Network


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

Hotspot


Dive into the research topics where Frederik J. Van Schooten is active.

Publication


Featured researches published by Frederik J. Van Schooten.


The FASEB Journal | 2013

Paternal lifestyle as a potential source of germline mutations transmitted to offspring

Joost O. Linschooten; Nicole Verhofstad; Kristine B. Gutzkow; Ann-Karin Olsen; Carole L. Yauk; Yvonne Oligschläger; Gunnar Brunborg; Frederik J. Van Schooten; Roger W. L. Godschalk

Paternal exposure to high levels of radioactivity causes heritable germline minisatellite mutations. However, the effect of more general paternal exposures, such as cigarette smoking, on germline mutations remains unexplored. We analyzed two of the most commonly used minisatellite loci (CEB1 and B6.7) to identify germline mutations in blood samples of complete mother‐father‐child triads from the Norwegian Mother and Child Cohort Study (MoBa). The presence of mutations was subsequently related to general lifestyle factors, including paternal smoking before the partner became pregnant. Paternally derived mutations at the B6.7 locus (mutation frequency 0.07) were not affected by lifestyle. In contrast, high gross yearly income as a general measure of a healthy lifestyle coincided with low‐mutation frequencies at the CEB1 locus (P=0.047). Income was inversely related to smoking behavior, and paternally derived CEB1 mutations were dose dependently increased when the father smoked in the 6 mo before pregnancy, 0.21 vs. 0.05 in smoking and nonsmoking fathers, respectively (P=0.061). These results suggest that paternal lifestyle can affect the chance of heritable mutations in unstable repetitive DNA sequences. To our knowledge, this is the first study reporting an effect of lifestyle on germline minisatellite mutation frequencies in a human population with moderate paternal exposures.—Linschooten, J. O., Verhofstad, N., Gutzkow, K., Olsen, A.‐K., Yauk, C., Oligschläger, Y., Brunborg, G., van Schooten, F. J., Godschalk, R. W. L. Paternal lifestyle as a potential source of germline mutations transmitted to offspring. FASEB J. 27, 2873‐2879 (2013). www.fasebj.org


Proceedings of the National Academy of Sciences of the United States of America | 2008

Germ-line mutations, DNA damage, and global hypermethylation in mice exposed to particulate air pollution in an urban/industrial location

Carole L. Yauk; Aris Polyzos; Andrea Rowan-Carroll; Christopher M. Somers; Roger W. L. Godschalk; Frederik J. Van Schooten; M. Lynn Berndt; Igor P. Pogribny; Igor Koturbash; Andrew Williams; George R. Douglas; Olga Kovalchuk

Particulate air pollution is widespread, yet we have little understanding of the long-term health implications associated with exposure. We investigated DNA damage, mutation, and methylation in gametes of male mice exposed to particulate air pollution in an industrial/urban environment. C57BL/CBA mice were exposed in situ to ambient air near two integrated steel mills and a major highway, alongside control mice breathing high-efficiency air particulate (HEPA) filtered ambient air. PCR analysis of an expanded simple tandem repeat (ESTR) locus revealed a 1.6-fold increase in sperm mutation frequency in mice exposed to ambient air for 10 wks, followed by a 6-wk break, compared with HEPA-filtered air, indicating that mutations were induced in spermatogonial stem cells. DNA collected after 3 or 10 wks of exposure did not exhibit increased mutation frequency. Bulky DNA adducts were below the detection threshold in testes samples, suggesting that DNA reactive chemicals do not reach the germ line and cause ESTR mutation. In contrast, DNA strand breaks were elevated at 3 and 10 wks, possibly resulting from oxidative stress arising from exposure to particles and associated airborne pollutants. Sperm DNA was hypermethylated in mice breathing ambient relative to HEPA-filtered air and this change persisted following removal from the environmental exposure. Increased germ-line DNA mutation frequencies may cause population-level changes in genetic composition and disease. Changes in methylation can have widespread repercussions for chromatin structure, gene expression and genome stability. Potential health effects warrant extensive further investigation.


Journal of Breath Research | 2012

The versatile use of exhaled volatile organic compounds in human health and disease

Agnes W. Boots; Joep J B N van Berkel; J.W. Dallinga; Agnieszka Smolinska; Emile F Wouters; Frederik J. Van Schooten

Exhaled breath contains thousands of volatile organic compounds (VOCs) of which the composition varies depending on health status. Various metabolic processes within the body produce volatile products that are released into the blood and will be passed on to the airway once the blood reaches the lungs. Moreover, the occurrence of chronic inflammation and/or oxidative stress can result in the excretion of volatile compounds that generate unique VOC patterns. Consequently, measuring the total amount of VOCs in exhaled air, a kind of metabolomics also referred to as breathomics, for clinical diagnosis and monitoring purposes gained increased interest over the last years. This paper describes the currently available methodologies regarding sampling, sample analysis and data processing as well as their advantages and potential drawbacks. Additionally, different application possibilities of VOC profiling are discussed. Until now, breathomics has merely been applied for diagnostic purposes. Exhaled air analysis can, however, also be applied as an analytical or monitoring tool. Within the analytic perspective, the use of VOCs as biomarkers of oxidative stress, inflammation or carcinogenesis is described. As monitoring tool, breathomics can be applied to elucidate the heterogeneity observed in chronic diseases, to study the pathogen(s) responsible for occurring infections and to monitor treatment efficacy.


The FASEB Journal | 2007

Activated neutrophils inhibit nucleotide excision repair in human pulmonary epithelial cells: role of myeloperoxidase

Nejla Güngör; Roger W. L. Godschalk; D. M. F. A. Pachen; Frederik J. Van Schooten; Ad M. Knaapen

Neutrophils are thought to affect pulmonary carcinogenesis by promoting the metabolism of inhaled chemical carcinogens, causing enhanced formation of promutagenic DNA adducts. We hypothesized that neutrophils interfere with the removal of such DNA adducts by inhibiting nucleotide excision repair (NER) in target cells. Human alveolar epithelial cells (A549) were cocultured with activated neutrophils, and we observed a significant reduction of NER in the A549 cells, which was abrogated by addition of the myeloper‐oxidase (MPO) inhibitor 4‐aminobenzoic acid hydra‐zide. The inhibitory effect of neutrophils could be mimicked by the MPO product hypochlorous acid (HOCl), which caused an acute, dose‐dependent inhibition of NER in A549 cells. This was independent of cytotoxicity or ATP loss and persisted up to 24 h. These data were supported by showing that HOCl caused a delayed removal of DNA adducts in benzo[a]pyrene‐diolepoxide‐exposed A549 cells. The acute HOCl‐in‐duced inhibition of NER can only partly be explained by oxidative modification of repair proteins. To explain the more persistent effects of HOCl, we analyzed the expression of NER genes and found that HOCl significantly reduced XPC expression. In conclusion, these data indicate that neutrophils are potent inhibitors of nucleotide excision repair. This may provide a further biological explanation for the association between inflammation and lung cancer development.—Güngör N., Godschalk, R. W. L., Pachen, D. M., Van Schooten F. J., Knaapen A. M. Activated neutro‐phils inhibit nucleotide excision repair in human pulmonary epithelial cells: role of myeloperoxidase. FASEB J. 21, 2359–2367 (2007)


The FASEB Journal | 2005

Polycyclic aromatic hydrocarbons induce an inflammatory atherosclerotic plaque phenotype irrespective of their DNA binding properties

Daniëlle M.J. Curfs; Ad M. Knaapen; Daniëlle M.F.A. Pachen; Marion J. J. Gijbels; Esther Lutgens; Marjan Smook; Mark M. Kockx; Mat J.A.P. Daemen; Frederik J. Van Schooten

Although it has been demonstrated that carcinogenic environmental polycyclic aromatic hydrocarbons (PAHs) cause progression of atherosclerosis, the underlying mechanism remains unclear. In the present study, we aimed to investigate whether DNA binding events are critically involved in the progression of PAH‐mediated atherogenesis. Apolipoprotein E knockout mice were orally (24 wk, once/wk) exposed to 5 mg/kg benzo[a]pyrene (B[a]P), or its nonmutagenic, noncarcinogenic structural isoform benzo[e]pyrene (B[e]P). 32P‐postlabeling of lung tissue confirmed the presence of promutagenic PAH‐DNA adducts in B[a]P‐exposed animals, whereas in B[e]P‐exposed and vehicle control animals, these adducts were undetectable. Morphometrical analysis showed that both B[a]P and B[e]P caused an increase in plaque size, whereas location or number of plaques was unaffected. Immunohistochemistry revealed no differences in oxidative DNA damage (8‐OHdG) or apoptosis in the plaques. Also plasma lipoprotein levels remained unchanged after PAH‐exposure. However, T lymphocytes were increased ≥2‐fold in the plaques of B[a]P‐ and B[e]P‐exposed animals. Additionally, B[a]P and to a lesser extent B[e]P exposure resulted in increased TGFβ protein levels in the plaques, that was mainly localized in the plaque macrophages. In vitro studies using the murine macrophage like RAW264.7 cells showed that inhibition of TGFβ resulted in decreased tumor necrosis factor (TNF) α release, suggesting that enhanced TGFβ expression in the plaque macrophages contributes to the proinflammatory effects in the vessel wall. In general, this inflammatory reaction in the plaques appeared to be a local response since peripheral blood cell composition (T cells, B cells, granulocytes, and macrophages) was not changed upon PAH exposure. In conclusion, we showed that both B[a]P and B[e]P cause progression of atherosclerosis, irrespective of their DNA binding properties. Moreover, our data revealed a possible novel mechanism of PAH‐mediated atherogenesis, which likely involves a TGFβ‐mediated local inflammatory reaction in the vessel wall.


American Journal of Pathology | 2004

Chronic Exposure to the Carcinogenic Compound Benzo[a]Pyrene Induces Larger and Phenotypically Different Atherosclerotic Plaques in ApoE-Knockout Mice

Daniëlle M.J. Curfs; Esther Lutgens; Marion J. J. Gijbels; Mark M. Kockx; Mat J.A.P. Daemen; Frederik J. Van Schooten

Benzo[a]pyrene (B[a]P) is a polycyclic aromatic hydrocarbon with atherogenic and carcinogenic properties. The role of B[a]P in carcinogenesis is well established, and thought to exert via enzymatic activation into reactive metabolites that are capable of binding to the DNA leading to uncontrolled proliferation. However, the mechanism underlying the atherogenic properties of B[a]P is still unclear. Therefore, the effects of chronic B[a]P exposure on atherosclerotic plaque development in apolipoprotein E knockout (apoE-KO) mice were studied. ApoE-KO mice were orally treated with 5 mg/kg/bw B[a]P once per week for 12 or 24 consecutive weeks. Levels of reactive B[a]P metabolites in the arterial tree (from the aortic arch until the iliac artery bifurcations) were high as shown by the level of B[a]P DNA-binding products measured in DNA isolated from the entire aorta (38.9 +/- 4.8 adducts/10(8) nucleotides). Analysis of atherosclerotic lesions in the aortic arch showed no influence of B[a]P on location or number of lesions. Moreover, no increased levels of p53 nuclear protein accumulation or cell proliferation, as detected by immunohistochemistry, were seen in the plaques of the B[a]P-exposed animals. However, the effects of B[a]P on advanced lesions were obvious: advanced plaques were larger and more prone to lipid core development and plaque layering at both 12 and 24 weeks (P < 0.05). In the B[a]P-exposed animals advanced plaques contained more T-lymphocytes and macrophages than in the control animals at both end points (P < 0.05). These data suggest that B[a]P does not initiate atherosclerosis in apoE-KO mice, but accelerates the progression of atherosclerotic plaques via a local inflammatory response.


Toxicology and Applied Pharmacology | 2010

Transcriptomics-based identification of developmental toxicants through their interference with cardiomyocyte differentiation of embryonic stem cells.

Dorien A.M. van Dartel; Jeroen L. A. Pennings; Frederik J. Van Schooten; Aldert H. Piersma

The embryonic stem cell test (EST) predicts developmental toxicity based on the inhibition of cardiomyocyte differentiation of embryonic stem cells (ESC). The subjective endpoint, the long culture duration together with the undefined applicability domain and related predictivity need further improvement to facilitate implementation of the EST into regulatory strategies. These aspects may be improved by studying gene expression changes in the ESC differentiation cultures and their modulation by compound exposure using transcriptomics. Here, we tested the developmental toxicants monobutyl phthalate and 6-aminonicotinamide. ESC were allowed to differentiated, and cardiomyocyte differentiation was assessed after 10 days of culture. RNA of solvent controls was collected after 0, 24, 48, 72 and 96 h of exposure, and RNA of developmental-toxicant-exposed cultures was collected after 24 and 96 h. Samples were hybridized to DNA microarrays, and 1355 genes were found differentially expressed among the unexposed experimental groups. These regulated genes were involved in differentiation-related processes, and Principal Component Analysis (PCA) based on these genes showed that the unexposed experimental groups appeared in chronological order in the PCA, which can therefore be regarded as a continuous representation of the differentiation track. The developmental-toxicant-exposed cultures appeared to deviate significantly from this differentiation track, which confirms the compound-modulating effects on the differentiation process. The incorporation of transcriptomics in the EST is expected to provide a more informative and improved endpoint in the EST as compared with morphology, allowing early detection of differentiation modulation. Furthermore, this approach may improve the definition of the applicability domain and predictivity of the EST.


The FASEB Journal | 2011

Epigenetics: prenatal exposure to genistein leaves a permanent signature on the hematopoietic lineage

Kimberly Vanhees; Susan Steinbusch Coort; Erik J.B. Ruijters; Roger W. L. Godschalk; Frederik J. Van Schooten; Sahar Barjesteh van Waalwijk van Doorn-Khosrovani

Recent studies demonstrate that maternal diet during pregnancy results in long‐lasting effects on the progeny. Supplementation of maternal diet with genistein, a phytoestrogen ubiquitous in the daily diet, altered coat color of agouti mice due to epigenetic changes. We studied hematopoiesis of mice prenatally exposed to genistein (270 mg/kg feed) compared with that of mice prenatally exposed to phytoestrogen‐poor feed and observed a significant increase in granulopoi‐esis, erythropoiesis, and mild macrocytosis at the adult age of 12 wk. Genistein exposure was associated with hypermethylation of certain repetitive elements, which coincided with a significant down‐regulation of estrogen‐responsive genes and genes involved in hematopoi‐esis in bone marrow cells of genistein‐exposed mice, as assessed by microarray technology. Although genistein exposure did not affect global methylation in fetal liver of fetuses at embryonic day 14.5, it accelerated the switch from primitive to definitive erythroid lineage. Taken together, our data demonstrate that prenatal exposure to genistein affects fetal erythropoiesis and exerts lifelong alterations in gene expression and DNA methylation of hematopoietic cells.—Vanhees, K., Coort, S., Ruitjers, E.J. B., Godschalk, R. W. L., van Schooten, F. J., Barjesteh van Waalwijk van Doorn‐Khosrovani, S. Epigenetics: prenatal exposure to genistein leaves a permanent signature on the hematopoietic lineage. FASEB J. 25, 797–807 (2011). www.fasebj.org


Carcinogenesis | 2015

Causes of genome instability: the effect of low dose chemical exposures in modern society

Sabine A.S. Langie; Gudrun Koppen; Daniel Desaulniers; Fahd Al-Mulla; Rabeah Al-Temaimi; Amedeo Amedei; Amaya Azqueta; William H. Bisson; Dustin G. Brown; Gunnar Brunborg; Amelia K. Charles; Tao Chen; Annamaria Colacci; Firouz Darroudi; Stefano Forte; Laetitia Gonzalez; Roslida A. Hamid; Lisbeth E. Knudsen; Luc Leyns; Adela Lopez de Cerain Salsamendi; Lorenzo Memeo; Chiara Mondello; Carmel Mothersill; Ann-Karin Olsen; Sofia Pavanello; Jayadev Raju; Emilio Rojas; Rabindra Roy; Elizabeth P. Ryan; Patricia Ostrosky-Wegman

Genome instability is a prerequisite for the development of cancer. It occurs when genome maintenance systems fail to safeguard the genomes integrity, whether as a consequence of inherited defects or induced via exposure to environmental agents (chemicals, biological agents and radiation). Thus, genome instability can be defined as an enhanced tendency for the genome to acquire mutations; ranging from changes to the nucleotide sequence to chromosomal gain, rearrangements or loss. This review raises the hypothesis that in addition to known human carcinogens, exposure to low dose of other chemicals present in our modern society could contribute to carcinogenesis by indirectly affecting genome stability. The selected chemicals with their mechanisms of action proposed to indirectly contribute to genome instability are: heavy metals (DNA repair, epigenetic modification, DNA damage signaling, telomere length), acrylamide (DNA repair, chromosome segregation), bisphenol A (epigenetic modification, DNA damage signaling, mitochondrial function, chromosome segregation), benomyl (chromosome segregation), quinones (epigenetic modification) and nano-sized particles (epigenetic pathways, mitochondrial function, chromosome segregation, telomere length). The purpose of this review is to describe the crucial aspects of genome instability, to outline the ways in which environmental chemicals can affect this cancer hallmark and to identify candidate chemicals for further study. The overall aim is to make scientists aware of the increasing need to unravel the underlying mechanisms via which chemicals at low doses can induce genome instability and thus promote carcinogenesis.


The FASEB Journal | 2013

Maternal folate depletion and high-fat feeding from weaning affects DNA methylation and DNA repair in brain of adult offspring

Sabine A.S. Langie; Sebastian Achterfeldt; Joanna P. Gorniak; Kirstin J. A. Halley-Hogg; David Oxley; Frederik J. Van Schooten; Roger W. L. Godschalk; Jill A. McKay; John C. Mathers

The mechanisms through which environmental and dietary factors modulate DNA repair are still unclear but may include dysregulation of gene expression due to altered epigenetic markings. In a mouse model, we investigated the effect of maternal folate depletion during pregnancy and lactation, and high‐fat feeding from weaning, on base excision repair (BER) and DNA methylation and expression of selected BER‐related genes in the brain of adult offspring. While folate depletion did not affect BER activity of the mothers, BER increased in the offspring at weaning (P=0.052). In the long term, as observed in 6‐mo‐old offspring, the double insult, i.e., maternal low‐folate supply and high‐fat feeding from weaning, decreased BER activity significantly in the cortex, cerebellum, hippocampus, and subcortical regions (P <0.017). This fall in BER activity was associated with small changes in methylation or expression of BER‐related genes. Maternal folate depletion led to slightly increased oxidative DNA damage levels in subcortical regions of adult offspring, which may increase sensitivity to oxidative stress and predispose to neurological disorders. In summary, our data suggest that low‐folate supply during early life may leave an epigenetic mark that can predispose the offspring to further dietary insults, causing adverse effects during adult life.—Langie, S. A. S., Achterfeldt, S., Gorniak, J. P., Halley‐Hogg, K. J. A., Oxley, D., van Schooten, F. J., Godschalk, R. W. L., McKay, J. A., Mathers, J. C., Maternal folate depletion and high‐fat feeding from weaning affects DNA methylation and DNA repair in brain of adult offspring. FASEBJ. 27, 3323‐3334 (2013). www.fasebj.org

Collaboration


Dive into the Frederik J. Van Schooten's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge