Network


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

Hotspot


Dive into the research topics where Floris Foijer is active.

Publication


Featured researches published by Floris Foijer.


Cancer Research | 2011

Analysis of Mitosis and Antimitotic Drug Responses in Tumors by In Vivo Microscopy and Single-Cell Pharmacodynamics

James D. Orth; Rainer H. Kohler; Floris Foijer; Peter K. Sorger; Ralph Weissleder; Timothy J. Mitchison

Cancer relies upon frequent or abnormal cell division, but how the tumor microenvironment affects mitotic processes in vivo remains unclear, largely due to the technical challenges of optical access, spatial resolution, and motion. We developed high-resolution in vivo microscopy methods to visualize mitosis in a murine xenograft model of human cancer. Using these methods, we determined whether the single-cell response to the antimitotic drug paclitaxel (Ptx) was the same in tumors as in cell culture, observed the impact of Ptx on the tumor response as a whole, and evaluated the single-cell pharmacodynamics (PD) of Ptx (by in vivo PD microscopy). Mitotic initiation was generally less frequent in tumors than in cell culture, but subsequently it proceeded normally. Ptx treatment caused spindle assembly defects and mitotic arrest, followed by slippage from mitotic arrest, multinucleation, and apoptosis. Compared with cell culture, the peak mitotic index in tumors exposed to Ptx was lower and the tumor cells survived longer after mitotic arrest, becoming multinucleated rather than dying directly from mitotic arrest. Thus, the tumor microenvironment was much less proapoptotic than cell culture. The morphologies associated with mitotic arrest were dose and time dependent, thereby providing a semiquantitative, single-cell measure of PD. Although many tumor cells did not progress through Ptx-induced mitotic arrest, tumor significantly regressed in the model. Our findings show that in vivo microscopy offers a useful tool to visualize mitosis during tumor progression, drug responses, and cell fate at the single-cell level.


Genes & Development | 2010

Loss of Rb proteins causes genomic instability in the absence of mitogenic signaling

Tanja van Harn; Floris Foijer; Marcel A. T. M. van Vugt; Ruby Banerjee; Fentang Yang; Anneke B. Oostra; Hans Joenje; Hein te Riele

Loss of G1/S control is a hallmark of cancer, and is often caused by inactivation of the retinoblastoma pathway. However, mouse embryonic fibroblasts lacking the retinoblastoma genes RB1, p107, and p130 (TKO MEFs) are still subject to cell cycle control: Upon mitogen deprivation, they enter and complete S phase, but then firmly arrest in G2. We now show that G2-arrested TKO MEFs have accumulated DNA damage. Upon mitogen readdition, cells resume proliferation, although only part of the damage is repaired. As a result, mitotic cells show chromatid breaks and chromatid cohesion defects. These aberrations lead to aneuploidy in the descendent cell population. Thus, our results demonstrate that unfavorable growth conditions can cause genomic instability in cells lacking G1/S control. This mechanism may allow premalignant tumor cells to acquire additional genetic alterations that promote tumorigenesis.


Developmental Cell | 2017

Centrosome Amplification Is Sufficient to Promote Spontaneous Tumorigenesis in Mammals

Michelle S. Levine; Bjorn Bakker; Bram Boeckx; Julia Moyett; James Lu; Benjamin Vitre; Diana C. J. Spierings; Peter M. Lansdorp; Don W. Cleveland; Diether Lambrechts; Floris Foijer; Andrew J. Holland

Centrosome amplification is a common feature of human tumors, but whether this is a cause or a consequence of cancer remains unclear. Here, we test the consequence of centrosome amplification by creating mice in which centrosome number can be chronically increased in the absence of additional genetic defects. We show that increasing centrosome number elevated tumor initiation in a mouse model of intestinal neoplasia. Most importantly, we demonstrate that supernumerary centrosomes are sufficient to drive aneuploidy and the development of spontaneous tumors in multiple tissues. Tumors arising from centrosome amplification exhibit frequent mitotic errors and possess complex karyotypes, recapitulating a common feature of human cancer. Together, our data support a direct causal relationship among centrosome amplification, genomic instability, and tumor development.


Biochimica et Biophysica Acta | 2008

Studying chromosome instability in the mouse

Floris Foijer; Viji M. Draviam; Peter K. Sorger

Aneuploidy has long been recognized as one of the hallmarks of cancer. It nonetheless remains uncertain whether aneuploidy occurring early in the development of a cancer is a primary cause of oncogenic transformation, or whether it is an epiphenomenon that arises from a general breakdown in cell cycle control late in tumorigenesis. The accuracy of chromosome segregation is ensured both by the intrinsic mechanics of mitosis and by an error-checking spindle assembly checkpoint. Many cancers show altered expression of proteins involved in the spindle checkpoint or in proteins implicated in other mitotic processes. To understand the role of aneuploidy in the initiation and progression of cancer, a number of spindle checkpoint genes have been disrupted in mice, most through conventional gene targeting (to create germ-line knockouts). We describe the consequence of these mutations with respect to embryonic development, tumor progression and an unexpected link to premature aging; readers are referred elsewhere [1] for a discussion of other cell cycle regulators.


Cell Cycle | 2006

Check, double check: The G2 barrier to cancer

Floris Foijer; Hein te Riele

Loss of the G1/S checkpoint is recognized as a mandatory step in development of cancer. Nevertheless, both in vivo and in vitro experiments have indicated that this condition highly sensitizes cells to apoptosis. E.g., primary mouse embryonic fibroblasts that lack the complete retinoblastoma suppressor gene family (TKO MEFs) massively die under mitogen-deprived conditions. The prevailing hypothesis therefore is that the increased proliferative capacity of cells that have lost the G1/S checkpoint becomes apparent by suppression of apoptosis. However, this view was recently challenged by the finding that suppression of apoptotic cell death in TKO MEFs did not allow unconstrained proliferation; instead, cells became arrested in G2. This mechanism, which is dependent on p53, provides yet another barrier to oncogenic transformation. Thus, progression to malignancy of Rb-deficient lesions by alleviation of G2 arrest may offer an alternative explanation for the synergism between loss of Rb and p53 in tumorigenesis.


Genome Biology | 2016

Single-cell whole genome sequencing reveals no evidence for common aneuploidy in normal and Alzheimer's disease neurons

Hilda van den Bos; Diana C. J. Spierings; Aaron Taudt; Bjorn Bakker; David Porubský; Ester Falconer; Carolina Novoa; Nancy Halsema; Hinke G. Kazemier; Karina Hoekstra-Wakker; Victor Guryev; Wilfred F. A. den Dunnen; Floris Foijer; Maria Colomé-Tatché; Hendrikus Boddeke; Peter M. Landsdorp

BackgroundAlzheimer’s disease (AD) is a neurodegenerative disease of the brain and the most common form of dementia in the elderly. Aneuploidy, a state in which cells have an abnormal number of chromosomes, has been proposed to play a role in neurodegeneration in AD patients. Several studies using fluorescence in situ hybridization have shown that the brains of AD patients contain an increased number of aneuploid cells. However, because the reported rate of aneuploidy in neurons ranges widely, a more sensitive method is needed to establish a possible role of aneuploidy in AD pathology.ResultsIn the current study, we used a novel single-cell whole genome sequencing (scWGS) approach to assess aneuploidy in isolated neurons from the frontal cortex of normal control individuals (n = 6) and patients with AD (n = 10). The sensitivity and specificity of our method was shown by the presence of three copies of chromosome 21 in all analyzed neuronal nuclei of a Down’s syndrome sample (n = 36). Very low levels of aneuploidy were found in the brains from control individuals (n = 589) and AD patients (n = 893). In contrast to other studies, we observe no selective gain of chromosomes 17 or 21 in neurons of AD patients.ConclusionscWGS showed no evidence for common aneuploidy in normal and AD neurons. Therefore, our results do not support an important role for aneuploidy in neuronal cells in the pathogenesis of AD. This will need to be confirmed by future studies in larger cohorts.


Journal of Biological Chemistry | 2005

Characterization of the condensin component Cnap1 and protein kinase Melk as novel E2F target genes down-regulated by 1,25-dihydroxyvitamin D3

Lieve Verlinden; Guy Eelen; Ine Beullens; Mark Van Camp; Paul Van Hummelen; Kristof Engelen; Ruth Van Hellemont; Kathleen Marchal; Bart De Moor; Floris Foijer; Hein te Riele; Monique Beullens; Mathieu Bollen; Chantal Mathieu; Roger Bouillon; Annemieke Verstuyf

1,25-Dihydroxyvitamin D3 (1,25(OH)2D3) has potent antiproliferative effects characterized by a hampered G1/S transition. cDNA microarrays were used to monitor expression of 21,492 genes in MC3T3-E1 mouse osteoblasts at 1, 6, 12, 24, and 36 h after treatment with 1,25(OH)2D3. Statistical analysis revealed a cluster of genes that were strongly down-regulated by 1,25(OH)2D3 and which not only function in cell cycle regulation and DNA replication but also mediate checkpoint control, DNA repair, chromosome modifications, and mitosis. Because many of these genes were shown earlier to be regulated by the transcriptional repressor E2F4, the intergenic regions of these 1,25(OH)2D3-down-regulated genes were searched for the presence of E2F binding sites. This led to the characterization of two novel E2F target genes, chromosome condensation-related SMC-associated protein 1 (Cnap1) and maternal embryonic leucine zipper kinase (Melk). Transfection studies and site-directed mutagenesis confirmed Cnap1 and Melk to be bona fide E2F targets. Repression of Cnap1 and Melk by 1,25(OH)2D3 was confirmed not only in MC3T3-E1 cells but also in several other bone-unrelated cell types. This down-regulation as well as the antiproliferative effect of 1,25(OH)2D3 depended on the pocket proteins p107 and p130 because 1,25(OH)2D3 failed to repress these E2F target genes and lost its antiproliferative action in p107–/–;p130–/– cells but not in pRb–/– cells.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Chromosome instability induced by Mps1 and p53 mutation generates aggressive lymphomas exhibiting aneuploidy-induced stress

Floris Foijer; Stephanie Xie; Judith E. Simon; Petra L. Bakker; Nathalie Conte; Stephanie H. Davis; Eva Kregel; Jos Jonkers; Allan Bradley; Peter K. Sorger

Significance Normal cells rarely missegregate chromosomes, but the majority of cancer cells have a chromosomal instability (CIN) phenotype that makes errors more common and results in abnormal chromosomal content (aneuploidy). Although aneuploidy promotes transformation via gain of oncogenes and loss of tumor suppressors, it also slows cell proliferation and disrupts metabolic homeostasis. Aneuploidy therefore represents a liability as well as a source of selective advantage for cancer cells. We provoked CIN in murine T cells by weakening the spindle-assembly checkpoint and then studied the consequences. We found that CIN dramatically accelerates cancer in a genetically predisposed background and that the resulting aneuploid cancers are metabolically deranged, a vulnerability that may open new avenues to treating aneuploid cancers. Aneuploidy is a hallmark of human solid cancers that arises from errors in mitosis and results in gain and loss of oncogenes and tumor suppressors. Aneuploidy poses a growth disadvantage for cells grown in vitro, suggesting that cancer cells adapt to this burden. To understand better the consequences of aneuploidy in a rapidly proliferating adult tissue, we engineered a mouse in which chromosome instability was selectively induced in T cells. A flanked by Lox mutation was introduced into the monopolar spindle 1 (Mps1) spindle-assembly checkpoint gene so that Cre-mediated recombination would create a truncated protein (Mps1DK) that retained the kinase domain but lacked the kinetochore-binding domain and thereby weakened the checkpoint. In a sensitized p53+/− background we observed that Mps1DK/DK mice suffered from rapid-onset acute lymphoblastic lymphoma. The tumors were highly aneuploid and exhibited a metabolic burden similar to that previously characterized in aneuploid yeast and cultured cells. The tumors nonetheless grew rapidly and were lethal within 3–4 mo after birth.


Genome Biology | 2016

Single-cell sequencing reveals karyotype heterogeneity in murine and human malignancies

Bjorn Bakker; Aaron Taudt; Mirjam E. Belderbos; David Porubsky; Diana C. J. Spierings; Tristan V. de Jong; Nancy Halsema; Hinke G. Kazemier; Karina Hoekstra-Wakker; Allan Bradley; Eveline S. J. M. de Bont; Anke van den Berg; Victor Guryev; Peter M. Lansdorp; Maria Colomé-Tatché; Floris Foijer

BackgroundChromosome instability leads to aneuploidy, a state in which cells have abnormal numbers of chromosomes, and is found in two out of three cancers. In a chromosomal instable p53 deficient mouse model with accelerated lymphomagenesis, we previously observed whole chromosome copy number changes affecting all lymphoma cells. This suggests that chromosome instability is somehow suppressed in the aneuploid lymphomas or that selection for frequently lost/gained chromosomes out-competes the CIN-imposed mis-segregation.ResultsTo distinguish between these explanations and to examine karyotype dynamics in chromosome instable lymphoma, we use a newly developed single-cell whole genome sequencing (scWGS) platform that provides a complete and unbiased overview of copy number variations (CNV) in individual cells. To analyse these scWGS data, we develop AneuFinder, which allows annotation of copy number changes in a fully automated fashion and quantification of CNV heterogeneity between cells. Single-cell sequencing and AneuFinder analysis reveals high levels of copy number heterogeneity in chromosome instability-driven murine T-cell lymphoma samples, indicating ongoing chromosome instability. Application of this technology to human B cell leukaemias reveals different levels of karyotype heterogeneity in these cancers.ConclusionOur data show that even though aneuploid tumours select for particular and recurring chromosome combinations, single-cell analysis using AneuFinder reveals copy number heterogeneity. This suggests ongoing chromosome instability that other platforms fail to detect. As chromosome instability might drive tumour evolution, karyotype analysis using single-cell sequencing technology could become an essential tool for cancer treatment stratification.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Spindle checkpoint deficiency is tolerated by murine epidermal cells but not hair follicle stem cells

Floris Foijer; Tia DiTommaso; Giacomo Donati; Katta Hautaviita; Stephanie Xie; Emma Heath; Ian Smyth; Fiona M. Watt; Peter K. Sorger; Allan Bradley

The spindle assembly checkpoint (SAC) ensures correct chromosome segregation during mitosis by preventing aneuploidy, an event that is detrimental to the fitness and survival of normal cells but oncogenic in tumor cells. Deletion of SAC genes is incompatible with early mouse development, and RNAi-mediated depletion of SAC components in cultured cells results in rapid death. Here we describe the use of a conditional KO of mouse Mad2, an essential component of the SAC signaling cascade, as a means to selectively induce chromosome instability and aneuploidy in the epidermis of the skin. We observe that SAC inactivation is tolerated by interfollicular epidermal cells but results in depletion of hair follicle bulge stem cells. Eventually, a histologically normal epidermis develops within ∼1 mo after birth, albeit without any hair. Mad2-deficient cells in this epidermis exhibited abnormal transcription of metabolic genes, consistent with aneuploid cell state. Hair follicle bulge stem cells were completely absent, despite the continued presence of rudimentary hair follicles. These data demonstrate that different cell lineages within a single tissue respond differently to chromosome instability: some proliferating cell lineages can survive, but stem cells are highly sensitive.

Collaboration


Dive into the Floris Foijer's collaboration.

Top Co-Authors

Avatar

Bjorn Bakker

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Diana C. J. Spierings

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Hein te Riele

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Peter M. Lansdorp

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Victor Guryev

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Aaron Taudt

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Anne Margriet Heijink

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Hilda van den Bos

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

René H. Medema

Netherlands Cancer Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge