Network


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

Hotspot


Dive into the research topics where Martin C. Wapenaar is active.

Publication


Featured researches published by Martin C. Wapenaar.


Nature Genetics | 2008

Newly identified genetic risk variants for celiac disease related to the immune response

Karen A. Hunt; Alexandra Zhernakova; Graham Turner; Graham A. Heap; Lude Franke; Marcel Bruinenberg; Jihane Romanos; Lotte C. Dinesen; Anthony W. Ryan; Davinder Panesar; Rhian Gwilliam; Fumihiko Takeuchi; William M. McLaren; Geoffrey Holmes; Peter D. Howdle; Julian R. Walters; David S. Sanders; Raymond J. Playford; Gosia Trynka; Chris Jj Mulder; M. Luisa Mearin; Wieke H. Verbeek; Valerie Trimble; Fiona M. Stevens; Colm O'Morain; N. P. Kennedy; Dermot Kelleher; Daniel J. Pennington; David P. Strachan; Wendy L. McArdle

Our genome-wide association study of celiac disease previously identified risk variants in the IL2–IL21 region. To identify additional risk variants, we genotyped 1,020 of the most strongly associated non-HLA markers in an additional 1,643 cases and 3,406 controls. Through joint analysis including the genome-wide association study data (767 cases, 1,422 controls), we identified seven previously unknown risk regions (P < 5 × 10−7). Six regions harbor genes controlling immune responses, including CCR3, IL12A, IL18RAP, RGS1, SH2B3 (nsSNP rs3184504) and TAGAP. Whole-blood IL18RAP mRNA expression correlated with IL18RAP genotype. Type 1 diabetes and celiac disease share HLA-DQ, IL2–IL21, CCR3 and SH2B3 risk regions. Thus, this extensive genome-wide association follow-up study has identified additional celiac disease risk variants in relevant biological pathways.


Nature Genetics | 2005

Myosin IXB variant increases the risk of celiac disease and points toward a primary intestinal barrier defect

Alienke J. Monsuur; Paul I. W. de Bakker; Behrooz Z. Alizadeh; Alexandra Zhernakova; Marianna Bevova; Eric Strengman; Lude Franke; Ruben van 't Slot; Martine van Belzen; I.C.M. Lavrijsen; Begoña Diosdado; Mark J. Daly; Chris J. Mulder; M. Luisa Mearin; Jos W. R. Meijer; Gerrit A. Meijer; Erica van Oort; Martin C. Wapenaar; Bobby P. C. Koeleman; Cisca Wijmenga

Celiac disease is probably the best-understood immune-related disorder. The disease presents in the small intestine and results from the interplay between multiple genes and gluten, the triggering environmental factor. Although HLA class II genes explain 40% of the heritable risk, non-HLA genes accounting for most of the familial clustering have not yet been identified. Here we report significant and replicable association (P = 2.1 × 10−6) to a common variant located in intron 28 of the gene myosin IXB (MYO9B), which encodes an unconventional myosin molecule that has a role in actin remodeling of epithelial enterocytes. Individuals homozygous with respect to the at-risk allele have a 2.3-times higher risk of celiac disease (P = 1.55 × 10−5). This result is suggestive of a primary impairment of the intestinal barrier in the etiology of celiac disease, which may explain why immunogenic gluten peptides are able to pass through the epithelial barrier.


Gut | 2007

Associations with tight junction genes PARD3 and MAGI2 in Dutch patients point to a common barrier defect for coeliac disease and ulcerative colitis.

Martin C. Wapenaar; Alienke J. Monsuur; A.A. van Bodegraven; Rinse K. Weersma; Marianna Bevova; R. K. Linskens; Peter D. Howdle; Geoffrey Holmes; Chris Jj Mulder; Gerard Dijkstra; D A van Heel; Cisca Wijmenga

Background: Coeliac disease (gluten-sensitive enteropathy; GSE) and inflammatory bowel disease (IBD) are common gastrointestinal disorders. Both display enhanced intestinal permeability, initiated by gluten exposure (GSE) or bacterial interactions (IBD). Previous studies showed the association of both diseases with variants in MYO9B, presumably involved in epithelial permeability. Aim: It was hypothesised that genetic variants in tight junction genes might affect epithelial barrier function, thus contributing to a shared pathogenesis of GSE and IBD. Methods: This hypothesis was tested with a comprehensive genetic association analysis of 41 genes from the tight junction pathway, represented by 197 tag single nucleotide polymorphism (SNP) markers. Results: Two genes, PARD3 (two SNPs) and MAGI2 (two SNPs), showed weak association with GSE in a Dutch cohort. Replication in a British GSE cohort yielded significance for one SNP in PARD3 and suggestive associations for two additional SNPs, one each in PARD3 and MAGI2. Joint analysis of the British and Dutch data further substantiated the association for both PARD3 (rs10763976, p = 6.4×10−5; OR 1.23, 95% CI 1.11 to 1.37) and MAGI2 (rs6962966, p = 7.6×10−4; OR 1.19, 95% CI 1.08 to 1.32). Association was also observed in Dutch ulcerative colitis patients with MAGI2 (rs6962966, p = 0.0036; OR 1.26, 95% CI 1.08 to 1.47), and suggestive association with PARD3 (rs4379776, p = 0.068). Conclusions: These results suggest that coeliac disease and ulcerative colitis may share a common aetiology through tight junction-mediated barrier defects, although the observations need further replication.


Genomics | 1988

A deletion hot spot in the Duchenne muscular dystrophy gene

Martin C. Wapenaar; T. Kievits; K.A. Hart; Stephen Abbs; L. A. J. Blonden; J.T. den Dunnen; P. M. Grootscholten; E. Bakker; Ch. Verellen-Dumoulin; M. Bobrow; G.J.B. van Ommen; P.L. Pearson

We have made a detailed study of a deletion hot spot in the distal half of the Duchenne muscular dystrophy (DMD) gene, using intragenic probe P20 (DXS269), isolated by a hybrid cell-mediated cloning procedure. P20 detects 16% deletions in patients suffering from either DMD or Becker muscular dystrophy (BMD), in sharp contrast to the adjacent intragenic markers JBir (7%) and J66 (less than 1%), mapping respectively 200-320 kb proximal and 380-500 kb distal to P20. Of the P20 deletions, 30% start within a region of 25-40 kb, the majority extending distally. P20 was confirmed to map internal to a distal intron of the DMD gene. This region was recently shown by both cDNA analysis (M. Koenig et al., 1987; Cell 50: 509-517), and field inversion electrophoresis studies (J.T. Den Dunnen et al., 1987, Nature (London) 329: 640-642) to be specifically prone to deletions. In addition, P20 detects MspI and EcoRV RFLPs, informative in 48% of the carrier females. Together, these properties make P20 useful for carrier detection, prenatal diagnosis, and the study of deletion induction in both DMD and BMD.


Gut | 2004

A microarray screen for novel candidate genes in coeliac disease pathogenesis

Begoña Diosdado; Martin C. Wapenaar; Lude Franke; Karen Duran; M J Goerres; M Hadithi; J B A Crusius; Jos W. R. Meijer; David Duggan; Cjj Mulder; Frank C. P. Holstege; Cisca Wijmenga

Background and aims: The causative molecular pathways underlying the pathogenesis of coeliac disease are poorly understood. To unravel novel aspects of disease pathogenesis, we used microarrays to determine changes in gene expression of duodenal biopsies. Methods: cDNA microarrays representing 19 200 genes were used to compare gene expression profiles of duodenal biopsies from 15 coeliac disease patients with villous atrophy (Marsh III) and seven control individuals with normal biopsies (Marsh 0). In addition, the specific effect of gluten was studied by comparing the expression profiles of Marsh III lesions of seven patients exposed to gluten with four patients on a gluten free diet. Results: Comparing Marsh III with Marsh 0 lesions identified 109 genes that differed significantly (p<0.001) in expression levels between patients and controls. A large number of these genes have functions in proliferation and differentiation pathways and might be important for correct development of crypt-villous units. Alterations in these pathways may lead to the characteristic hyperplasia and villous atrophy seen in coeliac disease. The analyses also revealed 120 differentially expressed genes (p<0.005) when comparing patients on a gluten free diet with those exposed to gluten. These genes further strengthen our observation of increased cell proliferation in the presence of gluten. Conclusions: Our study provides new candidate genes in the pathogenesis of coeliac disease. Based on our results, we hypothesise that villous atrophy in coeliac disease patients is due to failure in cell differentiation. These genes are involved in pathways not previously implicated in coeliac disease pathogenesis and they may provide new targets for therapy.


Journal of Pediatric Gastroenterology and Nutrition | 2011

Functional characterization of mutations in the myosin Vb gene associated with microvillus inclusion disease

Agata Szperl; Magdalena R. Golachowska; Marcel Bruinenberg; Rytis Prekeris; Andy-Mark W. H. Thunnissen; Arend Karrenbeld; Gerard Dijkstra; Dick Hoekstra; David F. Mercer; Janusz Ksiazyk; Cisca Wijmenga; Martin C. Wapenaar; Edmond H. H. M. Rings; Sven C.D. van IJzendoorn

Objectives: Microvillus inclusion disease (MVID) is a rare autosomal recessive enteropathy characterized by intractable diarrhea and malabsorption. Recently, various MYO5B gene mutations have been identified in patients with MVID. Interestingly, several patients with MVID showed only a MYO5B mutation in 1 allele (heterozygous) or no mutations in the MYO5B gene, illustrating the need to further functionally characterize the cell biological effects of the MYO5B mutations. Patients and Methods: The genomic DNA of 9 patients diagnosed as having MVID was screened for MYO5B mutations, and quantitative polymerase chain reaction and immunohistochemistry on the material of 2 patients was performed to investigate resultant cellular consequences. Results: We demonstrate for the first time that MYO5B mutations can be correlated with altered myosin Vb messenger RNA expression and with an aberrant subcellular distribution of the myosin Vb protein. Moreover, we demonstrate that the typical and myosin Vb–controlled accumulation of Rab11a- and FIP5-positive recycling endosomes in the apical cytoplasm of the cells is abolished in MVID enterocytes, which is indicative of altered myosin Vb function. Moreover, we report 8 novel MYO5B mutations in 9 patients of various ethnic backgrounds with MVID, including compound heterozygous mutations. Conclusions: Our functional analysis indicates that MYO5B mutations can be correlated with an aberrant subcellular distribution of the myosin Vb protein, and apical recycling endosomes, which, together with the additional compound heterozygous mutations, significantly strengthen the link between MYO5B and MVID.


Genomics | 1991

Generation and fluorescent in situ hybridization mapping of yeast artificial chromosomes of 1p, 17p, 17q, and 19q from a hybrid cell line by high-density screening of an amplified library

M.S. Driesen; J.G. Dauwerse; Martin C. Wapenaar; E. J. Meershoek; P. Mollevanger; K.L. Chen; Kenneth H. Fischbeck; G.J.B. van Ommen

A yeast artificial chromosome (YAC) library has been constructed from a somatic cell hybrid containing a t(1p;19q) chromosome and chromosome 17. After amplification, part of this library was analyzed by high-density colony filter screening with a repetitive human DNA probe (Alu). The human YACs distinguished by the screening were further analyzed by Alu fingerprinting and Alu PCR. Fluorescent in situ hybridization (FISH) was performed to localize the YACs to subchromosomal regions of chromosome 1p, 17, or 19q. We have obtained a panel of 123 individual YACs with a mean size of 160 kb, and 77 of these were regionally localized by FISH: 33 to 1p, 10 to 17p, 25 to 17q, and 9 to 19q. The YACs cover a total of 19.7 Mb or 9% of the 220 Mb of human DNA contained in the hybrid. No overlapping YACs have yet been detected. These YACs are available upon request and should be helpful in mapping studies of disease loci, e.g., Charcot-Marie-Tooth disease, Miller-Dieker syndrome, hereditary breast tumor, myotonic dystrophy, and malignant hyperthermia.


Epilepsia | 2006

A Novel Splicing Mutation in KCNQ2 in a Multigenerational Family with BFNC Followed for 25 Years

Gerrit-Jan de Haan; Dalila Pinto; Dietbrandt Carton; Adri J. Bader; Jorien Witte; Edith Peters; Gerard van Erp; Willem Vandereyken; Eduard H. J. F. Boezeman; Martin C. Wapenaar; Paul Boon; Dicky Halley; Bobby P. C. Koeleman; Dick Lindhout

Summary:  Purpose: A large multigenerational family with benign familial neonatal convulsions (BFNC) was revisited to identify the disease‐causing mutation and to assess long‐term outcome.


European Journal of Human Genetics | 2006

A functional candidate screen for coeliac disease genes.

Christine R. Curley; Alienke J. Monsuur; Martin C. Wapenaar; John D. Rioux; Cisca Wijmenga

It is increasingly evident that different inflammatory disorders show some overlap in their pathological features, concurrence in families and individuals, and shared genetic factors. This might also be true for coeliac disease, a chronic inflammatory disorder of the gastrointestinal system, which shares two linkage regions with inflammatory bowel disease: on chromosome 5q31 (CELIAC2 and IBD5) and 19p13 (CELIAC4 and IBD6). We hypothesised that these regions contain genes that contribute to susceptibility to both disorders. The overlapping 5q31 region contains only five positional candidate genes, whereas the overlapping 19p13 region has 141 genes. As the common disease gene probably plays a role in inflammation, we selected five functional candidate genes from the 19p13 region. We studied these 10 positional and functional candidate genes in our Dutch coeliac disease cohort using 44 haplotype tagging single-nucleotide polymorphisms. Two genes from 19p13 showed a small effect on familial clustering: the cytochrome P450 F3 gene CYP4F3 (Pnominal 0.0375, odds ratio (OR) 1.77) and CYP4F2 (Pnominal 0.013, OR 1.33). CYP4F3 and CYP4F2 catalyse the inactivation of leukotriene B4 (LTB4), a potent mediator of inflammation responsible for recruitment and activation of neutrophils. The genetic association of LTB4-regulating gene variants connects the innate immune response of neutrophil mobilisation with that of the established Th1 adaptive immunity present in coeliac disease patients. The findings in coeliac disease need to be replicated. Expanding genetic association studies of these cytochrome genes to other inflammatory conditions should reveal whether their causative influence extends beyond coeliac disease.


Immunogenetics | 2007

The SPINK gene family and celiac disease susceptibility

Martin C. Wapenaar; Alienke J. Monsuur; Jos Bernard Poell; Ruben van 't Slot; Jos W. R. Meijer; Gerrit A. Meijer; Chris J. Mulder; Maria Luisa Mearin; Cisca Wijmenga

The gene family of serine protease inhibitors of the Kazal type (SPINK) are functional and positional candidate genes for celiac disease (CD). Our aim was to assess the gut mucosal gene expression and genetic association of SPINK1, -2, -4, and -5 in the Dutch CD population. Gene expression was determined for all four SPINK genes by quantitative reverse-transcription polymerase chain reaction in duodenal biopsy samples from untreated (n = 15) and diet-treated patients (n = 31) and controls (n = 16). Genetic association of the four SPINK genes was tested within a total of 18 haplotype tagging SNPs, one coding SNP, 310 patients, and 180 controls. The SPINK4 study cohort was further expanded to include 479 CD cases and 540 controls. SPINK4 DNA sequence analysis was performed on six members of a multigeneration CD family to detect possible point mutations or deletions. SPINK4 showed differential gene expression, which was at its highest in untreated patients and dropped sharply upon commencement of a gluten-free diet. Genetic association tests for all four SPINK genes were negative, including SPINK4 in the extended case/control cohort. No SPINK4 mutations or deletions were observed in the multigeneration CD family with linkage to chromosome 9p21-13 nor was the coding SNP disease-specific. SPINK4 exhibits CD pathology-related differential gene expression, likely derived from altered goblet cell activity. All of the four SPINK genes tested do not contribute to the genetic risk for CD in the Dutch population.

Collaboration


Dive into the Martin C. Wapenaar's collaboration.

Top Co-Authors

Avatar

Cisca Wijmenga

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

G.J.B. van Ommen

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lude Franke

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alexandra Zhernakova

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Chris J. Mulder

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

J.T. den Dunnen

Leiden University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge