Network


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

Hotspot


Dive into the research topics where Cor Breukel is active.

Publication


Featured researches published by Cor Breukel.


Nature Cell Biology | 2001

Mutations in the APC tumour suppressor gene cause chromosomal instability

Riccardo Fodde; Jeroen Kuipers; Carla Rosenberg; Ron Smits; Menno F. Kielman; Claudia Gaspar; Johan H. van Es; Cor Breukel; Joop Wiegant; Rachel H. Giles; Hans Clevers

Two forms of genetic instability have been described in colorectal cancer: microsatellite instability and chromosomal instability. Microsatellite instability results from mutations in mismatch repair genes; chromosomal instability is the hallmark of many colorectal cancers, although it is not completely understood at the molecular level. As truncations of the Adenomatous Polyposis Coli (APC) gene are found in most colorectal tumours, we thought that mutations in APC might be responsible for chromosomal instability. To test this hypothesis, we examined mouse embryonic stem (ES) cells homozygous for Min (multiple intestinal neoplasia) or Apc1638T alleles. Here we show that Apc mutant ES cells display extensive chromosome and spindle aberrations, providing genetic evidence for a role of APC in chromosome segregation. Consistent with this, APC accumulates at the kinetochore during mitosis. Apc mutant cells form mitotic spindles with an abundance of microtubules that inefficiently connect with kinetochores. This phenotype is recapitulated by the induced expression of a 253-amino-acid carboxy-terminal fragment of APC in microsatellite unstable colorectal cancer cells. We conclude that loss of APC sequences that lie C-terminal to the β-catenin regulatory domain contributes to chromosomal instability in colorectal cancer.


Nature Genetics | 2002

Apc modulates embryonic stem-cell differentiation by controlling the dosage of β-catenin signaling

Menno F. Kielman; Maaret Rindapää; Claudia Gaspar; Nicole van Poppel; Cor Breukel; Sandra van Leeuwen; Makoto M. Taketo; Scott Roberts; Ron Smits; Riccardo Fodde

The Wnt signal-transduction pathway induces the nuclear translocation of membrane-bound β-catenin (Catnb) and has a key role in cell-fate determination. Tight somatic regulation of this signal is essential, as uncontrolled nuclear accumulation of β-catenin can cause developmental defects and tumorigenesis in the adult organism. The adenomatous polyposis coli gene (APC) is a major controller of the Wnt pathway and is essential to prevent tumorigenesis in a variety of tissues and organs. Here, we have investigated the effect of different mutations in Apc on the differentiation potential of mouse embryonic stem (ES) cells. We provide genetic and molecular evidence that the ability and sensitivity of ES cells to differentiate into the three germ layers is inhibited by increased doses of β-catenin by specific Apc mutations. These range from a severe differentiation blockade in Apc alleles completely deficient in β-catenin regulation to more specific neuroectodermal, dorsal mesodermal and endodermal defects in more hypomorphic alleles. Accordingly, a targeted oncogenic mutation in Catnb also affects the differentiation potential of ES cells. Expression profiling of wildtype and Apc-mutated teratomas supports the differentiation defects at the molecular level and pinpoints a large number of downstream structural and regulating genes. Chimeric experiments showed that this effect is cell-autonomous. Our results imply that constitutive activation of the Apc/β-catenin signaling pathway results in differentiation defects in tissue homeostasis, and possibly underlies tumorigenesis in the colon and other self-renewing tissues.


American Journal of Human Genetics | 2003

Molecular analysis of hereditary nonpolyposis colorectal cancer in the United States: High mutation detection rate among clinically selected families and characterization of an American founder genomic deletion of the MSH2 gene

Anja Wagner; Alicia Barrows; Juul T. Wijnen; Heleen M. van der Klift; Patrick Franken; Paul Verkuijlen; Hidewaki Nakagawa; Marjan Geugien; Shantie Jaghmohan-Changur; Cor Breukel; Hanne Meijers-Heijboer; Hans Morreau; Marjo van Puijenbroek; John Burn; Stephany Coronel; Yulia Kinarski; Ross A. Okimoto; Patrice Watson; Jane F. Lynch; Albert de la Chapelle; Henry T. Lynch; Riccardo Fodde

The identification of germline mutations in families with HNPCC is hampered by genetic heterogeneity and clinical variability. In previous studies, MSH2 and MLH1 mutations were found in approximately two-thirds of the Amsterdam-criteria-positive families and in much lower percentages of the Amsterdam-criteria-negative families. Therefore, a considerable proportion of HNPCC seems not to be accounted for by the major mismatch repair (MMR) genes. Does the latter result from a lack of sensitivity of mutation detection techniques, or do additional genes underlie the remaining cases? In this study we address these questions by thoroughly investigating a cohort of clinically selected North American families with HNPCC. We analyzed 59 clinically well-defined U.S. families with HNPCC for MSH2, MLH1, and MSH6 mutations. To maximize mutation detection, different techniques were employed, including denaturing gradient gel electrophoresis, Southern analysis, microsatellite instability, immunohistochemistry, and monoallelic expression analysis. In 45 (92%) of the 49 Amsterdam-criteria-positive families and in 7 (70%) of the 10 Amsterdam-criteria-negative families, a mutation was detected in one of the three analyzed MMR genes. Forty-nine mutations were in MSH2 or MLH1, and only three were in MSH6. A considerable proportion (27%) of the mutations were genomic rearrangements (12 in MSH2 and 2 in MLH1). Notably, a deletion encompassing exons 1-6 of MSH2 was detected in seven apparently unrelated families (12% of the total cohort) and was subsequently proven to be a founder. Screening of a second U.S. cohort with HNPCC from Ohio allowed the identification of two additional kindreds with the identical founder deletion. In the present study, we show that optimal mutation detection in HNPCC is achieved by combining accurate and expert clinical selection with an extensive mutation detection strategy. Notably, we identified a common North American deletion in MSH2, accounting for approximately 10% of our cohort. Genealogical, molecular, and haplotype studies showed that this deletion represents a North American founder mutation that could be traced back to the 19th century.


Human Genetics | 1996

Germline mutations in the 3′ part of APC exon 15 do not result in truncated proteins and are associated with attenuated adenomatous polyposis coli

R. B. van der Luijt; P. Meera Khan; Hans F. A. Vasen; Cor Breukel; Carli M. J. Tops; Rodney J. Scott; Riccardo Fodde

Abstract Familial adenomatous polyposis (FAP) is an inherited predisposition to colorectal cancer characterized by the development of numerous adenomatous polyps predominantly in the colorectal region. Germline mutations in the adenomatous polyposis coli (APC) gene are responsible for most cases of FAP. Mutations at the 5′ end of APC are known to be associated with a relatively mild form of the disease, called attenuated adenomatous polyposis coli (AAPC). We identified a frameshift mutation in the 3′ part of exon 15, resulting in a stop codon at 1862, in a large Dutch kindred with AAPC. Western blot analysis of lymphoblastoid cell lines derived from affected family members from this kindred, as well as from a previously reported Swiss family carrying a frameshift mutation at codon 1987 and displaying a similar attenuated phenotype, showed only the wild-type APC protein. Our study indicates that chain-terminating mutations located in the 3′ part of APC do not result in detectable truncated polypeptides and we hypothesize that this is likely to be the basis for the observed AAPC phenotype.


Gastroenterology | 1998

Apc1638N: A mouse model for familial adenomatous polyposis–associated desmoid tumors and cutaneous cysts☆☆☆

Ron Smits; Willemien van der Houven van Oordt; Arne Luz; Chris Zurcher; Shantie Jagmohan-Changur; Cor Breukel; P. Meera Khan; Riccardo Fodde

BACKGROUND & AIMS Germline mutations in the adenomatous polyposis coli (APC) gene are responsible for familial adenomatous polyposis (FAP), an autosomal dominant predisposition to the formation of multiple colorectal adenomas. Moreover, patients with FAP are at high risk of developing several extracolonic manifestations, including desmoids, cutaneous cysts, and tumors of the upper gastrointestinal tract. Although by definition desmoids are nonmalignant, because of their aggressive invasion of local structures, they represent one of the major causes of morbidity and mortality among patients with FAP. METHODS This study describes the histopathologic and molecular characterization of Apc1638N, a mouse model for the broad spectrum of extracolonic manifestations characteristic of FAP. RESULTS Heterozygous Apc+/Apc1638N animals develop fully penetrant and multifocal cutaneous follicular cysts and desmoid tumors in addition to attenuated polyposis of the upper gastrointestinal tract. Moreover, breeding of Apc+/Apc1638N mice in a p53-deficient background results in a dramatic seven-fold increase of the desmoid multiplicity. CONCLUSIONS Because of the attenuated nature of their intestinal phenotype, these mice survive longer than other murine models for Apc-driven tumorigenesis. Therefore, Apc1638N represents an ideal laboratory tool to test various therapeutic intervention strategies for the management of intestinal as well as extraintestinal tumors.


Human Genetics | 1995

APC mutation in the alternatively spliced region of exon 9 associated with late onset familial adenomatous polyposis

Rob B. van der Luijt; Hans F. A. Vasen; Carli M. J. Tops; Cor Breukel; Riccardo Fodde; P. Meera Khan

Germ-line mutations in the adenomatous polyposis coli (APC) gene are responsible for familial adenomatous polyposis (FAP). Genotype-phenotype correlation studies in patients with FAP have demonstrated associations of certain variants of the disease with mutations at specific sites within the APC gene. In a large FAP family, we identified a frameshift mutation located in the alternatively spliced region of exon 9. Phenotypic studies of affected family members showed that the clinical course of FAP was delayed, with gastrointestinal symptoms and death from colorectal carcinoma occurring on average 25 and 20 years later than usual, respectively. The numbers of colorectal adenomas differed markedly among affected individuals and the location of colorectal cancer lay frequently in the proximal colon. Our findings suggest that the exon 9 mutation identified in the pedigree is associated with late onset of FAP. The atypical phenotype may be explained by the site of the mutation in the APC gene. Analysis of the APC protein product indicated that the exon 9 mutation did not result in a detectable truncated APC protein. Given the location of the mutation within an alternatively spliced exon of APC, it is conceivable that normal APC proteins are produced from the mutant allele by alternative splicing.


Genes, Chromosomes and Cancer | 2000

SomaticApc mutations are selected upon their capacity to inactivate the ?-catenin downregulating activity

Ron Smits; Nandy Hofland; Winfried Edelmann; Marjan Geugien; Shantie Jagmohan-Changur; Cristina Albuquerque; Cor Breukel; Raju Kucherlapati; Menno F. Kielman; Riccardo Fodde

The APC gene, originally identified as the gene for familial adenomatous polyposis (FAP), is now considered as the true “gatekeeper” of colonic epithelial proliferation. Its main tumor suppressing activity seems to reside in the capacity to properly regulate intracellular β‐catenin signaling. Most somatic APC mutations are detected between codons 1286 and 1513, the mutation cluster region (MCR). This clustering can be explained either by the presence of mutation‐prone sequences within the MCR, or by the selective advantage provided by the resulting truncated polypeptides. Here, a Msh2‐deficient mouse model (Msh2Δ7N ) was generated and bred with Apc1638N and ApcMin that allowed the comparison of the somatic mutation spectra along the Apc gene in the different allelic combinations. Mutations identified in Msh2Δ7N/Δ7N tumors are predominantly dinucleotide deletions at simple sequence repeats leading to truncated Apc polypeptides that partially retain the 20 a.a. β‐catenin downregulating motifs. In contrast, the somatic mutations identified in the wild type Apc allele of Msh2Δ7N/Δ7N /Apc+/1638N and Msh2Δ7N/Δ7N /Apc+/Min tumors are clustered more to the 5′ end, thereby completely inactivating the β‐catenin downregulating activity of APC. These results indicate that somatic Apc mutations are selected during intestinal tumorigenesis and that inactivation of the β‐catenin downregulating function of APC is likely to represent the main selective factor.


Journal of Immunology | 2011

The Inhibiting Fc Receptor for IgG, FcγRIIB, Is a Modifier of Autoimmune Susceptibility

Peter Boross; Victoria L. Arandhara; Javier Martin-Ramirez; Marie-Laure Santiago-Raber; Francesco Carlucci; Roelof Flierman; Jos van der Kaa; Cor Breukel; Jill W. C. Claassens; Marcel Camps; Erik Lubberts; Daniela Salvatori; Maria Pia Rastaldi; Ferry Ossendorp; Mohamed R. Daha; H. Terence Cook; Shozo Izui; Marina Botto; J. Sjef Verbeek

FcγRIIB-deficient mice generated in 129 background (FcγRIIB129−/−) if back-crossed into C57BL/6 background exhibit a hyperactive phenotype and develop lethal lupus. Both in mice and humans, the Fcγr2b gene is located within a genomic interval on chromosome 1 associated with lupus susceptibility. In mice, the 129-derived haplotype of this interval, named Sle16, causes loss of self-tolerance in the context of the B6 genome, hampering the analysis of the specific contribution of FcγRIIB deficiency to the development of lupus in FcγRIIB129−/− mice. Moreover, in humans genetic linkage studies revealed contradictory results regarding the association of “loss of function” mutations in the Fcγr2b gene and susceptibility to systemic lupus erythematosis. In this study, we demonstrate that FcγRIIB−/− mice generated by gene targeting in B6-derived ES cells (FcγRIIBB6−/−), lacking the 129-derived flanking Sle16 region, exhibit a hyperactive phenotype but fail to develop lupus indicating that in FcγRIIB129−/− mice, not FcγRIIB deficiency but epistatic interactions between the C57BL/6 genome and the 129-derived Fcγr2b flanking region cause loss of tolerance. The contribution to the development of autoimmune disease by the resulting autoreactive B cells is amplified by the absence of FcγRIIB, culminating in lethal lupus. In the presence of the Yaa lupus-susceptibility locus, FcγRIIBB6−/− mice do develop lethal lupus, confirming that FcγRIIB deficiency only amplifies spontaneous autoimmunity determined by other loci.


Human Genetics | 1988

Close linkage of a highly polymorphic marker (D5S37) to familial adenomatous polyposis (FAP) and confirmation of FAP localization on chromosome 5q21-q22.

P. Meera Khan; C. Tops; Marianne van den Broek; Cor Breukel; Juul T. Wijnen; M. Oldenburg; J. v. d. Bos; I. S. J. Van Leeuwen-Cornelisse; Hans F. A. Vasen; G. Griffioen; H. M. Verspaget; F.C.A. den Hartog Jager

SummaryFifteen apparently unrelated Dutch families with familial adenomatous polyposis (FAP) also known as familial polyposis coli (FPC; McKusick No. 17510) were screened for linkage with the DNA probe C11p11 localized on chromosome 5q21–22 and previously reported to be closely linked to FAP (Bodmer et al. 1987; Leppert et al. 1987). In our study C11p11 was minimally informative, which is ascribable to its low heterozygosity in the North European populations. Of the above families, 12 were investigated also for linkage with D5S37 (DNA probe Pi227). Data from 11 of them were found to be informative and showed that FAP is closely linked to D5S37 previously localized on chromosome 5q21 (peak lod score 7.85 at a recombination fraction of 0.048 with 95% probability limits 0.005–0.145). Results discussed below indicate for the first time that the most likely location of the FAP gene is in the band 5q22 very close to 5q21, if not in the transitional zone between these two bands. The probe Pi227 recognizes 4 restriction fragment length polymorphism (RFLP) sites, exhibiting a total of 9 alleles with 24 theoretically possible haplotypes in the Dutch population. Therefore, this probe appears to have potential as a generally useful predictive marker for FAP until much closer and similarly useful markers become available.


PLOS Genetics | 2009

A targeted constitutive mutation in the APC tumor suppressor gene underlies mammary but not intestinal tumorigenesis.

Claudia Gaspar; Patrick Franken; Lia Molenaar; Cor Breukel; Martin van der Valk; Ron Smits; Riccardo Fodde

Germline mutations in the adenomatous polyposis coli (APC) gene are responsible for familial adenomatous polyposis (FAP), an autosomal dominant hereditary predisposition to the development of multiple colorectal adenomas and of a broad spectrum of extra-intestinal tumors. Moreover, somatic APC mutations play a rate-limiting and initiating role in the majority of sporadic colorectal cancers. Notwithstanding its multifunctional nature, the main tumor suppressing activity of the APC gene resides in its ability to regulate Wnt/β-catenin signaling. Notably, genotype–phenotype correlations have been established at the APC gene between the length and stability of the truncated proteins encoded by different mutant alleles, the corresponding levels of Wnt/β-catenin signaling activity they encode for, and the incidence and distribution of intestinal and extra-intestinal tumors. Here, we report a novel mouse model, Apc1572T, obtained by targeting a truncated mutation at codon 1572 in the endogenous Apc gene. This hypomorphic mutant allele results in intermediate levels of Wnt/β-catenin signaling activation when compared with other Apc mutations associated with multifocal intestinal tumors. Notwithstanding the constitutive nature of the mutation, Apc +/1572T mice have no predisposition to intestinal cancer but develop multifocal mammary adenocarcinomas and subsequent pulmonary metastases in both genders. The histology of the Apc1572T primary mammary tumours is highly heterogeneous with luminal, myoepithelial, and squamous lineages and is reminiscent of metaplastic carcinoma of the breast in humans. The striking phenotype of Apc +/1572T mice suggests that specific dosages of Wnt/β-catenin signaling activity differentially affect tissue homeostasis and initiate tumorigenesis in an organ-specific fashion.

Collaboration


Dive into the Cor Breukel's collaboration.

Top Co-Authors

Avatar

Riccardo Fodde

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

J. Sjef Verbeek

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jill W. C. Claassens

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Carli M. J. Tops

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Menno F. Kielman

Leiden University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge