Network


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

Hotspot


Dive into the research topics where Christel Kockx is active.

Publication


Featured researches published by Christel Kockx.


Genes & Development | 2010

The genome-wide dynamics of the binding of Ldb1 complexes during erythroid differentiation

Eric Soler; Charlotte Andrieu-Soler; Ernie de Boer; Jan Christian Bryne; Supat Thongjuea; Ralph Stadhouders; Robert-Jan Palstra; Mary Stevens; Christel Kockx; Wilfred van IJcken; Jun Hou; Christine Steinhoff; Erikjan Rijkers; Boris Lenhard; Frank Grosveld

One of the complexes formed by the hematopoietic transcription factor Gata1 is a complex with the Ldb1 (LIM domain-binding protein 1) and Tal1 proteins. It is known to be important for the development and differentiation of the erythroid cell lineage and is thought to be implicated in long-range interactions. Here, the dynamics of the composition of the complex-in particular, the binding of the negative regulators Eto2 and Mtgr1-are studied, in the context of their genome-wide targets. This shows that the complex acts almost exclusively as an activator, binding a very specific combination of sequences, with a positioning relative to transcription start site, depending on the type of the core promoter. The activation is accompanied by a net decrease in the relative binding of Eto2 and Mtgr1. A Chromosome Conformation Capture sequencing (3C-seq) assay also shows that the binding of the Ldb1 complex marks genomic interaction sites in vivo. This establishes the Ldb1 complex as a positive regulator of the final steps of erythroid differentiation that acts through the shedding of negative regulators and the active interaction between regulatory sequences.


Nature Genetics | 2011

Sox2 cooperates with Chd7 to regulate genes that are mutated in human syndromes

Erik Engelen; Umut Akinci; Jan Christian Bryne; Jun Hou; Cristina Gontan; Maaike Moen; Dorota Szumska; Christel Kockx; Wilfred van IJcken; Dick H. W. Dekkers; Jeroen Demmers; Erikjan Rijkers; Shoumo Bhattacharya; Sjaak Philipsen; Larysa Pevny; Frank Grosveld; Robbert J. Rottier; Boris Lenhard; Raymond A. Poot

The HMG-box transcription factor Sox2 plays a role throughout neurogenesis and also acts at other stages of development, as illustrated by the multiple organs affected in the anophthalmia syndrome caused by SOX2 mutations. Here we combined proteomic and genomic approaches to characterize gene regulation by Sox2 in neural stem cells. Chd7, a chromatin remodeling ATPase associated with CHARGE syndrome, was identified as a Sox2 transcriptional cofactor. Sox2 and Chd7 physically interact, have overlapping genome-wide binding sites and regulate a set of common target genes including Jag1, Gli3 and Mycn, genes mutated in Alagille, Pallister-Hall and Feingold syndromes, which show malformations also associated with SOX2 anophthalmia syndrome or CHARGE syndrome. Regulation of disease-associated genes by a Sox2-Chd7 complex provides a plausible explanation for several malformations associated with SOX2 anophthalmia syndrome or CHARGE syndrome. Indeed, we found that Chd7-haploinsufficient embryos showed severely reduced expression of Jag1 in the developing inner ear.


The EMBO Journal | 2012

Dynamic long-range chromatin interactions control Myb proto-oncogene transcription during erythroid development

Ralph Stadhouders; Supat Thongjuea; Charlotte Andrieu-Soler; Robert-Jan Palstra; Jan Christian Bryne; Anita van den Heuvel; Mary Stevens; Ernie de Boer; Christel Kockx; Antoine van der Sloot; Mirjam C. G. N. van den Hout; Wilfred van IJcken; Dirk Eick; Boris Lenhard; Frank Grosveld; Eric Soler

The key haematopoietic regulator Myb is essential for coordinating proliferation and differentiation. ChIP‐Sequencing and Chromosome Conformation Capture (3C)‐Sequencing were used to characterize the structural and protein‐binding dynamics of the Myb locus during erythroid differentiation. In proliferating cells expressing Myb, enhancers within the Myb‐Hbs1l intergenic region were shown to form an active chromatin hub (ACH) containing the Myb promoter and first intron. This first intron was found to harbour the transition site from transcription initiation to elongation, which takes place around a conserved CTCF site. Upon erythroid differentiation, Myb expression is downregulated and the ACH destabilized. We propose a model for Myb activation by distal enhancers dynamically bound by KLF1 and the GATA1/TAL1/LDB1 complex, which primarily function as a transcription elongation element through chromatin looping.


Molecular Cell | 2012

Snail Regulates MyoD Binding-Site Occupancy to Direct Enhancer Switching and Differentiation-Specific Transcription in Myogenesis

Vahab D. Soleimani; Hang Yin; Arezu Jahani-Asl; Hong Ming; Christel Kockx; Wilfred van IJcken; Frank Grosveld; Michael A. Rudnicki

In skeletal myogenesis, the transcription factor MyoD activates distinct transcriptional programs in progenitors compared to terminally differentiated cells. Using ChIP-Seq and gene expression analyses, we show that in primary myoblasts, Snail-HDAC1/2 repressive complex binds and excludes MyoD from its targets. Notably, Snail binds E box motifs that are G/C rich in their central dinucleotides, and such sites are almost exclusively associated with genes expressed during differentiation. By contrast, Snail does not bind the A/T-rich E boxes associated with MyoD targets in myoblasts. Thus, Snai1-HDAC1/2 prevent MyoD occupancy on differentiation-specific regulatory elements, and the change from Snail to MyoD binding often results in enhancer switching during differentiation. Furthermore, we show that a regulatory network involving myogenic regulatory factors (MRFs), Snai1/2, miR-30a, and miR-206 acts as a molecular switch that controls entry into myogenic differentiation. Together, these results reveal a regulatory paradigm that directs distinct gene expression programs in progenitors versus terminally differentiated cells.


Developmental Cell | 2012

Transcriptional Dominance of Pax7 in Adult Myogenesis Is Due to High-Affinity Recognition of Homeodomain Motifs

Vahab D. Soleimani; Vincent G. Punch; Yoh-ichi Kawabe; Andrew E. Jones; Gareth A. Palidwor; Christopher J. Porter; Joe W. Cross; Jaime J. Carvajal; Christel Kockx; Wilfred van IJcken; Theodore J. Perkins; Peter W.J. Rigby; Frank Grosveld; Michael A. Rudnicki

Pax3 and Pax7 regulate stem cell function in skeletal myogenesis. However, molecular insight into their distinct roles has remained elusive. Using gene expression data combined with genome-wide binding-site analysis, we show that both Pax3 and Pax7 bind identical DNA motifs and jointly activate a large panel of genes involved in muscle stem cell function. Surprisingly, in adult myoblasts Pax3 binds a subset (6.4%) of Pax7 targets. Despite a significant overlap in their transcriptional network, Pax7 regulates distinct panels of genes involved in the promotion of proliferation and inhibition of myogenic differentiation. We show that Pax7 has a higher binding affinity to the homeodomain-binding motif relative to Pax3, suggesting that intrinsic differences in DNA binding contribute to the observed functional difference between Pax3 and Pax7 binding in myogenesis. Together, our data demonstrate distinct attributes of Pax7 function and provide mechanistic insight into the nonredundancy of Pax3 and Pax7 in muscle development.


Modern Pathology | 2014

Clinical significance of immunohistochemistry for detection of BAP1 mutations in uveal melanoma.

Anna E. Koopmans; Robert M. Verdijk; Rutger W. W. Brouwer; Thierry van den Bosch; Mike M P van den Berg; Jolanda Vaarwater; Christel Kockx; Dion Paridaens; Nicole C. Naus; Mark Nellist; Wilfred van IJcken; Emine Kilic; Annelies de Klein

Uveal melanoma is a lethal cancer with a strong propensity to metastasize. Limited therapeutic options are available once the disease has disseminated. A strong predictor for metastasis is the loss of chromosome 3. Inactivating mutations in BAP1 encoding the BRCA1-associated protein 1 and located on chromosome 3p21.1, have been described in uveal melanoma and other types of cancer. In this study, we determined the prevalence of somatic BAP1 mutations and examined whether these mutations correlate with the functional expression of BAP1 in uveal melanoma tissue and with other clinical, histopathological and chromosomal parameters. We screened a cohort of 74 uveal melanomas for BAP1 mutations, using different deep sequencing methods. The frequency of BAP1 mutations in our study group was 47%. The expression of BAP1 protein was studied using immunohistochemistry. BAP1 staining was absent in 43% of the cases. BAP1 mutation status was strongly associated with BAP1 protein expression (P<0.001), loss of chromosome 3 (P<0.001), and other aggressive prognostic factors. Patients with a BAP1 mutation and absent BAP1 expression had an almost eightfold higher chance of developing metastases compared with those without these changes (P=0.002). We found a strong correlation between the immunohistochemical and sequencing data and therefore propose that, immunohistochemical screening for BAP1 should become routine in the histopathological work-up of uveal melanoma. Furthermore, our analysis indicates that loss of BAP1 may be particularly involved in the progression of uveal melanoma to an aggressive, metastatic phenotype.


Epigenetics & Chromatin | 2014

Targeted Chromatin Capture (T2C): a novel high resolution high throughput method to detect genomic interactions and regulatory elements

Petros Kolovos; Harmen J.G. van de Werken; Nick Kepper; Jessica Zuin; Rutger W. W. Brouwer; Christel Kockx; Kerstin S. Wendt; Wilfred van IJcken; Frank Grosveld; Tobias A. Knoch

BackgroundSignificant efforts have recently been put into the investigation of the spatial organization and the chromatin-interaction networks of genomes. Chromosome conformation capture (3C) technology and its derivatives are important tools used in this effort. However, many of these have limitations, such as being limited to one viewpoint, expensive with moderate to low resolution, and/or requiring a large sequencing effort. Techniques like Hi-C provide a genome-wide analysis. However, it requires massive sequencing effort with considerable costs. Here we describe a new technique termed Targeted Chromatin Capture (T2C), to interrogate large selected regions of the genome. T2C provides an unbiased view of the spatial organization of selected loci at superior resolution (single restriction fragment resolution, from 2 to 6 kbp) at much lower costs than Hi-C due to the lower sequencing effort.ResultsWe applied T2C on well-known model regions, the mouse β-globin locus and the human H19/IGF2 locus. In both cases we identified all known chromatin interactions. Furthermore, we compared the human H19/IGF2 locus data obtained from different chromatin conformation capturing methods with T2C data. We observed the same compartmentalization of the locus, but at a much higher resolution (single restriction fragments vs. the common 40 kbp bins) and higher coverage. Moreover, we compared the β-globin locus in two different biological samples (mouse primary erythroid cells and mouse fetal brain), where it is either actively transcribed or not, to identify possible transcriptional dependent interactions. We identified the known interactions in the β-globin locus and the same topological domains in both mouse primary erythroid cells and in mouse fetal brain with the latter having fewer interactions probably due to the inactivity of the locus. Furthermore, we show that interactions due to the important chromatin proteins, Ldb1 and Ctcf, in both tissues can be analyzed easily to reveal their role on transcriptional interactions and genome folding.ConclusionsT2C is an efficient, easy, and affordable with high (restriction fragment) resolution tool to address both genome compartmentalization and chromatin-interaction networks for specific genomic regions at high resolution for both clinical and non-clinical research.


Genome Research | 2013

Dynamic regulation of the transcription initiation landscape at single nucleotide resolution during vertebrate embryogenesis

Chirag Nepal; Yavor Hadzhiev; Christopher Previti; Vanja Haberle; Nan Li; Hazuki Takahashi; Ana Maria Suzuki; Ying Sheng; Rehab F. Abdelhamid; Santosh Anand; Jochen Gehrig; Altuna Akalin; Christel Kockx; Antoine van der Sloot; Wilfred van IJcken; Olivier Armant; Sepand Rastegar; Craig A. Watson; Uwe Strähle; Elia Stupka; Piero Carninci; Boris Lenhard; Ferenc Müller

Spatiotemporal control of gene expression is central to animal development. Core promoters represent a previously unanticipated regulatory level by interacting with cis-regulatory elements and transcription initiation in different physiological and developmental contexts. Here, we provide a first and comprehensive description of the core promoter repertoire and its dynamic use during the development of a vertebrate embryo. By using cap analysis of gene expression (CAGE), we mapped transcription initiation events at single nucleotide resolution across 12 stages of zebrafish development. These CAGE-based transcriptome maps reveal genome-wide rules of core promoter usage, structure, and dynamics, key to understanding the control of gene regulation during vertebrate ontogeny. They revealed the existence of multiple classes of pervasive intra- and intergenic post-transcriptionally processed RNA products and their developmental dynamics. Among these RNAs, we report splice donor site-associated intronic RNA (sRNA) to be specific to genes of the splicing machinery. For the identification of conserved features, we compared the zebrafish data sets to the first CAGE promoter map of Tetraodon and the existing human CAGE data. We show that a number of features, such as promoter type, newly discovered promoter properties such as a specialized purine-rich initiator motif, as well as sRNAs and the genes in which they are detected, are conserved in mammalian and Tetraodon CAGE-defined promoter maps. The zebrafish developmental promoterome represents a powerful resource for studying developmental gene regulation and revealing promoter features shared across vertebrates.


Epigenetics & Chromatin | 2012

Genome-wide DNA methylation profiling of non-small cell lung carcinomas

Rejane Hughes Carvalho; Vanja Haberle; Jun Hou; Teus van Gent; Supat Thongjuea; Wilfred van IJcken; Christel Kockx; Rutger W. W. Brouwer; Erikjan Rijkers; Anieta M. Sieuwerts; John A. Foekens; Mirjam van Vroonhoven; Joachim Aerts; Frank Grosveld; Boris Lenhard; Sjaak Philipsen

BackgroundNon-small cell lung carcinoma (NSCLC) is a complex malignancy that owing to its heterogeneity and poor prognosis poses many challenges to diagnosis, prognosis and patient treatment. DNA methylation is an important mechanism of epigenetic regulation involved in normal development and cancer. It is a very stable and specific modification and therefore in principle a very suitable marker for epigenetic phenotyping of tumors. Here we present a genome-wide DNA methylation analysis of NSCLC samples and paired lung tissues, where we combine MethylCap and next generation sequencing (MethylCap-seq) to provide comprehensive DNA methylation maps of the tumor and paired lung samples. The MethylCap-seq data were validated by bisulfite sequencing and methyl-specific polymerase chain reaction of selected regions.ResultsAnalysis of the MethylCap-seq data revealed a strong positive correlation between replicate experiments and between paired tumor/lung samples. We identified 57 differentially methylated regions (DMRs) present in all NSCLC tumors analyzed by MethylCap-seq. While hypomethylated DMRs did not correlate to any particular functional category of genes, the hypermethylated DMRs were strongly associated with genes encoding transcriptional regulators. Furthermore, subtelomeric regions and satellite repeats were hypomethylated in the NSCLC samples. We also identified DMRs that were specific to two of the major subtypes of NSCLC, adenocarcinomas and squamous cell carcinomas.ConclusionsCollectively, we provide a resource containing genome-wide DNA methylation maps of NSCLC and their paired lung tissues, and comprehensive lists of known and novel DMRs and associated genes in NSCLC.


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.

Collaboration


Dive into the Christel Kockx's collaboration.

Top Co-Authors

Avatar

Wilfred van IJcken

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Frank Grosveld

Erasmus University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Rutger W. W. Brouwer

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Petros Kolovos

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kerstin S. Wendt

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Jessica Zuin

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge