Network


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

Hotspot


Dive into the research topics where Gijs W.E. Santen is active.

Publication


Featured researches published by Gijs W.E. Santen.


Nature Genetics | 2012

Digenic inheritance of an SMCHD1 mutation and an FSHD-permissive D4Z4 allele causes facioscapulohumeral muscular dystrophy type 2

Richard J.L.F. Lemmers; Rabi Tawil; Lisa M. Petek; Judit Balog; Gregory J. Block; Gijs W.E. Santen; Amanda M. Amell; Patrick J. van der Vliet; Rowida Almomani; Kirsten R. Straasheijm; Yvonne D. Krom; Rinse Klooster; Yu-chun Sun; Johan T. den Dunnen; Quinta Helmer; Colleen M. Donlin-Smith; George W. Padberg; Baziel G.M. van Engelen; Jessica C. de Greef; Annemieke Aartsma-Rus; Rune R. Frants; Marianne de Visser; Claude Desnuelle; Sabrina Sacconi; Galina N. Filippova; Bert Bakker; Michael J. Bamshad; Stephen J. Tapscott; Daniel G. Miller; Silvère M. van der Maarel

Facioscapulohumeral dystrophy (FSHD) is characterized by chromatin relaxation of the D4Z4 macrosatellite array on chromosome 4 and expression of the D4Z4-encoded DUX4 gene in skeletal muscle. The more common form, autosomal dominant FSHD1, is caused by contraction of the D4Z4 array, whereas the genetic determinants and inheritance of D4Z4 array contraction–independent FSHD2 are unclear. Here, we show that mutations in SMCHD1 (encoding structural maintenance of chromosomes flexible hinge domain containing 1) on chromosome 18 reduce SMCHD1 protein levels and segregate with genome-wide D4Z4 CpG hypomethylation in human kindreds. FSHD2 occurs in individuals who inherited both the SMCHD1 mutation and a normal-sized D4Z4 array on a chromosome 4 haplotype permissive for DUX4 expression. Reducing SMCHD1 levels in skeletal muscle results in D4Z4 contraction–independent DUX4 expression. Our study identifies SMCHD1 as an epigenetic modifier of the D4Z4 metastable epiallele and as a causal genetic determinant of FSHD2 and possibly other human diseases subject to epigenetic regulation.


Nature Genetics | 2012

Mutations in SWI/SNF chromatin remodeling complex gene ARID1B cause Coffin-Siris syndrome

Gijs W.E. Santen; Emmelien Aten; Yu Sun; Rowida Almomani; Christian Gilissen; Maartje Nielsen; Sarina G. Kant; Irina Snoeck; Els Peeters; Yvonne Hilhorst-Hofstee; Marja W. Wessels; Nicolette S. den Hollander; Claudia Ruivenkamp; Gert-Jan B. van Ommen; Martijn H. Breuning; Johan T. den Dunnen; Arie van Haeringen; Marjolein Kriek

We identified de novo truncating mutations in ARID1B in three individuals with Coffin-Siris syndrome (CSS) by exome sequencing. Array-based copy-number variation (CNV) analysis in 2,000 individuals with intellectual disability revealed deletions encompassing ARID1B in 3 subjects with phenotypes partially overlapping that of CSS. Taken together with published data, these results indicate that haploinsufficiency of the ARID1B gene, which encodes an epigenetic modifier of chromatin structure, is an important cause of CSS and is potentially a common cause of intellectual disability and speech impairment.


Nature Genetics | 2014

Loss-of-function mutations in MICU1 cause a brain and muscle disorder linked to primary alterations in mitochondrial calcium signaling

Clare V. Logan; Gyorgy Szabadkai; Jenny A. Sharpe; David A. Parry; Silvia Torelli; Anne-Marie Childs; Marjolein Kriek; Rahul Phadke; Colin A. Johnson; Nicola Roberts; David T. Bonthron; Karen A. Pysden; Tamieka Whyte; Iulia Munteanu; A. Reghan Foley; Gabrielle Wheway; Katarzyna Szymanska; Subaashini Natarajan; Zakia Abdelhamed; J.E. Morgan; Helen Roper; Gijs W.E. Santen; Erik H. Niks; W. Ludo van der Pol; Dick Lindhout; Anna Raffaello; Diego De Stefani; Johan T. den Dunnen; Yu Sun; Ieke B. Ginjaar

Mitochondrial Ca2+ uptake has key roles in cell life and death. Physiological Ca2+ signaling regulates aerobic metabolism, whereas pathological Ca2+ overload triggers cell death. Mitochondrial Ca2+ uptake is mediated by the Ca2+ uniporter complex in the inner mitochondrial membrane, which comprises MCU, a Ca2+-selective ion channel, and its regulator, MICU1. Here we report mutations of MICU1 in individuals with a disease phenotype characterized by proximal myopathy, learning difficulties and a progressive extrapyramidal movement disorder. In fibroblasts from subjects with MICU1 mutations, agonist-induced mitochondrial Ca2+ uptake at low cytosolic Ca2+ concentrations was increased, and cytosolic Ca2+ signals were reduced. Although resting mitochondrial membrane potential was unchanged in MICU1-deficient cells, the mitochondrial network was severely fragmented. Whereas the pathophysiology of muscular dystrophy and the core myopathies involves abnormal mitochondrial Ca2+ handling, the phenotype associated with MICU1 deficiency is caused by a primary defect in mitochondrial Ca2+ signaling, demonstrating the crucial role of mitochondrial Ca2+ uptake in humans.


Clinical Pharmacokinectics | 2009

Morphine glucuronidation in preterm neonates, infants and children younger than 3 years

Catherijne A. J. Knibbe; Elke H. J. Krekels; Johannes N. van den Anker; Joost DeJongh; Gijs W.E. Santen; Monique van Dijk; Sinno Simons; Richard A. van Lingen; Evelyne Jacqz-Aigrain; Meindert Danhof; Dick Tibboel

Background and objectiveA considerable amount of drug use in children is still unlicensed or off-label. In order to derive rational dosing schemes, the influence of aging on glucuronidation capacity in newborns, including preterms, infants and children under the age of 3 years was studied using morphine and its major metabolites as a model drug.MethodsA population pharmacokinetic model was developed with the nonlinear mixed-effects modelling software NONMEM® V, on the basis of 2159 concentrations of morphine and its glucuronides from 248 infants receiving intravenous morphine ranging in bodyweight from 500 g to 18 kg (median 2.8 kg). The model was internally validated using normalized prediction distribution errors.ResultsFormation clearances of morphine to its glucuronides and elimination clearances of the glucuronides were found to be primarily influenced by bodyweight, which was parameterized using an allometric equation with an estimated exponential scaling factor of 1.44. Additionally, a postnatal age of less than 10 days was identified as a covariate for formation clearance to the glucuronides, independent of birthweight or postmenstrual age. Distribution volumes scaled linearly with bodyweight.ConclusionsModel-based simulations show that in newborns, including preterms, infants and children under the age of 3 years, a loading dose in µg/kg and a maintenance dose expressed in µg/kg1.5/h, with a 50% reduction of the maintenance dose in newborns younger than 10 days, results in a narrow range of morphine and metabolite serum concentrations throughout the studied age range. Future pharmacodynamic investigations are needed to reveal target concentrations in this population, after which final dosing recommendations can be made.


Human Mutation | 2013

Coffin-siris syndrome and the BAF complex: Genotype-phenotype study in 63 patients

Gijs W.E. Santen; Emmelien Aten; Anneke T. Vulto-van Silfhout; Caroline Pottinger; Bregje W.M. Bon; Ivonne J.H.M. Minderhout; Ronelle Snowdowne; Christian A.C. Lans; Merel W. Boogaard; Margot M.L. Linssen; Linda Vijfhuizen; Michiel J.R. Wielen; M.J. (Ellen) Vollebregt; Martijn H. Breuning; Marjolein Kriek; Arie van Haeringen; Johan T. den Dunnen; Alexander Hoischen; Jill Clayton-Smith; Bert B.A. Vries; Raoul C. M. Hennekam; Martine J. van Belzen; Mariam Almureikhi; Anwar Baban; Mafalda Barbosa; Tawfeg Ben-Omran; Katherine Berry; Stefania Bigoni; Odile Boute; Louise Brueton

De novo germline variants in several components of the SWI/SNF‐like BAF complex can cause Coffin–Siris syndrome (CSS), Nicolaides–Baraitser syndrome (NCBRS), and nonsyndromic intellectual disability. We screened 63 patients with a clinical diagnosis of CSS for these genes (ARID1A, ARID1B, SMARCA2, SMARCA4, SMARCB1, and SMARCE1) and identified pathogenic variants in 45 (71%) patients. We found a high proportion of variants in ARID1B (68%). All four pathogenic variants in ARID1A appeared to be mosaic. By using all variants from the Exome Variant Server as test data, we were able to classify variants in ARID1A, ARID1B, and SMARCB1 reliably as being pathogenic or nonpathogenic. For SMARCA2, SMARCA4, and SMARCE1 several variants in the EVS remained unclassified, underlining the importance of parental testing. We have entered all variant and clinical information in LOVD‐powered databases to facilitate further genotype–phenotype correlations, as these will become increasingly important because of the uptake of targeted and untargeted next generation sequencing in diagnostics. The emerging phenotype–genotype correlation is that SMARCB1 patients have the most marked physical phenotype and severe cognitive and growth delay. The variability in phenotype seems most marked in ARID1A and ARID1B patients. Distal limbs anomalies are most marked in ARID1A patients and least in SMARCB1 patients. Numbers are small however, and larger series are needed to confirm this correlation.


Nature Genetics | 2017

Targeted sequencing identifies 91 neurodevelopmental-disorder risk genes with autism and developmental-disability biases

Holly A.F. Stessman; Bo Xiong; Bradley P. Coe; Tianyun Wang; Kendra Hoekzema; Michaela Fenckova; Malin Kvarnung; Jennifer Gerdts; Sandy Trinh; Nele Cosemans; Laura Vives; Janice Lin; Tychele N. Turner; Gijs W.E. Santen; Claudia Ruivenkamp; Marjolein Kriek; Arie van Haeringen; Emmelien Aten; Kathryn Friend; Jan Liebelt; Christopher Barnett; Eric Haan; Marie Shaw; Jozef Gecz; Britt Marie Anderlid; Ann Nordgren; Anna Lindstrand; Charles E. Schwartz; R. Frank Kooy; Geert Vandeweyer

Gene-disruptive mutations contribute to the biology of neurodevelopmental disorders (NDDs), but most of the related pathogenic genes are not known. We sequenced 208 candidate genes from >11,730 cases and >2,867 controls. We identified 91 genes, including 38 new NDD genes, with an excess of de novo mutations or private disruptive mutations in 5.7% of cases. Drosophila functional assays revealed a subset with increased involvement in NDDs. We identified 25 genes showing a bias for autism versus intellectual disability and highlighted a network associated with high-functioning autism (full-scale IQ >100). Clinical follow-up for NAA15, KMT5B, and ASH1L highlighted new syndromic and nonsyndromic forms of disease.


Human Mutation | 2015

Next-Generation Diagnostics: Gene Panel, Exome, or Whole Genome?

Yu Sun; Claudia Ruivenkamp; Mariëtte J.V. Hoffer; Terry Vrijenhoek; Marjolein Kriek; Christi J. van Asperen; Johan T. den Dunnen; Gijs W.E. Santen

Although the benefits of next‐generation sequencing (NGS) for the diagnosis of heterogeneous diseases such as intellectual disability (ID) are undisputed, there is little consensus on the relative merits of targeted enrichment, whole‐exome sequencing (WES) or whole‐genome sequencing (WGS). To answer this question, WES and WGS data from the same nine samples were compared, and WES was shown not to miss any variants identified by WGS in a gene panel including ∼500 genes linked to ID (500GP). Additionally, deeply sequenced WES data were shown to adequately cover ∼99% of the 500GP; thus, little additional benefit was to be expected from a targeted enrichment approach. To reduce costs, minimal sequencing criteria were determined by investigating the relation between sequenced reads and outcome parameters such as coverage and variant yield. Our analysis indicated that 60 million reads yielded a mean coverage of ∼60×: ∼97% of the 500GP sequences were sufficiently covered to exclude variants, whereas variant yield was ∼99.5% and false‐positive and false‐negative rates were controlled. Our findings indicate that WES is currently the optimal approach to ID diagnostics. This result depends on the capture kit and sequencing strategy used. The developed framework however is amenable to other sequencing approaches.


Journal of Psychiatric Research | 2008

Sensitivity of the individual items of the Hamilton depression rating scale to response and its consequences for the assessment of efficacy

Gijs W.E. Santen; Roberto Gomeni; Meindert Danhof; Oscar Della Pasqua

The Hamilton depression rating scale (HAM-D(17)) has been the gold standard in depression trials since its introduction in 1960 by Max Hamilton. However, several authors have shown that the HAM-D(17) is multi-dimensional and that subscales of the HAM-D(17) outperform the total scale. In the current study, we assess the sensitivity of the individual HAM-D(17) items in differentiating responders from non-responders over the typical treatment period used in clinical efficacy trials. Based on data from randomised, placebo-controlled trials with paroxetine, a graphical analysis and a statistical analysis were performed to identify the items that are most sensitive to the rate and extent of response irrespective of treatment. From these analyses, two subscales consisting of seven items each were derived and compared to the Bech and Maier and Philip subscales using a linear mixed-effects modelling approach for repeated measures. The evaluation of two clinical trials revealed endpoint sensitivity comparable to the existing subscales. Using a bootstrap technique, we show that the subscales consistently yield higher statistical power compared to the HAM-D(17), although no subscale consistently outperforms the others. In conclusion, this study provides further evidence that not all items of the HAM-D(17) scale are equally sensitive to detect responding patients in a clinical trial. A HAM-D(7) subscale with higher sensitivity to drug effect is proposed consisting of the HAM-D(6) and the suicide item. This response-based subscale increases signal-to-noise ratio and could reduce failure rate in efficacy trials with antidepressant drugs.


European Journal of Human Genetics | 2015

Next-generation sequencing-based genome diagnostics across clinical genetics centers: implementation choices and their effects

Terry Vrijenhoek; Ken Kraaijeveld; Martin Elferink; Joep de Ligt; Elcke Kranendonk; Gijs W.E. Santen; Isaac J. Nijman; Derek Butler; Godelieve Claes; Adalberto Costessi; Wim Dorlijn; Winfried van Eyndhoven; Dicky Halley; Mirjam C. G. N. van den Hout; Steven van Hove; Lennart F. Johansson; Jan D. H. Jongbloed; R. Kamps; Christel Kockx; Bart de Koning; Marjolein Kriek; Ronald Lekanne Dit Deprez; Hans Lunstroo; Marcel Mannens; Olaf R. Mook; Marcel R. Nelen; Corrette Ploem; Marco Rijnen; Jasper J. Saris; Richard J. Sinke

Implementation of next-generation DNA sequencing (NGS) technology into routine diagnostic genome care requires strategic choices. Instead of theoretical discussions on the consequences of such choices, we compared NGS-based diagnostic practices in eight clinical genetic centers in the Netherlands, based on genetic testing of nine pre-selected patients with cardiomyopathy. We highlight critical implementation choices, including the specific contributions of laboratory and medical specialists, bioinformaticians and researchers to diagnostic genome care, and how these affect interpretation and reporting of variants. Reported pathogenic mutations were consistent for all but one patient. Of the two centers that were inconsistent in their diagnosis, one reported to have found ‘no causal variant’, thereby underdiagnosing this patient. The other provided an alternative diagnosis, identifying another variant as causal than the other centers. Ethical and legal analysis showed that informed consent procedures in all centers were generally adequate for diagnostic NGS applications that target a limited set of genes, but not for exome- and genome-based diagnosis. We propose changes to further improve and align these procedures, taking into account the blurring boundary between diagnostics and research, and specific counseling options for exome- and genome-based diagnostics. We conclude that alternative diagnoses may infer a certain level of ‘greediness’ to come to a positive diagnosis in interpreting sequencing results. Moreover, there is an increasing interdependence of clinic, diagnostics and research departments for comprehensive diagnostic genome care. Therefore, we invite clinical geneticists, physicians, researchers, bioinformatics experts and patients to reconsider their role and position in future diagnostic genome care.


The Journal of Clinical Endocrinology and Metabolism | 2013

An activating mutation in the kinase homology domain of the natriuretic peptide receptor-2 causes extremely tall stature without skeletal deformities

Sabine Hannema; Hermine A. van Duyvenvoorde; Thomas Premsler; Ruey-Bing Yang; Thomas D. Mueller; Birgit Gassner; Heike Oberwinkler; Ferdinand Roelfsema; Gijs W.E. Santen; Timothy C. R. Prickett; Sarina G. Kant; Annemieke J. M. H. Verkerk; André G. Uitterlinden; Eric A. Espiner; Claudia Ruivenkamp; Wilma Oostdijk; Alberto M. Pereira; Monique Losekoot; Michaela Kuhn; J.M. Wit

BACKGROUND C-type natriuretic peptide (CNP)/natriuretic peptide receptor 2 (NPR2) signaling is essential for long bone growth. Enhanced CNP production caused by chromosomal translocations results in tall stature, a Marfanoid phenotype, and skeletal abnormalities. A similar phenotype was described in a family with an activating NPR2 mutation within the guanylyl cyclase domain. CASE Here we describe an extremely tall male without skeletal deformities, with a novel NPR2 mutation (p.Arg655Cys) located in the kinase homology domain. OBJECTIVES The objective of the study was to investigate the functional and structural effects of the NPR2 mutation. METHODS Guanylyl cyclase activities of wild-type vs mutant NPR2 were analyzed in transfected human embryonic kidney 293 cells and in skin fibroblasts. The former were also used to study possible interactions between both isoforms. Homology modeling was performed to understand the molecular impact of the mutation. RESULTS CNP-stimulated cGMP production by the mutant NPR2 was markedly increased in patient skin fibroblasts and transfected human embryonic kidney 293 cells. The stimulatory effects of ATP on CNP-dependent guanylyl cyclase activity were augmented, suggesting that this novel mutation enhances both the responsiveness of NPR2 to CNP and its allosteric modulation/stabilization by ATP. Coimmunoprecipitation showed that wild-type and mutant NPR2 can form stable heterodimers, suggesting a dominant-positive effect. In accordance with augmented endogenous receptor activity, plasma N-terminal pro-CNP (a marker of CNP production in tissues) was reduced in the proband. CONCLUSIONS We report the first activating mutation within the kinase homology domain of NPR2, resulting in extremely tall stature. Our observations emphasize the important role of this domain in the regulation of guanylyl cyclase activity and bone growth in response to CNP.

Collaboration


Dive into the Gijs W.E. Santen's collaboration.

Top Co-Authors

Avatar

Claudia Ruivenkamp

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emmelien Aten

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Arie van Haeringen

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Johan T. den Dunnen

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Mariëtte J.V. Hoffer

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Martijn H. Breuning

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Monique Losekoot

Leiden University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge