expression analysis in articular cartilage; the RAAK study

Y.F. Ramos; Wouter den Hollander; Judith V. M. G. Bovée; Nils Bomer; Ruud van der Breggen; N. Lakenberg; J. Christiaan Keurentjes; Jelle J. Goeman; P. Eline Slagboom; Rob G. H. H. Nelissen; S.D. Bos; Ingrid Meulenbelt

Objective Identify gene expression profiles associated with OA processes in articular cartilage and determine pathways changing during the disease process. Methods Genome wide gene expression was determined in paired samples of OA affected and preserved cartilage of the same joint using microarray analysis for 33 patients of the RAAK study. Results were replicated in independent samples by RT-qPCR and immunohistochemistry. Profiles were analyzed with the online analysis tools DAVID and STRING to identify enrichment for specific pathways and protein-protein interactions. Results Among the 1717 genes that were significantly differently expressed between OA affected and preserved cartilage we found significant enrichment for genes involved in skeletal development (e.g. TNFRSF11B and FRZB). Also several inflammatory genes such as CD55, PTGES and TNFAIP6, previously identified in within-joint analyses as well as in analyses comparing preserved cartilage from OA affected joints versus healthy cartilage were among the top genes. Of note was the high up-regulation of NGF in OA cartilage. RT-qPCR confirmed differential expression for 18 out of 19 genes with expression changes of 2-fold or higher, and immunohistochemistry of selected genes showed a concordant change in protein expression. Most of these changes associated with OA severity (Mankin score) but were independent of joint-site or sex. Conclusion We provide further insights into the ongoing OA pathophysiological processes in cartilage, in particular into differences in macroscopically intact cartilage compared to OA affected cartilage, which seem relatively consistent and independent of sex or joint. We advocate that development of treatment could benefit by focusing on these similarities in gene expression changes and/or pathways.


Annals of the Rheumatic Diseases | 2014

Underlying molecular mechanisms of DIO2 susceptibility in symptomatic osteoarthritis.

Nils Bomer; Wouter den Hollander; Y.F. Ramos; S.D. Bos; Ruud van der Breggen; N. Lakenberg; Barry Antonius Pepers; Annelies E van Eeden; Arash Darvishan; Elmar W. Tobi; B.J. Duijnisveld; Erik B. van den Akker; Bastiaan T. Heijmans; Willeke Mc van Roon-Mom; Fons J. Verbeek; Gerjo J.V.M. van Osch; Rob G. H. H. Nelissen; P. Eline Slagboom; Ingrid Meulenbelt

Objectives To investigate how the genetic susceptibility gene DIO2 confers risk to osteoarthritis (OA) onset in humans and to explore whether counteracting the deleterious effect could contribute to novel therapeutic approaches. Methods Epigenetically regulated expression of DIO2 was explored by assessing methylation of positional CpG-dinucleotides and the respective DIO2 expression in OA-affected and macroscopically preserved articular cartilage from end-stage OA patients. In a human in vitro chondrogenesis model, we measured the effects when thyroid signalling during culturing was either enhanced (excess T3 or lentiviral induced DIO2 overexpression) or decreased (iopanoic acid). Results OA-related changes in methylation at a specific CpG dinucleotide upstream of DIO2 caused significant upregulation of its expression (β=4.96; p=0.0016). This effect was enhanced and appeared driven specifically by DIO2 rs225014 risk allele carriers (β=5.58, p=0.0006). During in vitro chondrogenesis, DIO2 overexpression resulted in a significant reduced capacity of chondrocytes to deposit extracellular matrix (ECM) components, concurrent with significant induction of ECM degrading enzymes (ADAMTS5, MMP13) and markers of mineralisation (ALPL, COL1A1). Given their concurrent and significant upregulation of expression, this process is likely mediated via HIF-2α/RUNX2 signalling. In contrast, we showed that inhibiting deiodinases during in vitro chondrogenesis contributed to prolonged cartilage homeostasis as reflected by significant increased deposition of ECM components and attenuated upregulation of matrix degrading enzymes. Conclusions Our findings show how genetic variation at DIO2 could confer risk to OA and raised the possibility that counteracting thyroid signalling may be a novel therapeutic approach.


Arthritis & Rheumatism | 2015

Transcriptional Associations of Osteoarthritis‐Mediated Loss of Epigenetic Control in Articular Cartilage

Wouter den Hollander; Y.F. Ramos; Nils Bomer; Stefan Elzinga; Ruud van der Breggen; N. Lakenberg; Wesley J. de Dijcker; H. Eka D. Suchiman; B.J. Duijnisveld; Jeanine J. Houwing-Duistermaat; P. Eline Slagboom; S.D. Bos; Rob G. H. H. Nelissen; Ingrid Meulenbelt

To identify osteoarthritis (OA) progression–modulating pathways in articular cartilage and their respective regulatory epigenetic and genetic determinants in end‐stage disease.


Annals of the Rheumatic Diseases | 2016

The effect of forced exercise on knee joints in Dio2−/− mice: type II iodothyronine deiodinase-deficient mice are less prone to develop OA-like cartilage damage upon excessive mechanical stress

Nils Bomer; F.M. Cornelis; Y.F. Ramos; Wouter den Hollander; L. Storms; Ruud van der Breggen; N. Lakenberg; P. Eline Slagboom; Ingrid Meulenbelt; Rik Jl Lories

Objective To further explore deiodinase iodothyronine type 2 (DIO2) as a therapeutic target in osteoarthritis (OA) by studying the effects of forced mechanical loading on in vivo joint cartilage tissue homeostasis and the modulating effect herein of Dio2 deficiency. Methods Wild-type and C57BL/6-Dio2−/− -mice were subjected to a forced running regime for 1 h per day for 3 weeks. Severity of OA was assessed by histological scoring for cartilage damage and synovitis. Genome-wide gene expression was determined in knee cartilage by microarray analysis (Illumina MouseWG-6 v2). STRING-db analyses were applied to determine enrichment for specific pathways and to visualise protein–protein interactions. Results In total, 158 probes representing 147 unique genes showed significantly differential expression with a fold-change ≥1.5 upon forced exercise. Among these are genes known for their association with OA (eg, Mef2c, Egfr, Ctgf, Prg4 and Ctnnb1), supporting the use of forced running as an OA model in mice. Dio2-deficient mice showed significantly less cartilage damage and signs of synovitis. Gene expression response upon exercise between wild-type and knockout mice was significantly different for 29 genes. Conclusions Mice subjected to a running regime have significant increased cartilage damage and synovitis scores. Lack of Dio2 protected against cartilage damage in this model and was reflected in a specific gene expression profile, and either mark a favourable effect in the Dio2 knockout (eg, Gnas) or an unfavourable effect in wild-type cartilage homeostasis (eg, Hmbg2 and Calr). These data further support DIO2 activity as a therapeutic target in OA.


Best Practice & Research: Clinical Rheumatology | 2015

Translating genomics into mechanisms of disease: Osteoarthritis

Nils Bomer; Wouter den Hollander; Y.F. Ramos; Ingrid Meulenbelt

Osteoarthritis (OA) is the most common age-related arthritic disorder and is characterized by aberrant extracellular matrix (ECM) content and surface disruptions that range from fibrillation, clefting and delamination, leading to articular surface erosion. Worldwide, over 20% of the population is affected with OA and 80% of these patients have limitations in movement, whereas 25% experience inhibition in major daily activities of life. OA is the most common disabling arthritic disease; nevertheless, no disease-modifying treatment is available except for the expensive total joint replacement surgery at end-stage disease. Lack of insight into the underlying pathophysiological mechanisms of OA has considerably contributed to the inability of the scientific community to develop disease-modifying drugs. To overcome this critical barrier, focus should be on translation of identified robust gene deviations towards the underlying biological mechanisms.


Arthritis & Rheumatism | 2018

Annotating transcriptional effects of genetic variants in disease relevant tissue: Transcriptome-wide allelic imbalance in osteoarthritic cartilage

Wouter den Hollander; Irina Pulyakhina; C.G. Boer; Nils Bomer; Ruud van der Breggen; Wibowo Arindrarto; Rodrigo Couthino de Almeida; N. Lakenberg; Thom Sentner; Jeroen F.J. Laros; Peter A. C. 't Hoen; Eline Slagboom; Rob G. H. H. Nelissen; Joyce B. J. van Meurs; Y.F. Ramos; Ingrid Meulenbelt

Multiple single‐nucleotide polymorphisms (SNPs) conferring susceptibility to osteoarthritis (OA) mark imbalanced expression of positional genes in articular cartilage, reflected by unequally expressed alleles among heterozygotes (allelic imbalance [AI]). We undertook this study to explore the articular cartilage transcriptome from OA patients for AI events to identify putative disease‐driving genetic variation.


PLOS ONE | 2016

Aberrant Calreticulin Expression in Articular Cartilage of Dio2 Deficient Mice

Nils Bomer; F.M. Cornelis; Y.F. Ramos; Wouter den Hollander; N. Lakenberg; Ruud van der Breggen; L. Storms; P. Eline Slagboom; Rik Lories; Ingrid Meulenbelt

Objective To identify intrinsic differences in cartilage gene expression profiles between wild-type- and Dio2-/--mice, as a mechanism to investigate factors that contribute to prolonged healthy tissue homeostasis. Methods Previously generated microarray-data (Illumina MouseWG-6 v2) of knee cartilage of wild-type and Dio2 -/- -mice were re-analyzed to identify differential expressed genes independent of mechanical loading conditions by forced treadmill-running. RT-qPCR and western blot analyses of overexpression and knockdown of Calr in mouse chondro-progenitor cells (ATDC5) were applied to assess the direct effect of differential Calr expression on cartilage deposition. Results Differential expression analyses of articular cartilage of Dio2-/- (N = 9) and wild-type-mice (N = 11) while applying a cutoff threshold (P < 0.05 (FDR) and FC > |1,5|) resulted in 1 probe located in Calreticulin (Calr) that was found significantly downregulated in Dio2-/- mice (FC = -1.731; P = 0.044). Furthermore, overexpression of Calr during early chondrogenesis in ATDC5 cells leads to decreased proteoglycan deposition and corresponding lower Aggrecan expression, whereas knocking down Calr expression does not lead to histological differences of matrix composition. Conclusion We here demonstrate that the beneficial homeostatic state of articular cartilage in Dio2-/- mice is accompanied with significant lower expression of Calr. Functional analyses further showed that upregulation of Calr expression could act as an initiator of cartilage destruction. The consistent association between Calr and Dio2 expression suggests that enhanced expression of these genes facilitate detrimental effects on cartilage integrity.


Annals of the Rheumatic Diseases | 2015

Extended report: Underlying molecular mechanisms of DIO2 susceptibility in symptomatic osteoarthritis

Nils Bomer; Wouter den Hollander; Y.F. Ramos; S.D. Bos; Ruud van der Breggen; N. Lakenberg; Barry A. Pepers; Annelies E van Eeden; Arash Darvishan; Elmar W. Tobi; B.J. Duijnisveld; Erik B. van den Akker; Bastiaan T. Heijmans; Willeke M. C. van Roon-Mom; Fons J. Verbeek; Gerjo J.V.M. van Osch; Rob G. H. H. Nelissen; P. Eline Slagboom; Ingrid Meulenbelt

Objectives To investigate how the genetic susceptibility gene DIO2 confers risk to osteoarthritis (OA) onset in humans and to explore whether counteracting the deleterious effect could contribute to novel therapeutic approaches. Methods Epigenetically regulated expression of DIO2 was explored by assessing methylation of positional CpG-dinucleotides and the respective DIO2 expression in OA-affected and macroscopically preserved articular cartilage from end-stage OA patients. In a human in vitro chondrogenesis model, we measured the effects when thyroid signalling during culturing was either enhanced (excess T3 or lentiviral induced DIO2 overexpression) or decreased (iopanoic acid). Results OA-related changes in methylation at a specific CpG dinucleotide upstream of DIO2 caused significant upregulation of its expression (β=4.96; p=0.0016). This effect was enhanced and appeared driven specifically by DIO2 rs225014 risk allele carriers (β=5.58, p=0.0006). During in vitro chondrogenesis, DIO2 overexpression resulted in a significant reduced capacity of chondrocytes to deposit extracellular matrix (ECM) components, concurrent with significant induction of ECM degrading enzymes (ADAMTS5, MMP13) and markers of mineralisation (ALPL, COL1A1). Given their concurrent and significant upregulation of expression, this process is likely mediated via HIF-2α/RUNX2 signalling. In contrast, we showed that inhibiting deiodinases during in vitro chondrogenesis contributed to prolonged cartilage homeostasis as reflected by significant increased deposition of ECM components and attenuated upregulation of matrix degrading enzymes. Conclusions Our findings show how genetic variation at DIO2 could confer risk to OA and raised the possibility that counteracting thyroid signalling may be a novel therapeutic approach.

Collaboration


Dive into the Nils Bomer's collaboration.

Top Co-Authors

Avatar

Ingrid Meulenbelt

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Y.F. Ramos

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Rob G. H. H. Nelissen

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

N. Lakenberg

Loyola University Medical Center

View shared research outputs
Top Co-Authors

Avatar

W. den Hollander

Loyola University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wouter den Hollander

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

B.J. Duijnisveld

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

R. van der Breggen

Loyola University Medical Center

View shared research outputs
Top Co-Authors

Avatar

P. Eline Slagboom

Leiden University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge