Network


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

Hotspot


Dive into the research topics where Margit Schraders is active.

Publication


Featured researches published by Margit Schraders.


Science | 2009

SDH5, a Gene Required for Flavination of Succinate Dehydrogenase, Is Mutated in Paraganglioma

Huai Xiang Hao; Oleh Khalimonchuk; Margit Schraders; Noah Dephoure; Jean-Pierre Bayley; H.P.M. Kunst; Peter Devilee; C.W.R.J. Cremers; Joshua D. Schiffman; Brandon G. Bentz; Steven P. Gygi; Dennis R. Winge; H. Kremer; Jared Rutter

Tapping the Mitochondrial Proteome Mitochondria produce the energy that cells need to survive, function, and divide. A growing list of human disorders has been traced to defects in mitochondrial function. About 300 mammalian mitochondrial proteins are functionally uncharacterized, and Hao et al. (p. 1139, published online 23 July) reasoned that the most highly conserved proteins within this group might provide insights into human disease. A combination of bioinformatics, yeast genetics, biochemistry, and human genetics was used to show that a previously uncharacterized mitochondrial protein (Sdh5) is required for the activity of respiratory complex II. Inactivating mutations in the human gene encoding SDH5 were found in individuals with hereditary paraganglioma, a rare neuroendocrine tumor. Thus, analysis of a mitochondrial protein in yeast has revealed a human tumor susceptibility gene. Analysis of a yeast mitochondrial protein reveals a human tumor susceptibility gene. Mammalian mitochondria contain about 1100 proteins, nearly 300 of which are uncharacterized. Given the well-established role of mitochondrial defects in human disease, functional characterization of these proteins may shed new light on disease mechanisms. Starting with yeast as a model system, we investigated an uncharacterized but highly conserved mitochondrial protein (named here Sdh5). Both yeast and human Sdh5 interact with the catalytic subunit of the succinate dehydrogenase (SDH) complex, a component of both the electron transport chain and the tricarboxylic acid cycle. Sdh5 is required for SDH-dependent respiration and for Sdh1 flavination (incorporation of the flavin adenine dinucleotide cofactor). Germline loss-of-function mutations in the human SDH5 gene, located on chromosome 11q13.1, segregate with disease in a family with hereditary paraganglioma, a neuroendocrine tumor previously linked to mutations in genes encoding SDH subunits. Thus, a mitochondrial proteomics analysis in yeast has led to the discovery of a human tumor susceptibility gene.


Nature Genetics | 2012

Mutations in ISPD cause Walker-Warburg syndrome and defective glycosylation of α-dystroglycan.

Tony Roscioli; Erik-Jan Kamsteeg; K Buysse; Isabelle Maystadt; Jeroen van Reeuwijk; Christa van den Elzen; Ellen van Beusekom; Moniek Riemersma; Rolph Pfundt; Lisenka E.L.M. Vissers; Margit Schraders; Umut Altunoglu; Michael Buckley; Han G. Brunner; Bernard Grisart; Huiqing Zhou; Joris A. Veltman; Christian Gilissen; Grazia M.S. Mancini; Paul Delrée; M.A.A.P. Willemsen; Danijela Petković Ramadža; David Chitayat; Christopher L. Bennett; Eamonn Sheridan; Els Peeters; Gita M. B. Tan-Sindhunata; Christine E.M. de Die-Smulders; Koenraad Devriendt; Hülya Kayserili

Walker-Warburg syndrome (WWS) is an autosomal recessive multisystem disorder characterized by complex eye and brain abnormalities with congenital muscular dystrophy (CMD) and aberrant α-dystroglycan glycosylation. Here we report mutations in the ISPD gene (encoding isoprenoid synthase domain containing) as the second most common cause of WWS. Bacterial IspD is a nucleotidyl transferase belonging to a large glycosyltransferase family, but the role of the orthologous protein in chordates is obscure to date, as this phylum does not have the corresponding non-mevalonate isoprenoid biosynthesis pathway. Knockdown of ispd in zebrafish recapitulates the human WWS phenotype with hydrocephalus, reduced eye size, muscle degeneration and hypoglycosylated α-dystroglycan. These results implicate ISPD in α-dystroglycan glycosylation in maintaining sarcolemma integrity in vertebrates.


American Journal of Human Genetics | 2011

Next-generation sequencing identifies mutations of SMPX, which encodes the small muscle protein, X-linked, as a cause of progressive hearing impairment

Margit Schraders; Stefan A. Haas; Nicole J.D. Weegerink; Jaap Oostrik; Hao Hu; Lies H. Hoefsloot; Sriram Kannan; P.L.M. Huygen; R.J.E. Pennings; Ronald J.C. Admiraal; Vera M. Kalscheuer; H.P.M. Kunst; Hannie Kremer

In a Dutch family with an X-linked postlingual progressive hearing impairment, a critical linkage interval was determined to span a region of 12.9 Mb flanked by the markers DXS7108 and DXS7110. This interval overlaps with the previously described DFNX4 locus and contains 75 annotated genes. Subsequent next-generation sequencing (NGS) detected one variant within the linkage interval, a nonsense mutation in SMPX. SMPX encodes the small muscle protein, X-linked (SMPX). Further screening was performed on 26 index patients from small families for which X-linked inheritance of nonsyndromic hearing impairment (NSHI) was not excluded. We detected a frameshift mutation in SMPX in one of the patients. Segregation analysis of both mutations in the families in whom they were found revealed that the mutations cosegregated with hearing impairment. Although we show that SMPX is expressed in many different organs, including the human inner ear, no obvious symptoms other than hearing impairment were observed in the patients. SMPX had previously been demonstrated to be specifically expressed in striated muscle and, therefore, seemed an unlikely candidate gene for hearing impairment. We hypothesize that SMPX functions in inner ear development and/or maintenance in the IGF-1 pathway, the integrin pathway through Rac1, or both.


American Journal of Human Genetics | 2012

Mutations in OTOGL, encoding the inner ear protein otogelin-like, cause moderate sensorineural hearing loss

Kemal O. Yariz; Duygu Duman; Celia Zazo Seco; Julia E. Dallman; Mingqian Huang; Theo A. Peters; Asli Sirmaci; Na Lu; Margit Schraders; Isaac Skromne; Jaap Oostrik; Oscar Diaz-Horta; Juan I. Young; Suna Tokgoz-Yilmaz; Ozlem Konukseven; Hashem Shahin; Lisette Hetterschijt; Moien Kanaan; Anne M.M. Oonk; Yvonne J. K. Edwards; Huawei Li; Semra Atalay; Susan H. Blanton; Alexandra DeSmidt; Xue Zhong Liu; R.J.E. Pennings; Zhongmin Lu; Zheng-Yi Chen; Hannie Kremer; Mustafa Tekin

Hereditary hearing loss is characterized by a high degree of genetic heterogeneity. Here we present OTOGL mutations, a homozygous one base pair deletion (c.1430 delT) causing a frameshift (p.Val477Glufs(∗)25) in a large consanguineous family and two compound heterozygous mutations, c.547C>T (p.Arg183(∗)) and c.5238+5G>A, in a nonconsanguineous family with moderate nonsyndromic sensorineural hearing loss. OTOGL maps to the DFNB84 locus at 12q21.31 and encodes otogelin-like, which has structural similarities to the epithelial-secreted mucin protein family. We demonstrate that Otogl is expressed in the inner ear of vertebrates with a transcription level that is high in embryonic, lower in neonatal, and much lower in adult stages. Otogelin-like is localized to the acellular membranes of the cochlea and the vestibular system and to a variety of inner ear cells located underneath these membranes. Knocking down of otogl with morpholinos in zebrafish leads to sensorineural hearing loss and anatomical changes in the inner ear, supporting that otogelin-like is essential for normal inner ear function. We propose that OTOGL mutations affect the production and/or function of acellular structures of the inner ear, which ultimately leads to sensorineural hearing loss.


Nature Communications | 2011

Gipc3 mutations associated with audiogenic seizures and sensorineural hearing loss in mouse and human

Nikoletta Charizopoulou; Andrea Lelli; Margit Schraders; Kausik Ray; Michael S. Hildebrand; A. Ramesh; C. R. Srikumari Srisailapathy; Jaap Oostrik; Ronald J.C. Admiraal; Harold R. Neely; Joseph R. Latoche; Richard J.H. Smith; John K. Northup; H. Kremer; Jeffrey R. Holt; Konrad Noben-Trauth

Sensorineural hearing loss affects the quality of life and communication of millions of people, but the underlying molecular mechanisms remain elusive. Here, we identify mutations in Gipc3 underlying progressive sensorineural hearing loss (age-related hearing loss 5, ahl5) and audiogenic seizures (juvenile audiogenic monogenic seizure 1, jams1) in mice and autosomal recessive deafness DFNB15 and DFNB95 in humans. Gipc3 localizes to inner ear sensory hair cells and spiral ganglion. A missense mutation in the PDZ domain has an attenuating effect on mechanotransduction and the acquisition of mature inner hair cell potassium currents. Magnitude and temporal progression of wave I amplitude of afferent neurons correlate with susceptibility and resistance to audiogenic seizures. The Gipc3343A allele disrupts the structure of the stereocilia bundle and affects long-term function of auditory hair cells and spiral ganglion neurons. Our study suggests a pivotal role of Gipc3 in acoustic signal acquisition and propagation in cochlear hair cells.


Jaro-journal of The Association for Research in Otolaryngology | 2011

Genotype-Phenotype Correlation in DFNB8/10 Families with TMPRSS3 Mutations

Nicole J. D. Weegerink; Margit Schraders; Jaap Oostrik; P.L.M. Huygen; Tim M. Strom; Susanne Granneman; R.J.E. Pennings; Hanka Venselaar; Lies H. Hoefsloot; Mariet W. Elting; C.W.R.J. Cremers; Ronald J.C. Admiraal; Hannie Kremer; H.P.M. Kunst

In the present study, genotype–phenotype correlations in eight Dutch DFNB8/10 families with compound heterozygous mutations in TMPRSS3 were addressed. We compared the phenotypes of the families by focusing on the mutation data. The compound heterozygous variants in the TMPRSS3 gene in the present families included one novel variant, p.Val199Met, and four previously described pathogenic variants, p.Ala306Thr, p.Thr70fs, p.Ala138Glu, and p.Cys107Xfs. In addition, the p.Ala426Thr variant, which had previously been reported as a possible polymorphism, was found in one family. All affected family members reported progressive bilateral hearing impairment, with variable onset ages and progression rates. In general, the hearing impairment affected the high frequencies first, and sooner or later, depending on the mutation, the low frequencies started to deteriorate, which eventually resulted in a flat audiogram configuration. The ski-slope audiogram configuration is suggestive for the involvement of TMPRSS3. Our data suggest that not only the protein truncating mutation p.T70fs has a severe effect but also the amino acid substitutions p.Ala306Thr and p.Val199Met. A combination of two of these three mutations causes prelingual profound hearing impairment. However, in combination with the p.Ala426Thr or p.Ala138Glu mutations, a milder phenotype with postlingual onset of the hearing impairment is seen. Therefore, the latter mutations are likely to be less detrimental for protein function. Further studies are needed to distinguish possible phenotypic differences between different TMPRSS3 mutations. Evaluation of performance of patients with a cochlear implant indicated that this is a good treatment option for patients with TMPRSS3 mutations as satisfactory speech reception was reached after implantation.


American Journal of Human Genetics | 2010

Mutations in TPRN Cause a Progressive Form of Autosomal-Recessive Nonsyndromic Hearing Loss

Yun Li; Esther Pohl; Redouane Boulouiz; Margit Schraders; Gudrun Nürnberg; Majida Charif; Ronald J.C. Admiraal; Simon von Ameln; Ingelore Baessmann; Mostafa Kandil; Joris A. Veltman; Peter Nürnberg; Christian Kubisch; Abdelhamid Barakat; Hannie Kremer; Bernd Wollnik

We performed genome-wide homozygosity mapping in a large consanguineous family from Morocco and mapped the autosomal-recessive nonsyndromic hearing loss (ARNSHL) in this family to the DFNB79 locus on chromosome 9q34. By sequencing of 62 positional candidate genes of the critical region, we identified a causative homozygous 11 bp deletion, c.42_52del, in the TPRN gene in all seven affected individuals. The deletion is located in exon 1 and results in a frameshift and premature protein truncation (p.Gly15AlafsX150). Interestingly, the deleted sequence is part of a repetitive and CG-rich motive predicted to be prone to structural aberrations during crossover formation. We identified another family with progressive ARNSHL linked to this locus, whose affected members were shown to carry a causative 1 bp deletion (c.1347delG) in exon 1 of TPRN. The function of the encoded protein, taperin, is unknown; yet, partial homology to the actin-caping protein phostensin suggests a role in actin dynamics.


American Journal of Human Genetics | 2010

Homozygosity Mapping Reveals Mutations of GRXCR1 as a Cause of Autosomal-Recessive Nonsyndromic Hearing Impairment

Margit Schraders; Kwanghyuk Lee; Jaap Oostrik; P.L.M. Huygen; Ghazanfar Ali; Lies H. Hoefsloot; Joris A. Veltman; Frans P.M. Cremers; Sulman Basit; Muhammad Ansar; C.W.R.J. Cremers; H.P.M. Kunst; Wasim Ahmad; Ronald J.C. Admiraal; Suzanne M. Leal; Hannie Kremer

We identified overlapping homozygous regions within the DFNB25 locus in two Dutch and ten Pakistani families with sensorineural autosomal-recessive nonsyndromic hearing impairment (arNSHI). Only one of the families, W98-053, was not consanguineous, and its sibship pointed toward a reduced critical region of 0.9 Mb. This region contained the GRXCR1 gene, and the orthologous mouse gene was described to be mutated in the pirouette (pi) mutant with resulting hearing loss and circling behavior. Sequence analysis of the GRXCR1 gene in hearing-impaired family members revealed splice-site mutations in two Dutch families and a missense and nonsense mutation, respectively, in two Pakistani families. The splice-site mutations are predicted to cause frameshifts and premature stop codons. In family W98-053, this could be confirmed by cDNA analysis. GRXCR1 is predicted to contain a GRX-like domain. GRX domains are involved in reversible S-glutathionylation of proteins and thereby in the modulation of activity and/or localization of these proteins. The missense mutation is located in this domain, whereas the nonsense and splice-site mutations may result in complete or partial absence of the GRX-like domain or of the complete protein. Hearing loss in patients with GRXCR1 mutations is congenital and is moderate to profound. Progression of the hearing loss was observed in family W98-053. Vestibular dysfunction was observed in some but not all affected individuals. Quantitative analysis of GRXCR1 transcripts in fetal and adult human tissues revealed a preferential expression of the gene in fetal cochlea, which may explain the nonsyndromic nature of the hearing impairment.


PLOS ONE | 2014

Genetic spectrum of autosomal recessive non-syndromic hearing loss in Pakistani families.

Sobia Shafique; Saima Siddiqi; Margit Schraders; Jaap Oostrik; Humaira Ayub; Ammad Bilal; Muhammad Ajmal; Cecilia Zazo Seco; Tim M. Strom; Atika Mansoor; Kehkashan Mazhar; Syed Tahir A. Shah; Alamdar Hussain; Maleeha Azam; Hannie Kremer; Raheel Qamar

The frequency of inherited bilateral autosomal recessive non-syndromic hearing loss (ARNSHL) in Pakistan is 1.6/1000 individuals. More than 50% of the families carry mutations in GJB2 while mutations in MYO15A account for about 5% of recessive deafness. In the present study a cohort of 30 ARNSHL families was initially screened for mutations in GJB2 and MYO15A. Homozygosity mapping was performed by employing whole genome single nucleotide polymorphism (SNP) genotyping in the families that did not carry mutations in GJB2 or MYO15A. Mutation analysis was performed for the known ARNSHL genes present in the homozygous regions to determine the causative mutations. This allowed the identification of a causative mutation in all the 30 families including 9 novel mutations, which were identified in 9 different families (GJB2 (c.598G>A, p.Gly200Arg); MYO15A (c.9948G>A, p.Gln3316Gln; c.3866+1G>A; c.8767C>T, p.Arg2923* and c.8222T>C, p.Phe2741Ser), TMC1 (c.362+18A>G), BSND (c.97G>C, p.Val33Leu), TMPRSS3 (c.726C>G, p.Cys242Trp) and MSRB3 (c.20T>G, p.Leu7Arg)). Furthermore, 12 recurrent mutations were detected in 21 other families. The 21 identified mutations included 10 (48%) missense changes, 4 (19%) nonsense mutations, 3 (14%) intronic mutations, 2 (9%) splice site mutations and 2 (9%) frameshift mutations. GJB2 accounted for 53% of the families, while mutations in MYO15A were the second most frequent (13%) cause of ARNSHL in these 30 families. The identification of novel as well as recurrent mutations in the present study increases the spectrum of mutations in known deafness genes which could lead to the identification of novel founder mutations and population specific mutated deafness genes causative of ARNSHL. These results provide detailed genetic information that has potential diagnostic implication in the establishment of cost-efficient allele-specific analysis of frequently occurring variants in combination with other reported mutations in Pakistani populations.


British Journal of Haematology | 2008

Integrated genomic and expression profiling in mantle cell lymphoma: identification of gene-dosage regulated candidate genes.

Margit Schraders; Pedro Jares; Sílvia Beà; E.F.P.M. Schoenmakers; Johan H. J. M. van Krieken; Elias Campo; Patricia J. T. A. Groenen

Mantle cell lymphoma (MCL) is characterized by the t(11;14)(q13;q32) translocation and several other cytogenetic aberrations, including heterozygous loss of chromosomal arms 1p, 6q, 11q and 13q and/or gains of 3q and 8q. The common intervals of chromosomal imbalance have been narrowed down using array‐comparative genomic hybridization (CGH). However, the chromosomal intervals still contain many genes potentially involved in MCL pathogeny. Combined analysis of tiling‐resolution array‐CGH with gene expression profiling on 11 MCL tumours enabled the identification of genomic alterations and their corresponding gene expression profiles. Only subsets of genes located within given cytogenetic anomaly‐intervals showed a concomitant change in mRNA expression level. The genes that showed consistent correlation between DNA copy number and RNA expression levels are likely to be important in MCL pathology. Besides several ‘anonymous genes’, we also identified various fully annotated genes, whose gene products are involved in cyclic adenosine monophosphate‐regulated pathways (PRKACB), DNA damage repair, maintenance of chromosome stability and prevention of rereplication (ATM, ERCC5, FBXO5), energy metabolism (such as genes that are involved in the synthesis of proteins encoded by the mitochondrial genome) and signal transduction (ARHGAP29). Deregulation of these gene products may interfere with the signalling pathways that are involved in MCL tumour development and maintenance.

Collaboration


Dive into the Margit Schraders's collaboration.

Top Co-Authors

Avatar

Hannie Kremer

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Jaap Oostrik

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

H.P.M. Kunst

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

R.J.E. Pennings

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lies H. Hoefsloot

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

P.L.M. Huygen

Radboud University Nijmegen Medical Centre

View shared research outputs
Top Co-Authors

Avatar

Celia Zazo Seco

Radboud University Nijmegen Medical Centre

View shared research outputs
Top Co-Authors

Avatar

C.W.R.J. Cremers

Radboud University Nijmegen Medical Centre

View shared research outputs
Top Co-Authors

Avatar

Helger G. Yntema

Radboud University Nijmegen

View shared research outputs
Researchain Logo
Decentralizing Knowledge