69 results on '"Dorsman JC"'
Search Results
2. Proper genomic profiling of (BRCA1-mutated) basal-like breast carcinomas requires prior removal of tumor infiltrating lymphocytes
- Author
-
Massink, MPG, Kooi, IE, van Mil, SE, Jordanova, ES, Arneziane, N, Dorsman, JC, van Beek, DM, van der Voorn, JP, Sie, D, Ylstra, B, van Deurzen, Carolien, Martens, John, Smid, Marcel, Sieuwerts, Anieta, Weerd, Vanja, Foekens, John, van den Ouweland, Ans, van Dyk, E, Nederlof, PM, Waisfisz, Q, Meijers-Heijboer, H, Massink, MPG, Kooi, IE, van Mil, SE, Jordanova, ES, Arneziane, N, Dorsman, JC, van Beek, DM, van der Voorn, JP, Sie, D, Ylstra, B, van Deurzen, Carolien, Martens, John, Smid, Marcel, Sieuwerts, Anieta, Weerd, Vanja, Foekens, John, van den Ouweland, Ans, van Dyk, E, Nederlof, PM, Waisfisz, Q, and Meijers-Heijboer, H
- Abstract
Introduction: BRCA1-mutated breast carcinomas may have distinct biological features, suggesting the involvement of specific oncogenic pathways in tumor development. The identification of genomic aberrations characteristic for BRCA1-mutated breast carcinomas could lead to a better understanding of BRCA1-associated oncogenic events and could prove valuable in clinical testing for BRCA1-involvement in patients. Methods: For this purpose, genomic and gene expression profiles of basal-like BRCA1-mutated breast tumors (n = 27) were compared with basal-like familial BRCAX (non-BRCA1/2/CHEK2*1100delC) tumors (n = 14) in a familial cohort of 120 breast carcinomas. Results: Genome wide copy number profiles of the BRCA1-mutated breast carcinomas in our data appeared heterogeneous. Gene expression analyses identified varying amounts of tumor infiltrating lymphocytes (TILs) as a major cause for this heterogeneity. Indeed, selecting tumors with relative low amounts of TILs, resulted in the identification of three known but also five previously unrecognized BRCA1-associated copy number aberrations. Moreover, these aberrations occurred with high frequencies in the BRCA1-mutated tumor samples. Using these regions it was possible to discriminate BRCA1-mutated from BRCAX breast carcinomas, and they were validated in two independent cohorts. To further substantiate our findings, we used flow cytometry to isolate cancer cells from formalin-fixed, paraffin-embedded, BRCA1-mutated triple negative breast carcinomas with estimated TIL percentages of 40% and higher. Genomic profiles of sorted and unsorted fractions were compared by shallow whole genome sequencing and confirm our findings. Conclusion: This study shows that genomic profiling of in particular basal-like, and thus BRCA1-mutated, breast carcinomas is severely affected by the presence of high numbers of TILs. Previous reports on genomic profiling of BRCA1-mutated breast carcinomas have largely neglected this. Therefore, our findin
- Published
- 2015
3. Cultures of ovarian surface epithelium from women with and without a hereditary predisposition to develop female adnexal carcinoma
- Author
-
Piek, JMJ, Dorsman, JC, Shvarts, A, Massuger, LF, Ansink, Anca, Weegenaar, J, Kenemans, P, Verheijen, RHM, and Obstetrics & Gynecology
- Published
- 2004
4. DNA profiling of primary serous ovarian and Fallopian tube carcinomas with array comparative genomic hybridization and multiplex ligation-dependent probe amplification
- Author
-
Nowee, ME, primary, Snijders, AM, additional, Rockx, DAP, additional, de Wit, RM, additional, Kosma, VM, additional, Hämäläinen, K, additional, Schouten, JP, additional, Verheijen, RHM, additional, van Diest, PJ, additional, Albertson, DG, additional, and Dorsman, JC, additional
- Published
- 2007
- Full Text
- View/download PDF
5. Differentiating MYCN-amplified RB1 wild-type retinoblastoma from biallelic RB1 mutant retinoblastoma using MR-based radiomics: a retrospective multicenter case-control study.
- Author
-
de Bloeme CM, Jansen RW, Cardoen L, Göricke S, van Elst S, Jessen JL, Ramasubramanian A, Skalet AH, Miller AK, Maeder P, Uner OE, Hubbard GB, Grossniklaus H, Boldt HC, Nichols KE, Brennan RC, Sen S, Koob M, Sirin S, Brisse HJ, Galluzzi P, Dommering CJ, Cysouw M, Boellaard R, Dorsman JC, Moll AC, de Jong MC, and de Graaf P
- Subjects
- Humans, Female, Case-Control Studies, Male, Retrospective Studies, Child, Preschool, Infant, Retinal Neoplasms genetics, Retinal Neoplasms diagnostic imaging, Retinal Neoplasms pathology, Machine Learning, Mutation, Diagnosis, Differential, Child, Radiomics, Retinoblastoma genetics, Retinoblastoma diagnostic imaging, Retinoblastoma pathology, N-Myc Proto-Oncogene Protein genetics, Magnetic Resonance Imaging methods, Retinoblastoma Binding Proteins genetics, Ubiquitin-Protein Ligases genetics
- Abstract
MYCN-amplified RB1 wild-type (MYCN
amp RB1+/+ ) retinoblastoma is a rare and aggressive subtype, often resistant to standard therapies. Identifying unique MRI features is crucial for diagnosing this subtype, as biopsy is not recommended. This study aimed to differentiate MYCNamp RB1+/+ from the most prevalent RB1-/- retinoblastoma using pretreatment MRI and radiomics. Ninety-eight unilateral retinoblastoma patients (19 MYCN cases and 79 matched controls) were included. Tumors on T2-weighted MR images were manually delineated and validated by experienced radiologists. Radiomics analysis extracted 120 features per tumor. Several combinations of feature selection methods, oversampling techniques and machine learning (ML) classifiers were evaluated in a repeated fivefold cross-validation machine learning pipeline to yield the best-performing prediction model for MYCN. The best model used univariate feature selection, data oversampling (duplicating MYCN cases), and logistic regression classifier, achieving a mean AUC of 0.78 (SD 0.12). SHAP analysis highlighted lower sphericity, higher flatness, and greater gray-level heterogeneity as predictive for MYCNamp RB1+/+ status, yielding an AUC of 0.81 (SD 0.11). This study shows the potential of MRI-based radiomics to distinguish MYCNamp RB1+/+ and RB1-/- retinoblastoma subtypes., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
6. CRISPR screens in sister chromatid cohesion defective cells reveal PAXIP1-PAGR1 as regulator of chromatin association of cohesin.
- Author
-
van Schie JJM, de Lint K, Molenaar TM, Moronta Gines M, Balk JA, Rooimans MA, Roohollahi K, Pai GM, Borghuis L, Ramadhin AR, Corazza F, Dorsman JC, Wendt KS, Wolthuis RMF, and de Lange J
- Abstract
The cohesin complex regulates higher order chromosome architecture through maintaining sister chromatid cohesion and folding chromatin by DNA loop extrusion. Impaired cohesin function underlies a heterogeneous group of genetic syndromes and is associated with cancer. Here, we mapped the genetic dependencies of human cell lines defective of cohesion regulators DDX11 and ESCO2. The obtained synthetic lethality networks are strongly enriched for genes involved in DNA replication and mitosis and support the existence of parallel sister chromatid cohesion establishment pathways. Among the hits, we identify the chromatin binding, BRCT-domain containing protein PAXIP1 as a novel cohesin regulator. Depletion of PAXIP1 severely aggravates cohesion defects in ESCO2 mutant cells, leading to mitotic cell death. PAXIP1 promotes global chromatin association of cohesin, independent of DNA replication, a function that cannot be explained by indirect effects of PAXIP1 on transcription or DNA repair. Cohesin regulation by PAXIP1 requires its binding partner PAGR1 and a conserved FDF motif in PAGR1. PAXIP1 co-localizes with cohesin on multiple genomic loci, including active gene promoters and enhancers. Possibly, this newly identified role of PAXIP1-PAGR1 in regulating cohesin occupancy on chromatin is also relevant for previously described functions of PAXIP1 in transcription, immune cell maturation and DNA repair., (© The Author(s) 2023. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2023
- Full Text
- View/download PDF
7. Genome-wide siRNA screens identify RBBP9 function as a potential target in Fanconi anaemia-deficient head-and-neck squamous cell carcinoma.
- Author
-
Pai G, Roohollahi K, Rockx D, de Jong Y, Stoepker C, Pennings C, Rooimans M, Vriend L, Piersma S, Jimenez CR, De Menezes RX, Van Beusechem VW, Brakenhoff RH, Te Riele H, Wolthuis RMF, and Dorsman JC
- Subjects
- Humans, Cell Cycle Proteins genetics, DNA, Emetine therapeutic use, Genome-Wide Association Study, Intracellular Signaling Peptides and Proteins genetics, Neoplasm Proteins genetics, RNA, Small Interfering genetics, Fanconi Anemia genetics, Fanconi Anemia pathology, Head and Neck Neoplasms, Squamous Cell Carcinoma of Head and Neck genetics
- Abstract
Fanconi anaemia (FA) is a rare chromosomal-instability syndrome caused by mutations of any of the 22 known FA DNA-repair genes. FA individuals have an increased risk of head-and-neck squamous-cell-carcinomas (HNSCC), often fatal. Systemic intolerance to standard cisplatin-based protocols due to somatic-cell hypersensitivity underscores the urgent need to develop novel therapies. Here, we performed unbiased siRNA screens to unveil genetic interactions synthetic-lethal with FA-pathway deficiency in FA-patient HNSCC cell lines. We identified based on differential-lethality scores between FA-deficient and FA-proficient cells, next to common-essential genes such as PSMC1, PSMB2, and LAMTOR2, the otherwise non-essential RBBP9 gene. Accordingly, low dose of the FDA-approved RBBP9-targeting drug Emetine kills FA-HNSCC. Importantly both RBBP9-silencing as well as Emetine spared non-tumour FA cells. This study provides a minable genome-wide analyses of vulnerabilities to address treatment challenges in FA-HNSCC. Our investigation divulges a DNA-cross-link-repair independent lead, RBBP9, for targeted treatment of FA-HNSCCs without systemic toxicity., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
8. Retinoblastoma: From genes to patient care.
- Author
-
Bouchoucha Y, Matet A, Berger A, Carcaboso AM, Gerrish A, Moll A, Jenkinson H, Ketteler P, Dorsman JC, Chantada G, Beck-Popovic M, Munier F, Aerts I, Doz F, and Golmard L
- Subjects
- Child, Humans, Genes, Retinoblastoma, Mutation, Patient Care, DNA Mutational Analysis methods, Retinoblastoma genetics, Retinal Neoplasms genetics, Retinal Neoplasms pathology
- Abstract
Retinoblastoma is the most common paediatric neoplasm of the retina, and one of the earliest model of cancer genetics since the identification of the master tumour suppressor gene RB1. Tumorigenesis has been shown to be driven by pathogenic variants of the RB1 locus, but also genomic and epigenomic alterations outside the locus. The increasing knowledge on this "mutational landscape" is used in current practice for precise genetic testing and counselling. Novel methods provide access to pre-therapeutic tumour DNA, by isolating cell-free DNA from aqueous humour or plasma. This is expected to facilitate assessment of the constitutional status of RB1, to provide an early risk stratification using molecular prognostic markers, to follow the response to the treatment in longitudinal studies, and to predict the response to targeted therapies. The aim of this review is to show how molecular genetics of retinoblastoma drives diagnosis, treatment, monitoring of the disease and surveillance of the patients and relatives. We first recap the current knowledge on retinoblastoma genetics and its use in every-day practice. We then focus on retinoblastoma subgrouping at the era of molecular biology, and the expected input of cell-free DNA in the field., Competing Interests: Declaration of competing interest The authors declare no conflict of interest., (Copyright © 2022. Published by Elsevier Masson SAS.)
- Published
- 2023
- Full Text
- View/download PDF
9. High-Level MYCN- Amplified RB1- Proficient Retinoblastoma Tumors Retain Distinct Molecular Signatures.
- Author
-
Roohollahi K, de Jong Y, van Mil SE, Fabius AWM, Moll AC, and Dorsman JC
- Abstract
Purpose: Retinoblastomas are malignant eye tumors diagnosed in young children. Most retinoblastomas are genetically characterized by biallelic inactivation of the RB1 gene. However, 1.5% of tumors demonstrate high-level amplification of the proto-oncogene MYCN . Patients with MYCN -amplified RB1 -proficient retinoblastoma receive a diagnosis at an earlier age and show a clinically and histologically more malignant phenotype. This study aimed to identify genome-wide molecular features that distinguish this subtype from other retinoblastomas., Design: Cohort study., Participants: Forty-seven retinoblastoma tumors, comprising 36 RB1
-/- , 4 RB1+/- , and 7 RB1+/+ tumors. In total, 5 retinoblastomas displayed high-level MYCN amplification, with 3 being RB1+/+ , 1 being RB1+/- , and 1 being RB1-/- ., Methods: Integrated analysis, based on gene expression, methylation, and methylation-expression correlations, was performed to identify distinct molecular components of MYCN -amplified RB1 -proficient retinoblastomas compared with other retinoblastoma subtypes. The methylation and methylation-expression correlation analysis was initially conducted within a subset of samples (n = 15) for which methylation profiles were available. The significant findings were cross-validated in the entire cohort (n = 47) and in publicly available data., Main Outcome Measures: Differentially expressed genes/pathways, differentially methylated genes, and methylation-driven differential gene expression., Results: A large number of genes (n = 3155) were identified with distinct expression patterns in MYCN -amplified RB1 -proficient retinoblastomas. The upregulated and downregulated genes were associated with translation and cell-cycle processes, respectively. Methylation analysis revealed distinct methylated patterns in MYCN -amplified RB1 -proficient tumors, many of which showing significant impact on gene expression. Data integration identified a 40-gene expression signature with hypermethylated state resulting in a significant downregulation in MYCN -amplified RB1 -proficient retinoblastomas. Cross-validation using the entire cohort and the public domain expression data verified the overall lower expression of these genes not only in retinoblastomas with a MYCN -amplified RB1 -proficient background, but also in MYCN- amplified neuroblastomas. These include the metabolism-associated TSTD1 gene and the cyclin-dependent kinase inhibitor gene CDKN2C ., Conclusions: MYCN -amplified RB1 -proficient retinoblastomas display significantly distinct molecular features compared with other retinoblastomas, including a set of 40 hypermethylation-driven downregulated genes. This gene set can give insight into the biology of MYCN -amplified retinoblastomas and may help us to understand the more aggressive clinical behavior., (© 2022 by the American Academy of Ophthalmology.)- Published
- 2022
- Full Text
- View/download PDF
10. Detection of cytogenetic changes and chromosomal aneuploidy with fluorescent in situ hybridization in cytological specimens of oral cancers in Fanconi anemia-Proof of concept.
- Author
-
Silva de Araujo BE, Velleuer E, Dietrich R, Pomjanski N, de Santana Almeida Araujo IK, Schlensog M, Wells SI, Dorsman JC, and Schramm M
- Subjects
- Aneuploidy, Chromosomes, Humans, In Situ Hybridization, Fluorescence methods, Carcinoma, Squamous Cell genetics, Carcinoma, Squamous Cell pathology, Fanconi Anemia genetics, Head and Neck Neoplasms, Mouth Neoplasms pathology
- Abstract
Objectives: Fanconi anemia (FA) is a rare inherited DNA instability disorder with a remarkably elevated risk of neoplasia compared with the general population, mainly leukemia and squamous cell carcinoma (SCC). Two thirds of the SCCs arise in the oral cavity and are typically preceded by visible lesions. These lesions can be classified with brush biopsy-based cytological methods regarding their risk of a malignant transformation. As a proof of concept, this study aims to investigate genetic changes and chromosomal aneuploidy using fluorescent in situ hybridization (FISH) on oral squamous cells derived from FA affected individuals., Material and Methods: Five FA oral SCC (OSCC) tumor cell lines, one FA OSCC cervical lymph node metastasis as well as tumor-negative and atypical smears from oral brush biopsies were analyzed with FISH probes covering 5p15.2, MYC, EGFR, TERC, 9q34.1, CCND1, 9p21 and centromeres of chromosomes 3, 6, 7, 9, 11, and 17., Results: OSCC specimens showed gains of all analyzed chromosomal regions. Chromosomal aneuploidy was observed in five of the six OSCC specimens in two multicolor FISH assays with panels of four probes each. Five out of six OSCC specimens displayed a relative deletion of 9p21. Applied on atypical brush biopsy-based smears, chromosomal aneuploidy was detected in malignant lesions but not in the smear derived from a severe parodontitis., Conclusions: As proof of concept, FISH was able to detect genetic changes and chromosomal aneuploidy discriminating oral cancer from noncancerous lesions in individuals with FA. This supports its application on oral brush biopsy-based cytology., (© 2021 The Authors. Clinical and Experimental Dental Research published by John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
11. ELOF1 is a transcription-coupled DNA repair factor that directs RNA polymerase II ubiquitylation.
- Author
-
van der Weegen Y, de Lint K, van den Heuvel D, Nakazawa Y, Mevissen TET, van Schie JJM, San Martin Alonso M, Boer DEC, González-Prieto R, Narayanan IV, Klaassen NHM, Wondergem AP, Roohollahi K, Dorsman JC, Hara Y, Vertegaal ACO, de Lange J, Walter JC, Noordermeer SM, Ljungman M, Ogi T, Wolthuis RMF, and Luijsterburg MS
- Subjects
- CRISPR-Cas Systems, Cell Line, Tumor, DNA Helicases genetics, DNA Helicases metabolism, DNA Repair Enzymes genetics, DNA Repair Enzymes metabolism, Humans, Peptide Elongation Factor 1 genetics, Poly-ADP-Ribose Binding Proteins genetics, Poly-ADP-Ribose Binding Proteins metabolism, Protein Binding, Protein Interaction Domains and Motifs, RNA Polymerase II genetics, Transcription Elongation, Genetic, Transcription Factors genetics, Transcription Factors metabolism, Ubiquitin-Protein Ligases genetics, DNA Damage, DNA Repair, Peptide Elongation Factor 1 metabolism, RNA Polymerase II metabolism, Ubiquitin-Protein Ligases metabolism, Ubiquitination
- Abstract
Cells employ transcription-coupled repair (TCR) to eliminate transcription-blocking DNA lesions. DNA damage-induced binding of the TCR-specific repair factor CSB to RNA polymerase II (RNAPII) triggers RNAPII ubiquitylation of a single lysine (K1268) by the CRL4
CSA ubiquitin ligase. How CRL4CSA is specifically directed towards K1268 is unknown. Here, we identify ELOF1 as the missing link that facilitates RNAPII ubiquitylation, a key signal for the assembly of downstream repair factors. This function requires its constitutive interaction with RNAPII close to K1268, revealing ELOF1 as a specificity factor that binds and positions CRL4CSA for optimal RNAPII ubiquitylation. Drug-genetic interaction screening also revealed a CSB-independent pathway in which ELOF1 prevents R-loops in active genes and protects cells against DNA replication stress. Our study offers key insights into the molecular mechanisms of TCR and provides a genetic framework of the interplay between transcriptional stress responses and DNA replication.- Published
- 2021
- Full Text
- View/download PDF
12. HSF2BP negatively regulates homologous recombination in DNA interstrand crosslink repair.
- Author
-
Sato K, Brandsma I, van Rossum-Fikkert SE, Verkaik N, Oostra AB, Dorsman JC, van Gent DC, Knipscheer P, Kanaar R, and Zelensky AN
- Subjects
- Animals, BRCA2 Protein metabolism, Cell Line, DNA Damage, Fanconi Anemia genetics, Humans, Mice, Protein Binding, Proteolysis, Rad51 Recombinase metabolism, Xenopus, Carrier Proteins metabolism, Cell Cycle Proteins metabolism, DNA Repair, Heat-Shock Proteins metabolism, Homologous Recombination
- Abstract
The tumor suppressor BRCA2 is essential for homologous recombination (HR), replication fork stability and DNA interstrand crosslink (ICL) repair in vertebrates. We show that ectopic production of HSF2BP, a BRCA2-interacting protein required for meiotic HR during mouse spermatogenesis, in non-germline human cells acutely sensitize them to ICL-inducing agents (mitomycin C and cisplatin) and PARP inhibitors, resulting in a phenotype characteristic of cells from Fanconi anemia (FA) patients. We biochemically recapitulate the suppression of ICL repair and establish that excess HSF2BP compromises HR by triggering the removal of BRCA2 from the ICL site and thereby preventing the loading of RAD51. This establishes ectopic expression of a wild-type meiotic protein in the absence of any other protein-coding mutations as a new mechanism that can lead to an FA-like cellular phenotype. Naturally occurring elevated production of HSF2BP in tumors may be a source of cancer-promoting genomic instability and also a targetable vulnerability., (© The Author(s) 2020. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2020
- Full Text
- View/download PDF
13. Effective CRISPR/Cas9-mediated correction of a Fanconi anemia defect by error-prone end joining or templated repair.
- Author
-
van de Vrugt HJ, Harmsen T, Riepsaame J, Alexantya G, van Mil SE, de Vries Y, Bin Ali R, Huijbers IJ, Dorsman JC, Wolthuis RMF, and Te Riele H
- Subjects
- Animals, Cells, Cultured, DNA Repair, Ear, Fibroblasts, Mice, Mouse Embryonic Stem Cells, CRISPR-Cas Systems genetics, Fanconi Anemia genetics, Fanconi Anemia therapy, Fanconi Anemia Complementation Group F Protein genetics, Gene Editing methods, Genetic Therapy methods
- Abstract
Fanconi anemia (FA) is a cancer predisposition syndrome characterized by congenital abnormalities, bone marrow failure, and hypersensitivity to aldehydes and crosslinking agents. For FA patients, gene editing holds promise for therapeutic applications aimed at functionally restoring mutated genes in hematopoietic stem cells. However, intrinsic FA DNA repair defects may obstruct gene editing feasibility. Here, we report on the CRISPR/Cas9-mediated correction of a disruptive mutation in Fancf. Our experiments revealed that gene editing could effectively restore Fancf function via error-prone end joining resulting in a 27% increased survival in the presence of mitomycin C. In addition, templated gene correction could be achieved after double strand or single strand break formation. Although templated gene editing efficiencies were low (≤6%), FA corrected embryonic stem cells acquired a strong proliferative advantage over non-corrected cells, even without imposing genotoxic stress. Notably, Cas9 nickase activity resulted in mono-allelic gene editing and avoidance of undesired mutagenesis. In conclusion: DNA repair defects associated with FANCF deficiency do not prohibit CRISPR/Cas9 gene correction. Our data provide a solid basis for the application of pre-clinical models to further explore the potential of gene editing against FA, with the eventual aim to obtain therapeutic strategies against bone marrow failure.
- Published
- 2019
- Full Text
- View/download PDF
14. Loss of p53 suppresses replication-stress-induced DNA breakage in G1/S checkpoint deficient cells.
- Author
-
Benedict B, van Harn T, Dekker M, Hermsen S, Kucukosmanoglu A, Pieters W, Delzenne-Goette E, Dorsman JC, Petermann E, Foijer F, and Te Riele H
- Subjects
- Animals, Apoptosis genetics, Cell Cycle Checkpoints genetics, Cell Line, Tumor, Cyclin-Dependent Kinase Inhibitor p21 genetics, DNA Breaks, Double-Stranded, DNA Repair genetics, Humans, Mice, Neoplasms pathology, S Phase genetics, Teratoma genetics, Teratoma pathology, DNA Damage genetics, DNA Replication genetics, Neoplasms genetics, Tumor Suppressor Protein p53 genetics
- Abstract
In cancer cells, loss of G1/S control is often accompanied by p53 pathway inactivation, the latter usually rationalized as a necessity for suppressing cell cycle arrest and apoptosis. However, we found an unanticipated effect of p53 loss in mouse and human G1-checkpoint-deficient cells: reduction of DNA damage. We show that abrogation of the G1/S-checkpoint allowed cells to enter S-phase under growth-restricting conditions at the expense of severe replication stress manifesting as decelerated DNA replication, reduced origin firing and accumulation of DNA double-strand breaks. In this system, loss of p53 allowed mitogen-independent proliferation, not by suppressing apoptosis, but rather by restoring origin firing and reducing DNA breakage. Loss of G1/S control also caused DNA damage and activation of p53 in an in vivo retinoblastoma model. Moreover, in a teratoma model, loss of p53 reduced DNA breakage. Thus, loss of p53 may promote growth of incipient cancer cells by reducing replication-stress-induced DNA damage., Competing Interests: BB, Tv, MD, SH, AK, WP, ED, JD, EP, FF, Ht No competing interests declared, (© 2018, Benedict et al.)
- Published
- 2018
- Full Text
- View/download PDF
15. MR Imaging Features of Retinoblastoma: Association with Gene Expression Profiles.
- Author
-
Jansen RW, de Jong MC, Kooi IE, Sirin S, Göricke S, Brisse HJ, Maeder P, Galluzzi P, van der Valk P, Cloos J, Eekhout I, Castelijns JA, Moll AC, Dorsman JC, and de Graaf P
- Subjects
- Child, Preschool, Female, Humans, Infant, Male, Reproducibility of Results, Retina diagnostic imaging, Retinal Neoplasms genetics, Retinoblastoma genetics, Genes, Retinoblastoma genetics, Magnetic Resonance Imaging methods, Retinal Neoplasms diagnostic imaging, Retinoblastoma diagnostic imaging, Transcriptome genetics
- Abstract
Purpose To identify associations between magnetic resonance (MR) imaging features and gene expression in retinoblastoma. Materials and Methods A retinoblastoma MR imaging atlas was validated by using anonymized MR images from referral centers in Essen, Germany, and Paris, France. Images were from 39 patients with retinoblastoma (16 male and 18 female patients [the sex in five patients was unknown]; age range, 5-90 months; inclusion criterion: pretreatment MR imaging). This atlas was used to compare MR imaging features with genome-wide messenger RNA (mRNA) expression data from 60 consecutive patients obtained from 1995 to 2012 (35 male patients [58%]; age range, 2-69 months; inclusion criteria: pretreatment MR imaging, genome-wide mRNA expression data available). Imaging pathway associations were analyzed by means of gene enrichment. In addition, imaging features were compared with a predefined gene expression signature of photoreceptorness. Statistical analysis was performed with generalized linear modeling of radiology traits on normalized log2-transformed expression values. P values were corrected for multiple hypothesis testing. Results Radiogenomic analysis revealed 1336 differentially expressed genes for qualitative imaging features (threshold P = .05 after multiple hypothesis correction). Loss of photoreceptorness gene expression correlated with advanced stage imaging features, including multiple lesions (P = .03) and greater eye size (P < .001). The number of lesions on MR images was associated with expression of MYCN (P = .04). A newly defined radiophenotype of diffuse-growing, plaque-shaped, multifocal tumors displayed overexpression of SERTAD3 (P = .003, P = .049, and P = .06, respectively), a protein that stimulates cell growth by activating the E2F network. Conclusion Radiogenomic biomarkers can potentially help predict molecular features, such as photoreceptorness loss, that indicate tumor progression. Results imply a possible role for radiogenomics in future staging and treatment decision making in retinoblastoma., (© RSNA, 2018 Online supplemental material is available for this article.)
- Published
- 2018
- Full Text
- View/download PDF
16. Non-invasive tumor genotyping using radiogenomic biomarkers, a systematic review and oncology-wide pathway analysis.
- Author
-
Jansen RW, van Amstel P, Martens RM, Kooi IE, Wesseling P, de Langen AJ, Menke-Van der Houven van Oordt CW, Jansen BHE, Moll AC, Dorsman JC, Castelijns JA, de Graaf P, and de Jong MC
- Abstract
With targeted treatments playing an increasing role in oncology, the need arises for fast non-invasive genotyping in clinical practice. Radiogenomics is a rapidly evolving field of research aimed at identifying imaging biomarkers useful for non-invasive genotyping. Radiogenomic genotyping has the advantage that it can capture tumor heterogeneity, can be performed repeatedly for treatment monitoring, and can be performed in malignancies for which biopsy is not available. In this systematic review of 187 included articles, we compiled a database of radiogenomic associations and unraveled networks of imaging groups and gene pathways oncology-wide. Results indicated that ill-defined tumor margins and tumor heterogeneity can potentially be used as imaging biomarkers for 1p/19q codeletion in glioma, relevant for prognosis and disease profiling. In non-small cell lung cancer, FDG-PET uptake and CT-ground-glass-opacity features were associated with treatment-informing traits including EGFR -mutations and ALK -rearrangements. Oncology-wide gene pathway analysis revealed an association between contrast enhancement (imaging) and the targetable VEGF-signalling pathway. Although the need of independent validation remains a concern, radiogenomic biomarkers showed potential for prognosis prediction and targeted treatment selection. Quantitative imaging enhanced the potential of multiparametric radiogenomic models. A wealth of data has been compiled for guiding future research towards robust non-invasive genomic profiling., Competing Interests: CONFLICTS OF INTEREST The authors declare no conflicts of interests.
- Published
- 2018
- Full Text
- View/download PDF
17. Genomic landscape of retinoblastoma in Rb -/- p130 -/- mice resembles human retinoblastoma.
- Author
-
Kooi IE, van Mil SE, MacPherson D, Mol BM, Moll AC, Meijers-Heijboer H, Kaspers GJ, Cloos J, Te Riele H, and Dorsman JC
- Subjects
- Animals, Embryonic Stem Cells cytology, Embryonic Stem Cells metabolism, Female, Genome, High-Throughput Nucleotide Sequencing methods, Humans, Male, Mice, Mice, Inbred C57BL, Biomarkers, Tumor genetics, DNA Copy Number Variations genetics, Retinoblastoma genetics, Retinoblastoma-Like Protein p107 physiology, Retinoblastoma-Like Protein p130 physiology
- Abstract
Several murine retinoblastoma models have been generated by deleting the genes encoding for retinoblastoma susceptibility protein pRb and one of its family members p107 or p130. In Rb
-/- p107-/- retinoblastomas, somatic copy number alterations (SCNAs) like Mdm2 amplification or Cdkn2a deletion targeting the p53-pathway occur, which is uncommon for human retinoblastoma. In our study, we determined SCNAs in retinoblastomas developing in Rb-/- p130-/- mice and compared this to murine Rb-/- p107-/- tumors and human tumors. Chimeric mice were made by injection of 129/Ola-derived Rb-/- p130-/- embryonic stem cells into wild type C57BL/6 blastocysts. SCNAs of retinoblastoma samples were determined by low-coverage (∼0.5×) whole genome sequencing. In Rb-/- p130-/- tumors, SCNAs included gain of chromosomes 1 (3/23 tumors), 8 (1/23 tumors), 10 (1/23 tumors), 11 (2/23 tumors), and 12 (4/23 tumors), which could be mapped to frequently altered chromosomes in human retinoblastomas. While the altered chromosomes in Rb-/- p130-/- tumors were similar to those in Rb-/- p107-/- tumors, the alteration frequencies were much lower in Rb-/- p130-/- tumors. Most of the Rb-/- p130-/- tumors (16/23 tumors, 70%) were devoid of SCNAs, in strong contrast to Rb-/- p107-/- tumors, which were never (0/15 tumors) SCNA-devoid. Similarly, to human retinoblastoma, increased age at diagnosis significantly correlated with increased SCNA frequencies. Additionally, focal loss of Cdh11 was observed in one Rb-/- p130-/- tumor, which enforces studies in human retinoblastoma that identified CDH11 as a retinoblastoma suppressor. Moreover, based on a comparison of genes altered in human and murine retinoblastoma, we suggest exploring the role of HMGA1 and SRSF3 in retinoblastoma development. © 2016 Wiley Periodicals, Inc., (© 2016 Wiley Periodicals, Inc.)- Published
- 2017
- Full Text
- View/download PDF
18. Somatic genomic alterations in retinoblastoma beyond RB1 are rare and limited to copy number changes.
- Author
-
Kooi IE, Mol BM, Massink MP, Ameziane N, Meijers-Heijboer H, Dommering CJ, van Mil SE, de Vries Y, van der Hout AH, Kaspers GJ, Moll AC, Te Riele H, Cloos J, and Dorsman JC
- Subjects
- Humans, Sequence Analysis, DNA, Gene Dosage, Mutation, Retinoblastoma pathology, Retinoblastoma Binding Proteins genetics, Ubiquitin-Protein Ligases genetics
- Abstract
Retinoblastoma is a rare childhood cancer initiated by RB1 mutation or MYCN amplification, while additional alterations may be required for tumor development. However, the view on single nucleotide variants is very limited. To better understand oncogenesis, we determined the genomic landscape of retinoblastoma. We performed exome sequencing of 71 retinoblastomas and matched blood DNA. Next, we determined the presence of single nucleotide variants, copy number alterations and viruses. Aside from RB1, recurrent gene mutations were very rare. Only a limited fraction of tumors showed BCOR (7/71, 10%) or CREBBP alterations (3/71, 4%). No evidence was found for the presence of viruses. Instead, specific somatic copy number alterations were more common, particularly in patients diagnosed at later age. Recurrent alterations of chromosomal arms often involved less than one copy, also in highly pure tumor samples, suggesting within-tumor heterogeneity. Our results show that retinoblastoma is among the least mutated cancers and signify the extreme sensitivity of the childhood retina for RB1 loss. We hypothesize that retinoblastomas arising later in retinal development benefit more from subclonal secondary alterations and therefore, these alterations are more selected for in these tumors. Targeted therapy based on these subclonal events might be insufficient for complete tumor control.
- Published
- 2016
- Full Text
- View/download PDF
19. A Meta-Analysis of Retinoblastoma Copy Numbers Refines the List of Possible Driver Genes Involved in Tumor Progression.
- Author
-
Kooi IE, Mol BM, Massink MP, de Jong MC, de Graaf P, van der Valk P, Meijers-Heijboer H, Kaspers GJ, Moll AC, Te Riele H, Cloos J, and Dorsman JC
- Subjects
- Cell Line, Tumor, Disease Progression, Gene Expression Profiling, Humans, Oligonucleotide Array Sequence Analysis, Oncogenes, Retinoblastoma Binding Proteins genetics, Ubiquitin-Protein Ligases genetics, Gene Dosage, Retinal Neoplasms genetics, Retinoblastoma genetics
- Abstract
Background: While RB1 loss initiates retinoblastoma development, additional somatic copy number alterations (SCNAs) can drive tumor progression. Although SCNAs have been identified with good concordance between studies at a cytoband resolution, accurate identification of single genes for all recurrent SCNAs is still challenging. This study presents a comprehensive meta-analysis of genome-wide SCNAs integrated with gene expression profiling data, narrowing down the list of plausible retinoblastoma driver genes., Methods: We performed SCNA profiling of 45 primary retinoblastoma samples and eight retinoblastoma cell lines by high-resolution microarrays. We combined our data with genomic, clinical and histopathological data of ten published genome-wide SCNA studies, which strongly enhanced the power of our analyses (N = 310)., Results: Comprehensive recurrence analysis of SCNAs in all studies integrated with gene expression data allowed us to reduce candidate gene lists for 1q, 2p, 6p, 7q and 13q to a limited gene set. Besides the well-established driver genes RB1 (13q-loss) and MYCN (2p-gain) we identified CRB1 and NEK7 (1q-gain), SOX4 (6p-gain) and NUP205 (7q-gain) as novel retinoblastoma driver candidates. Depending on the sample subset and algorithms used, alternative candidates were identified including MIR181 (1q-gain) and DEK (6p gain). Remarkably, our study showed that copy number gains rarely exceeded change of one copy, even in pure tumor samples with 100% homozygosity at the RB1 locus (N = 34), which is indicative for intra-tumor heterogeneity. In addition, profound between-tumor variability was observed that was associated with age at diagnosis and differentiation grades., Interpretation: Since focal alterations at commonly altered chromosome regions were rare except for 2p24.3 (MYCN), further functional validation of the oncogenic potential of the described candidate genes is now required. For further investigations, our study provides a refined and revised set of candidate retinoblastoma driver genes.
- Published
- 2016
- Full Text
- View/download PDF
20. A novel Fanconi anaemia subtype associated with a dominant-negative mutation in RAD51.
- Author
-
Ameziane N, May P, Haitjema A, van de Vrugt HJ, van Rossum-Fikkert SE, Ristic D, Williams GJ, Balk J, Rockx D, Li H, Rooimans MA, Oostra AB, Velleuer E, Dietrich R, Bleijerveld OB, Maarten Altelaar AF, Meijers-Heijboer H, Joenje H, Glusman G, Roach J, Hood L, Galas D, Wyman C, Balling R, den Dunnen J, de Winter JP, Kanaar R, Gelinas R, and Dorsman JC
- Subjects
- Acid Anhydride Hydrolases, Base Sequence, DNA Damage, DNA Repair, DNA Repair Enzymes metabolism, DNA-Binding Proteins metabolism, Fanconi Anemia genetics, Humans, Male, Molecular Sequence Data, Recombination, Genetic, Young Adult, DNA Repair Enzymes genetics, DNA-Binding Proteins genetics, Fanconi Anemia enzymology, Mutation, Missense
- Abstract
Fanconi anaemia (FA) is a hereditary disease featuring hypersensitivity to DNA cross-linker-induced chromosomal instability in association with developmental abnormalities, bone marrow failure and a strong predisposition to cancer. A total of 17 FA disease genes have been reported, all of which act in a recessive mode of inheritance. Here we report on a de novo g.41022153G>A; p.Ala293Thr (NM_002875) missense mutation in one allele of the homologous recombination DNA repair gene RAD51 in an FA-like patient. This heterozygous mutation causes a novel FA subtype, 'FA-R', which appears to be the first subtype of FA caused by a dominant-negative mutation. The patient, who features microcephaly and mental retardation, has reached adulthood without the typical bone marrow failure and paediatric cancers. Together with the recent reports on RAD51-associated congenital mirror movement disorders, our results point to an important role for RAD51-mediated homologous recombination in neurodevelopment, in addition to DNA repair and cancer susceptibility.
- Published
- 2015
- Full Text
- View/download PDF
21. The iron-sulfur cluster assembly network component NARFL is a key element in the cellular defense against oxidative stress.
- Author
-
Corbin MV, Rockx DA, Oostra AB, Joenje H, and Dorsman JC
- Subjects
- Aconitate Hydratase metabolism, Blotting, Western, Cell Survival, Cytosol metabolism, DNA Helicases metabolism, HeLa Cells, Humans, Hydrogenase antagonists & inhibitors, Hydrogenase genetics, Iron-Sulfur Proteins genetics, Mitochondria metabolism, Protein Interaction Domains and Motifs, RNA, Small Interfering genetics, Transcriptome, Chromosome Breakage, Hydrogenase metabolism, Iron-Sulfur Proteins metabolism, Oxidative Stress physiology
- Abstract
Aim of this study was to explore cellular changes associated with increased resistance to atmospheric oxygen using high-resolution DNA and RNA profiling combined with functional studies. Two independently selected oxygen-resistant substrains of HeLa cells (capable of proliferating at >80% O2, i.e. hyperoxia) were compared with their parental cells (adapted to growth at 20% O2, but unable to grow at >80% O2). A striking consistent alteration found to be associated with the oxygen-resistant state appeared to be an amplified and overexpressed region on chromosome 16p13.3 harboring 21 genes. The driver gene of this amplification was identified by functional studies as NARFL, which encodes a component of the cytosolic iron-sulfur cluster assembly system. In line with this result we found the cytosolic c-aconitase activity as well as the nuclear protein RTEL1, both Fe-S dependent proteins, to be protected by NARFL overexpression under hyperoxia. In addition, we observed a protective effect of NARFL against hyperoxia-induced loss of sister-chromatid cohesion. NARFL thus appeared to be a key factor in the cellular defense against hyperoxia-induced oxidative stress in human cells. Our findings suggest that new insight into age-related degenerative processes may come from studies that specifically address the involvement of iron-sulfur proteins., (Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
22. FANCJ protein is important for the stability of FANCD2/FANCI proteins and protects them from proteasome and caspase-3 dependent degradation.
- Author
-
Clark DW, Tripathi K, Dorsman JC, and Palle K
- Subjects
- Basic-Leucine Zipper Transcription Factors genetics, Cell Line, Tumor, DNA Damage, DNA Repair, Enzyme Stability, Fanconi Anemia drug therapy, Fanconi Anemia genetics, Fanconi Anemia Complementation Group D2 Protein genetics, Fanconi Anemia Complementation Group Proteins genetics, Gene Expression Regulation, Neoplastic, Humans, Hydroxyurea pharmacology, Proteasome Inhibitors pharmacology, Protein Binding, Proteolysis, RNA Interference, Time Factors, Transfection, Ubiquitination, Basic-Leucine Zipper Transcription Factors metabolism, Caspase 3 metabolism, Fanconi Anemia enzymology, Fanconi Anemia Complementation Group D2 Protein metabolism, Fanconi Anemia Complementation Group Proteins metabolism, Proteasome Endopeptidase Complex metabolism
- Abstract
Fanconi anemia (FA) is a rare genome instability syndrome with progressive bone marrow failure and cancer susceptibility. FANCJ is one of 17 genes mutated in FA-patients, comprises a DNA helicase that is vital for properly maintaining genomic stability and is known to function in the FA-BRCA DNA repair pathway. While exact role(s) of FANCJ in this repair process is yet to be determined, it is known to interact with primary effector FANCD2. However, FANCJ is not required for FANCD2 activation but is important for its ability to fully respond to DNA damage. In this report, we determined that transient depletion of FANCJ adversely affects stability of FANCD2 and its co-regulator FANCI in multiple cell lines. Loss of FANCJ does not significantly alter cell cycle progression or FANCD2 transcription. However, in the absence of FANCJ, the majority of FANCD2 is degraded by both the proteasome and Caspase-3 dependent mechanism. FANCJ is capable of complexing with and stabilizing FANCD2 even in the absence of a functional helicase domain. Furthermore, our data demonstrate that FANCJ is important for FANCD2 stability and proper activation of DNA damage responses to replication blocks induced by hydroxyurea.
- Published
- 2015
- Full Text
- View/download PDF
23. Loss of photoreceptorness and gain of genomic alterations in retinoblastoma reveal tumor progression.
- Author
-
Kooi IE, Mol BM, Moll AC, van der Valk P, de Jong MC, de Graaf P, van Mil SE, Schouten-van Meeteren AY, Meijers-Heijboer H, Kaspers GL, Te Riele H, Cloos J, and Dorsman JC
- Subjects
- Child, Preschool, Cluster Analysis, DNA Copy Number Variations genetics, Dactinomycin pharmacology, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic drug effects, Gene Ontology, Humans, Infant, Karyotyping, Male, Photoreceptor Cells, Vertebrate drug effects, RNA, Messenger genetics, RNA, Messenger metabolism, Reproducibility of Results, Retinoblastoma pathology, Disease Progression, Genome, Human, Photoreceptor Cells, Vertebrate metabolism, Retinoblastoma genetics
- Abstract
Background: Retinoblastoma is a pediatric eye cancer associated with RB1 loss or MYCN amplification (RB1 (+/+) MYCN(A) ). There are controversies concerning the existence of molecular subtypes within RB1(-/-) retinoblastoma. To test whether these molecular subtypes exist, we performed molecular profiling., Methods: Genome-wide mRNA expression profiling was performed on 76 primary human retinoblastomas. Expression profiling was complemented by genome-wide DNA profiling and clinical, histopathological, and ex vivo drug sensitivity data., Findings: RNA and DNA profiling identified major variability between retinoblastomas. While gene expression differences between RB1 (+/+) MYCN(A) and RB1(-/-) tumors seemed more dichotomous, differences within the RB1(-/-) tumors were gradual. Tumors with high expression of a photoreceptor gene signature were highly differentiated, smaller in volume and diagnosed at younger age compared with tumors with low photoreceptor signature expression. Tumors with lower photoreceptor expression showed increased expression of genes involved in M-phase and mRNA and ribosome synthesis and increased frequencies of somatic copy number alterations., Interpretation: Molecular, clinical and histopathological differences between RB1(-/-) tumors are best explained by tumor progression, reflected by a gradual loss of differentiation and photoreceptor expression signature. Since copy number alterations were more frequent in tumors with less photoreceptorness, genomic alterations might be drivers of tumor progression., Research in Context: Retinoblastoma is an ocular childhood cancer commonly caused by mutations in the RB1 gene. In order to determine optimal treatment, tumor subtyping is considered critically important. However, except for very rare retinoblastomas without an RB1 mutation, there are controversies as to whether subtypes of retinoblastoma do exist. Our study shows that retinoblastomas are highly diverse but rather than reflecting distinct tumor types with a different etiology, our data suggests that this diversity is a result of tumor progression driven by cumulative genetic alterations. Therefore, retinoblastomas should not be categorized in distinct subtypes, but be described according to their stage of progression.
- Published
- 2015
- Full Text
- View/download PDF
24. Proper genomic profiling of (BRCA1-mutated) basal-like breast carcinomas requires prior removal of tumor infiltrating lymphocytes.
- Author
-
Massink MP, Kooi IE, van Mil SE, Jordanova ES, Ameziane N, Dorsman JC, van Beek DM, van der Voorn JP, Sie D, Ylstra B, van Deurzen CH, Martens JW, Smid M, Sieuwerts AM, de Weerd V, Foekens JA, van den Ouweland AM, van Dyk E, Nederlof PM, Waisfisz Q, and Meijers-Heijboer H
- Subjects
- Cluster Analysis, DNA Copy Number Variations genetics, Female, Flow Cytometry, Gene Dosage, Gene Expression Regulation, Neoplastic, Genomics, Humans, Reproducibility of Results, Sequence Analysis, DNA, BRCA1 Protein genetics, Breast Neoplasms genetics, Breast Neoplasms immunology, Lymphocytes, Tumor-Infiltrating immunology, Mutation genetics
- Abstract
Introduction: BRCA1-mutated breast carcinomas may have distinct biological features, suggesting the involvement of specific oncogenic pathways in tumor development. The identification of genomic aberrations characteristic for BRCA1-mutated breast carcinomas could lead to a better understanding of BRCA1-associated oncogenic events and could prove valuable in clinical testing for BRCA1-involvement in patients., Methods: For this purpose, genomic and gene expression profiles of basal-like BRCA1-mutated breast tumors (n = 27) were compared with basal-like familial BRCAX (non-BRCA1/2/CHEK2*1100delC) tumors (n = 14) in a familial cohort of 120 breast carcinomas., Results: Genome wide copy number profiles of the BRCA1-mutated breast carcinomas in our data appeared heterogeneous. Gene expression analyses identified varying amounts of tumor infiltrating lymphocytes (TILs) as a major cause for this heterogeneity. Indeed, selecting tumors with relative low amounts of TILs, resulted in the identification of three known but also five previously unrecognized BRCA1-associated copy number aberrations. Moreover, these aberrations occurred with high frequencies in the BRCA1-mutated tumor samples. Using these regions it was possible to discriminate BRCA1-mutated from BRCAX breast carcinomas, and they were validated in two independent cohorts. To further substantiate our findings, we used flow cytometry to isolate cancer cells from formalin-fixed, paraffin-embedded, BRCA1-mutated triple negative breast carcinomas with estimated TIL percentages of 40% and higher. Genomic profiles of sorted and unsorted fractions were compared by shallow whole genome sequencing and confirm our findings., Conclusion: This study shows that genomic profiling of in particular basal-like, and thus BRCA1-mutated, breast carcinomas is severely affected by the presence of high numbers of TILs. Previous reports on genomic profiling of BRCA1-mutated breast carcinomas have largely neglected this. Therefore, our findings have direct consequences on the interpretation of published genomic data. Also, these findings could prove valuable in light of currently used genomic tools for assessing BRCA1-involvement in breast cancer patients and pathogenicity assessment of BRCA1 variants of unknown significance. The BRCA1-associated genomic aberrations identified in this study provide possible leads to a better understanding of BRCA1-associated oncogenesis., (Copyright © 2015 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
25. A reason for intermittent fasting to suppress the awakening of dormant breast tumors.
- Author
-
Lankelma J, Kooi B, Krab K, Dorsman JC, Joenje H, and Westerhoff HV
- Subjects
- Blood Glucose physiology, Female, Humans, Breast Neoplasms physiopathology, Cell Hypoxia physiology, Fasting physiology, Models, Biological, Neoplasm Recurrence, Local prevention & control, Neovascularization, Pathologic physiopathology
- Abstract
For their growth, dormant tumors, which lack angiogenesis may critically depend on gradients of nutrients and oxygen from the nearest blood vessel. Because for oxygen depletion the distance from the nearest blood vessel to depletion will generally be shorter than for glucose depletion, such tumors will contain anoxic living tumor cells. These cells are dangerous, because they are capable of inducing angiogenesis, which will "wake up" the tumor. Anoxic cells are dependent on anaerobic glucose breakdown for ATP generation. The local extracellular glucose concentration gradient is determined by the blood glucose concentration and by consumption by cells closer to the nearest blood vessel. The blood glucose concentration can be lowered by 20-40% during fasting. We calculated that glucose supply to the potentially hazardous anoxic cells can thereby be reduced significantly, resulting in cell death specifically of the anoxic tumor cells. We hypothesize that intermittent fasting will help to reduce the incidence of tumor relapse via reducing the number of anoxic tumor cells and tumor awakening., (Copyright © 2014 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
26. Coregulation of FANCA and BRCA1 in human cells.
- Author
-
Haitjema A, Mol BM, Kooi IE, Massink MP, Jørgensen JA, Rockx DA, Rooimans MA, de Winter JP, Meijers-Heijboer H, Joenje H, and Dorsman JC
- Abstract
Fanconi anemia (FA) is a genetically heterogeneous syndrome associated with increased cancer predisposition. The underlying genes govern the FA pathway which functions to protect the genome during the S-phase of the cell cycle. While upregulation of FA genes has been linked to chemotherapy resistance, little is known about their regulation in response to proliferative stimuli. The purpose of this study was to examine how FA genes are regulated, especially in relation to the cell cycle, in order to reveal their possible participation in biochemical networks. Expression of 14 FA genes was monitored in two human cell-cycle models and in two RB1/E2F pathway-associated primary cancers, retinoblastoma and basal breast cancer. In silico studies were performed to further evaluate coregulation and identify connected networks and diseases. Only FANCA was consistently induced over 2-fold; FANCF failed to exhibit any regulatory fluctuations. Two tools exploiting public data sets indicated coregulation of FANCA with BRCA1. Upregulation of FANCA and BRCA1 correlated with upregulation of E2F3. Genes coregulated with both FANCA and BRCA1 were enriched for MeSH-Term id(s) genomic instability, microcephaly, and Bloom syndrome, and enriched for the cellular component centrosome. The regulation of FA genes appears highly divergent. In RB1-linked tumors, upregulation of FA network genes was associated with reduced expression of FANCF. FANCA and BRCA1 may jointly act in a subnetwork - supporting vital function(s) at the subcellular level (centrosome) as well as at the level of embryonic development (mechanisms controlling head circumference).
- Published
- 2014
- Full Text
- View/download PDF
27. RB1 mutation spectrum in a comprehensive nationwide cohort of retinoblastoma patients.
- Author
-
Dommering CJ, Mol BM, Moll AC, Burton M, Cloos J, Dorsman JC, Meijers-Heijboer H, and van der Hout AH
- Subjects
- Child, Preschool, Chromosome Deletion, Cohort Studies, DNA Mutational Analysis methods, Female, Humans, Male, Netherlands epidemiology, Pedigree, Retinoblastoma epidemiology, Retinoblastoma genetics, Retinoblastoma Protein genetics
- Abstract
Background: Retinoblastoma (Rb) is a childhood cancer of the retina, commonly initiated by biallelic inactivation of the RB1 gene. Knowledge of the presence of a heritable RB1 mutation can help in risk management and reproductive decision making. We report here on RB1 mutation scanning in a unique nationwide cohort of Rb patients from the Netherlands., Methods: From the 1173 Rb patients registered in the Dutch National Retinoblastoma Register until January 2013, 529 patients from 433 unrelated families could be included. RB1 mutation scanning was performed with different detection methods, depending on the time period., Results: Our mutation detection methods revealed RB1 mutations in 92% of bilateral and/or familial Rb patients and in 10% of non-familial unilateral cases. Overall an RB1 germline mutation was detected in 187 (43%) of 433 Rb families, including 33 novel mutations. The distribution of the type of mutation was 37% nonsense, 20% frameshift, 21% splice, 9% large indel, 5% missense, 7% chromosomal deletions and 1% promoter. Ten per cent of patients were mosaic for the RB1 mutation. Six three-generation families with incomplete penetrance RB1 mutations were found. We found evidence that two variants, previously described as pathogenic RB1 mutations, are likely to be neutral variants., Conclusions: The frequency of the type of mutations in the RB1 gene in our unbiased national cohort is the same as the mutation spectrum described worldwide. Furthermore, our RB1 mutation detection regimen achieves a high scanning sensitivity., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://group.bmj.com/group/rights-licensing/permissions.)
- Published
- 2014
- Full Text
- View/download PDF
28. High resolution SNP array profiling identifies variability in retinoblastoma genome stability.
- Author
-
Mol BM, Massink MP, van der Hout AH, Dommering CJ, Zaman JM, Bosscha MI, Kors WA, Meijers-Heijboer HE, Kaspers GJ, Riele Ht, Moll AC, Cloos J, and Dorsman JC
- Subjects
- Child, Preschool, Chromosomes, Human, Pair 13 genetics, Chromosomes, Human, Pair 14 genetics, Cluster Analysis, Female, Gene Dosage, Genes, Retinoblastoma, Humans, Infant, Loss of Heterozygosity, Male, Oligonucleotide Array Sequence Analysis, Genomic Instability, Polymorphism, Single Nucleotide, Retinal Neoplasms genetics, Retinoblastoma genetics
- Abstract
Both hereditary and nonhereditary retinoblastoma (Rb) are commonly initiated by loss of both copies of the retinoblastoma tumor suppressor gene (RB1), while additional genomic changes are required for tumor initiation and progression. Our aim was to determine whether there is genomic heterogeneity between different clinical Rb subtypes. Therefore, 21 Rb tumors from 11 hereditary patients and 10 nonhereditary Rb patients were analyzed using high-resolution single nucleotide polymorphism (SNP) arrays and gene losses and gains were validated with Multiplex Ligation-dependent Probe Amplification. In these tumors only a few focal aberrations were detected. The most frequent was a focal gain on chromosome 2p24.3, the minimal region of gain encompassing the oncogene MYCN. The genes BAZ1A, OTX2, FUT8, and AKT1 were detected in four focal regions on chromosome 14 in one nonhereditary Rb. There was a large difference in number of copy number aberrations between tumors. A subset of nonhereditary Rbs turned out to be the most genomic unstable, while especially very young patients with hereditary Rb display stable genomes. Established Rb copy number aberrations, including gain of chromosome arm 1q and loss of chromosome arm 16q, turned out to be preferentially associated with the nonhereditary Rbs with later age of diagnosis. In contrast, copy number neutral loss of heterozygosity was detected mainly on chromosome 13, where RB1 resides, irrespective of hereditary status or age. Focal amplifications and deletions and copy number neutral loss of heterozygosity besides chromosome 13 appear to be rare events in retinoblastoma., (Copyright © 2013 Wiley Periodicals, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
29. A protein prioritization approach tailored for the FA/BRCA pathway.
- Author
-
Haitjema A, Brandt BW, Ameziane N, May P, Heringa J, de Winter JP, Joenje H, and Dorsman JC
- Subjects
- Cell Cycle genetics, Fanconi Anemia genetics, Fanconi Anemia metabolism, Haploinsufficiency genetics, Humans, Molecular Sequence Annotation, Proteome, Reproducibility of Results, Computational Biology methods, Fanconi Anemia Complementation Group Proteins metabolism, Signal Transduction, Tumor Suppressor Proteins metabolism
- Abstract
Fanconi anemia (FA) is a heterogeneous recessive disorder associated with a markedly elevated risk to develop cancer. To date sixteen FA genes have been identified, three of which predispose heterozygous mutation carriers to breast cancer. The FA proteins work together in a genome maintenance pathway, the so-called FA/BRCA pathway which is important during the S phase of the cell cycle. Since not all FA patients can be linked to (one of) the sixteen known complementation groups, new FA genes remain to be identified. In addition the complex FA network remains to be further unravelled. One of the FA genes, FANCI, has been identified via a combination of bioinformatic techniques exploiting FA protein properties and genetic linkage. The aim of this study was to develop a prioritization approach for proteins of the entire human proteome that potentially interact with the FA/BRCA pathway or are novel candidate FA genes. To this end, we combined the original bioinformatics approach based on the properties of the first thirteen FA proteins identified with publicly available tools for protein-protein interactions, literature mining (Nermal) and a protein function prediction tool (FuncNet). Importantly, the three newest FA proteins FANCO/RAD51C, FANCP/SLX4, and XRCC2 displayed scores in the range of the already known FA proteins. Likewise, a prime candidate FA gene based on next generation sequencing and having a very low score was subsequently disproven by functional studies for the FA phenotype. Furthermore, the approach strongly enriches for GO terms such as DNA repair, response to DNA damage stimulus, and cell cycle-regulated genes. Additionally, overlaying the top 150 with a haploinsufficiency probability score, renders the approach more tailored for identifying breast cancer related genes. This approach may be useful for prioritization of putative novel FA or breast cancer genes from next generation sequencing efforts.
- Published
- 2013
- Full Text
- View/download PDF
30. Characterisation of retinoblastomas without RB1 mutations: genomic, gene expression, and clinical studies.
- Author
-
Rushlow DE, Mol BM, Kennett JY, Yee S, Pajovic S, Thériault BL, Prigoda-Lee NL, Spencer C, Dimaras H, Corson TW, Pang R, Massey C, Godbout R, Jiang Z, Zacksenhaus E, Paton K, Moll AC, Houdayer C, Raizis A, Halliday W, Lam WL, Boutros PC, Lohmann D, Dorsman JC, and Gallie BL
- Subjects
- Alleles, Cell Line, Tumor, Child, Child, Preschool, Female, Gene Amplification, Gene Expression Regulation, Neoplastic, Genome, Human, Humans, Infant, Mutation, N-Myc Proto-Oncogene Protein, Polymorphism, Single Nucleotide, Gene Dosage, Nuclear Proteins genetics, Nuclear Proteins metabolism, Oncogene Proteins genetics, Oncogene Proteins metabolism, Retinoblastoma genetics, Retinoblastoma metabolism, Retinoblastoma pathology, Retinoblastoma Protein genetics, Retinoblastoma Protein metabolism
- Abstract
Background: Retinoblastoma is the childhood retinal cancer that defined tumour-suppressor genes. Previous work shows that mutation of both alleles of the RB1 retinoblastoma suppressor gene initiates disease. We aimed to characterise non-familial retinoblastoma tumours with no detectable RB1 mutations., Methods: Of 1068 unilateral non-familial retinoblastoma tumours, we compared those with no evidence of RB1 mutations (RB1(+/+)) with tumours carrying a mutation in both alleles (RB1(-/-)). We analysed genomic copy number, RB1 gene expression and protein function, retinal gene expression, histological features, and clinical data., Findings: No RB1 mutations (RB1(+/+)) were reported in 29 (2·7%) of 1068 unilateral retinoblastoma tumours. 15 of the 29 RB1(+/+) tumours had high-level MYCN oncogene amplification (28-121 copies; RB1(+/+)MYCN(A)), whereas none of 93 RB1(-/-) primary tumours tested showed MYCN amplification (p<0·0001). RB1(+/+)MYCN(A) tumours expressed functional RB1 protein, had fewer overall genomic copy-number changes in genes characteristic of retinoblastoma than did RB1(-/-) tumours, and showed distinct aggressive histological features. MYCN amplification was the sole copy-number change in one RB1(+/+)MYCN(A) retinoblastoma. One additional MYCN(A) tumour was discovered after the initial frequencies were determined, and this is included in further analyses. Median age at diagnosis of the 17 children with RB1(+/+)MYCN(A) tumours was 4·5 months (IQR 3·5-10), compared with 24 months (15-37) for 79 children with non-familial unilateral RB1(-/-) retinoblastoma., Interpretation: Amplification of the MYCN oncogene might initiate retinoblastoma in the presence of non-mutated RB1 genes. These unilateral RB1(+/+)MYCN(A) retinoblastomas are characterised by distinct histological features, only a few of the genomic copy-number changes that are characteristic of retinoblastoma, and very early age of diagnosis., Funding: National Cancer Institute-National Institutes of Health, Canadian Institutes of Health Research, German Research Foundation, Canadian Retinoblastoma Society, Hyland Foundation, Toronto Netralaya and Doctors Lions Clubs, Ontario Ministry of Health and Long Term Care, UK-Essen, and Foundations Avanti-STR and KiKa., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
31. Huntingtin with an expanded polyglutamine repeat affects the Jab1-p27(Kip1) pathway.
- Author
-
Cong SY, Pepers BA, Zhou TT, Kerkdijk H, Roos RA, van Ommen GJ, and Dorsman JC
- Subjects
- Blotting, Western, COP9 Signalosome Complex, Cells, Cultured, DNA Repeat Expansion, Fluorescent Antibody Technique, HeLa Cells, Humans, Huntingtin Protein, Huntington Disease metabolism, Huntington Disease pathology, Luciferases metabolism, Mutation genetics, Mutation physiology, PC12 Cells, Plasmids genetics, Polymerase Chain Reaction, Signal Transduction drug effects, Cyclin-Dependent Kinase Inhibitor p27 drug effects, Intracellular Signaling Peptides and Proteins drug effects, Nerve Tissue Proteins genetics, Nerve Tissue Proteins pharmacology, Peptide Hydrolases drug effects, Peptides genetics, Peptides pharmacology
- Abstract
Expansion of polyglutamine repeats is the cause of at least nine inherited human neurodegenerative disorders, including Huntington's disease (HD). It is widely accepted that deregulation of the transcriptional coactivator CBP by expanded huntingtin (htt) plays an important role in HD molecular pathogenesis. In this study, we report on a novel target of expanded polyglutamine stretches, the transcriptional coactivator Jun activation domain-binding protein 1 (Jab1), which shares DNA-sequence-specific transcription factor targets with CBP. Jab1 also plays a major role in the degradation of the cyclin-dependent-kinase inhibitor and putative transcription cofactor p27(Kip1). We found that Jab1 accumulates in aggregates when co-expressed with either expanded polyglutamine stretches or N-terminal fragments of mutant htt. In addition, the coactivator function of Jab1 was suppressed both by aggregated expanded polyglutamine solely and by mutant htt. Inhibition by mutant htt even preceded the appearance of microscopic aggregation. In an exon 1 HD cell model, we found that endogenous Jab1 could be recruited into aggregates and that this was accompanied by the accumulation of p27(Kip1). Accumulation of p27(Kip1) was also found in brains derived from HD patients. The repression of Jab1 by various mechanisms coupled with an increase of p27(Kip1) at late stages may have important transcriptional effects. In addition, the interference with the Jab1-p27(Kip1) pathway may contribute to the observed lower incidence of cancer in HD patients and may also be relevant for the understanding of the molecular pathogenesis of polyglutamine disorders in general., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
32. Diagnosis of fanconi anemia: mutation analysis by next-generation sequencing.
- Author
-
Ameziane N, Sie D, Dentro S, Ariyurek Y, Kerkhoven L, Joenje H, Dorsman JC, Ylstra B, Gille JJ, Sistermans EA, and de Winter JP
- Abstract
Fanconi anemia (FA) is a rare genetic instability syndrome characterized by developmental defects, bone marrow failure, and a high cancer risk. Fifteen genetic subtypes have been distinguished. The majority of patients (≈85%) belong to the subtypes A (≈60%), C (≈15%) or G (≈10%), while a minority (≈15%) is distributed over the remaining 12 subtypes. All subtypes seem to fit within the "classical" FA phenotype, except for D1 and N patients, who have more severe clinical symptoms. Since FA patients need special clinical management, the diagnosis should be firmly established, to exclude conditions with overlapping phenotypes. A valid FA diagnosis requires the detection of pathogenic mutations in a FA gene and/or a positive result from a chromosomal breakage test. Identification of the pathogenic mutations is also important for adequate genetic counselling and to facilitate prenatal or preimplantation genetic diagnosis. Here we describe and validate a comprehensive protocol for the molecular diagnosis of FA, based on massively parallel sequencing. We used this approach to identify BRCA2, FANCD2, FANCI and FANCL mutations in novel unclassified FA patients.
- Published
- 2012
- Full Text
- View/download PDF
33. Array comparative genomic hybridization analysis indicates that serous carcinomas of the ovary, fallopian tube and endometrium are distinct entities.
- Author
-
Nowee ME, Seeber LM, Horrée N, Snijders AM, Van Diest PJ, Verheijen RH, and Dorsman JC
- Subjects
- Chromosomes, Artificial, Bacterial, Comparative Genomic Hybridization, Female, Gene Expression Profiling, Humans, Microarray Analysis, Biomarkers, Tumor genetics, Cystadenocarcinoma, Serous genetics, Endometrial Neoplasms genetics, Fallopian Tube Neoplasms genetics, Ovarian Neoplasms genetics
- Published
- 2010
- Full Text
- View/download PDF
34. Lack of large genomic deletions in BRIP1, PALB2, and FANCD2 genes in BRCA1/2 negative familial breast cancer.
- Author
-
Ameziane N, van den Ouweland AM, Adank MA, Vijzelaar RN, Errami A, Dorsman JC, Joenje H, Meijers-Heijboer H, and Waisfisz Q
- Subjects
- Fanconi Anemia Complementation Group N Protein, Fanconi Anemia Complementation Group Proteins, Female, Gene Deletion, Genes, BRCA1, Genes, BRCA2, Humans, Netherlands, Breast Neoplasms genetics, DNA-Binding Proteins genetics, Fanconi Anemia Complementation Group D2 Protein genetics, Genetic Predisposition to Disease, Nuclear Proteins genetics, RNA Helicases genetics, Tumor Suppressor Proteins genetics
- Published
- 2009
- Full Text
- View/download PDF
35. v-Raf murine sarcoma viral oncogene mutation status in serous borderline ovarian tumors and the effect on clinical behavior.
- Author
-
Verbruggen MB, Sieben NL, Roemen GM, Rockx DA, van Diest PJ, Verheijen RH, and Dorsman JC
- Subjects
- Adult, Cystadenocarcinoma, Serous pathology, DNA Mutational Analysis, Disease Progression, Female, Follow-Up Studies, Genetic Predisposition to Disease, HT29 Cells, Humans, Middle Aged, Mutation physiology, Ovarian Neoplasms pathology, Prognosis, Retrospective Studies, Cystadenocarcinoma, Serous diagnosis, Cystadenocarcinoma, Serous genetics, Ovarian Neoplasms diagnosis, Ovarian Neoplasms genetics, Proto-Oncogene Proteins B-raf genetics
- Abstract
Aims: To determine the incidence of activating v-raf murine sarcoma viral oncogene (BRAF) mutations in 30 serous borderline tumors (SBTs) of the ovary and the accompanying implants and to link BRAF mutation status to the clinical behavior of these tumors., Methods and Results: Serous borderline tumors and noninvasive implants of 30 patients were analyzed for the presence of the BRAF V599E mutation, and mutation status was correlated to 70 months of clinical follow-up. Mutation status could be assessed in 27 SBTs. Eleven (41%) showed a BRAF mulation. Four (80%) of 5 patients with bilateral SBT showed a BRAF mutation in both ovaries. From the 8 implants that were analyzed for BRAF, 2 (25%) were mutated together with their primary tumor. v-Raf murine sarcoma viral oncogene mutation positive SBTs tend to present with a lower International Federation of Gynecology and Obstetrics stage and a higher tumor volume and are less frequently aneuploid. Seventy months' follow-up indicated no significant recurrence-free survival difference between these groups., Conclusions: v-Raf murine sarcoma viral oncogene mutations are common in ovarian SBT, are strongly associated with bilateral tumors, and are also found in implants. A larger number of tumors should be investigated to assess clinical importance of BRAF mutation status in SBTs.
- Published
- 2009
- Full Text
- View/download PDF
36. Mutant huntingtin activates Nrf2-responsive genes and impairs dopamine synthesis in a PC12 model of Huntington's disease.
- Author
-
van Roon-Mom WM, Pepers BA, 't Hoen PA, Verwijmeren CA, den Dunnen JT, Dorsman JC, and van Ommen GB
- Subjects
- Animals, Disease Models, Animal, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Huntingtin Protein, Huntington Disease metabolism, NF-E2-Related Factor 2 genetics, Nerve Tissue Proteins metabolism, Nuclear Proteins metabolism, Oxidative Stress, PC12 Cells, RNA, Messenger metabolism, Rats, Up-Regulation, Dopamine biosynthesis, Huntington Disease genetics, Mutation, NF-E2-Related Factor 2 metabolism, Nerve Tissue Proteins genetics, Nuclear Proteins genetics
- Abstract
Background: Huntington's disease is a progressive autosomal dominant neurodegenerative disorder that is caused by a CAG repeat expansion in the HD or Huntington's disease gene. Although micro array studies on patient and animal tissue provide valuable information, the primary effect of mutant huntingtin will inevitably be masked by secondary processes in advanced stages of the disease. Thus, cell models are instrumental to study early, direct effects of mutant huntingtin. mRNA changes were studied in an inducible PC12 model of Huntington's disease, before and after aggregates became visible, to identify groups of genes that could play a role in the early pathology of Huntington's disease., Results: Before aggregation, up-regulation of gene expression predominated, while after aggregates became visible, down-regulation and up-regulation occurred to the same extent. After aggregates became visible there was a down-regulation of dopamine biosynthesis genes accompanied by down-regulation of dopamine levels in culture, indicating the utility of this model to identify functionally relevant pathways. Furthermore, genes of the anti-oxidant Nrf2-ARE pathway were up-regulated, possibly as a protective mechanism. In parallel, we discovered alterations in genes which may result in increased oxidative stress and damage., Conclusion: Up-regulation of gene expression may be more important in HD pathology than previously appreciated. In addition, given the pathogenic impact of oxidative stress and neuroinflammation, the Nrf2-ARE signaling pathway constitutes a new attractive therapeutic target for HD.
- Published
- 2008
- Full Text
- View/download PDF
37. HER-2/neu and p27Kip1 in progression of Fallopian tube carcinoma: an immunohistochemical and array comparative genomic hybridization study.
- Author
-
Nowee ME, Dorsman JC, Piek JM, Kosma VM, Hämäläinen K, Verheijen RH, and van Diest PJ
- Subjects
- Carcinoma metabolism, Carcinoma pathology, Cell Differentiation, Cyclin-Dependent Kinase Inhibitor p27, Disease Progression, Fallopian Tube Neoplasms metabolism, Fallopian Tube Neoplasms pathology, Female, Genomics methods, Humans, Immunohistochemistry, Intracellular Signaling Peptides and Proteins metabolism, Neoplasm Staging, Oligonucleotide Array Sequence Analysis, Receptor, ErbB-2 metabolism, Tumor Suppressor Protein p53 genetics, Carcinoma genetics, Fallopian Tube Neoplasms genetics, Intracellular Signaling Peptides and Proteins genetics, Receptor, ErbB-2 genetics, Tumor Suppressor Protein p53 metabolism
- Abstract
Aims: To determine expression of p53, HER-2/neu and p27(Kip1) in serous Fallopian tube carcinoma (FTC) in relation to stage and grade, and to investigate DNA copy number changes of HER-2 and P27KIP1 as a potential mechanism of altered expression status., Methods and Results: Immunohistochemistry was performed on 28 serous FTCs and 10 normal Fallopian tubes. p53 protein accumulated and p27(Kip1) was down-regulated significantly in early-stage FTCs compared with normal Fallopian tubes. HER-2/neu overexpression was absent in normal Fallopian tubes and in all stage I FTCs (n = 6) but present in 57% (12/21) of advanced-stage FTCs. No differences in expression between grade 2 and 3 tumours were detected. HER-2 gain/amplification was found by array comparative genomic hybridization in 23% (3/13) of analysed FTCs and all showed overexpression. HER-2/neu overexpression also occurred without DNA copy number changes in three other cases. For p27(Kip1), expression and DNA copy number were unrelated., Conclusions: p53 accumulation and p27(Kip1) down-regulation seem to be early events in Fallopian tube carcinogenesis. HER-2/neu showed overexpression, caused by gain/amplification in 50%, and may be involved in progression of FTC. These data contribute to a better understanding of the molecular carcinogenesis of FTC and to possible new therapeutic approaches.
- Published
- 2007
- Full Text
- View/download PDF
38. A case of loss of heterozygosity in the BRCA2 gene of a borderline ovarian tumor: case report and review of literature.
- Author
-
Verbruggen MB, Zweemer RP, Piek JM, van Unnik GA, van Diest PJ, Gille JJ, Menko FH, Dorsman JC, and Verheijen RH
- Subjects
- DNA Mutational Analysis, Female, Humans, Intracellular Signaling Peptides and Proteins, Middle Aged, Mutation, Ovarian Neoplasms diagnosis, Proteins analysis, Genes, BRCA2, Heterozygote, Loss of Heterozygosity genetics, Ovarian Neoplasms genetics
- Abstract
Germline BRCA1 and BRCA2 mutations highly increase the risk of breast and female adnexal cancer. The role of these genes in the tumorigenesis of other malignancies is still under debate. Borderline ovarian tumors (BOT) are occasionally found in families with a strong history of breast and/or female adnexal cancer with or without proven germline mutations. We investigated whether a BOT arising in a germline BRCA2 mutation carrier could be attributed to this mutation, in which case BOT should be added to the BRCA2 related tumor spectrum. Tumor DNA of a serous borderline ovarian tumor (sBOT) of a 55-year-old female carrier of a pathogenic BRCA2 mutation (6085G>T) was analyzed for loss of heterozygosity (LOH) of BRCA2. The sBOT cells, unexpectedly, revealed loss of the mutant allele of BRCA2, while ovarian stroma cells and peripheral blood lymphocytes contained both wild-type and mutant allele of BRCA2. The finding that no loss of the wild-type BRCA2 allele was found in the tumor tissue but loss of the mutant allele was seen suggests that sBOT are not part of the BRCA2 related tumor spectrum. In the literature BOT's in germline BRCA1 and BRCA2 mutation carriers are described incidentally, while in patients with a BOT a germline BRCA1 or BRCA2 mutation is rarely found. Therefore, we conclude that borderline ovarian tumors are neither part of the BRCA1- nor the BRCA2- related tumor spectrum.
- Published
- 2007
- Full Text
- View/download PDF
39. Cisplatin and doxorubicin repress Vascular Endothelial Growth Factor expression and differentially down-regulate Hypoxia-inducible Factor I activity in human ovarian cancer cells.
- Author
-
Duyndam MC, van Berkel MP, Dorsman JC, Rockx DA, Pinedo HM, and Boven E
- Subjects
- Antigens, Neoplasm genetics, Carbonic Anhydrase IX, Carbonic Anhydrases genetics, Carrier Proteins genetics, Cell Line, Tumor, Corticosterone, Down-Regulation, Female, Humans, Ovarian Neoplasms metabolism, Ovarian Neoplasms pathology, p300-CBP Transcription Factors genetics, Antineoplastic Agents pharmacology, Cisplatin pharmacology, Doxorubicin pharmacology, Gene Expression Regulation, Neoplastic drug effects, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Ovarian Neoplasms drug therapy, Vascular Endothelial Growth Factor A genetics
- Abstract
Vascular Endothelial Growth Factor (VEGF) and its transcriptional regulator Hypoxia-inducible Factor 1 (HIF-1) play an important role in the process of angiogenesis in many types of cancer, including ovarian cancer. We have examined whether the DNA-damaging drugs cisplatin and doxorubicin and the microtubule inhibitors docetaxel and paclitaxel can affect VEGF expression and HIF-1 activity in three human ovarian cancer cell lines. We demonstrate that cisplatin and doxorubicin abolish hypoxia-induced VEGF mRNA expression in all cell lines, while basal VEGF mRNA expression was also downregulated. Transient transfection with a HIF-1-responsive luciferase construct indicated that cisplatin and doxorubicin inhibited hypoxic activation of HIF-1. Cisplatin repressed HIF-1alpha protein expression in all cell lines. Stimulation of HIF-1alpha protein degradation by cisplatin was observed in the only cell line expressing wild-type p53. Cisplatin also inhibited the synthesis of HIF-1alpha protein for which p53 was dispensable. Interestingly, cisplatin strongly reduced the protein levels of the HIF-1 coactivators p300 and CREB-binding protein (CBP) under hypoxia in all cell lines. Although doxorubicin inhibited hypoxic activation of HIF-1, this drug had no significant effect on the expression levels of HIF-1alpha and hypoxic expression of p300 and CBP was only weakly reduced. Docetaxel and paclitaxel did neither influence VEGF expression nor hypoxia-induced HIF-1 activity. In total, our findings indicate that cisplatin and doxorubicin can repress hypoxic induction of VEGF expression by inhibiting HIF-1 through different mechanisms. This knowledge may be useful for future treatment schedules including agents that target the HIF-1 signalling pathway.
- Published
- 2007
- Full Text
- View/download PDF
40. Fanconi anemia is associated with a defect in the BRCA2 partner PALB2.
- Author
-
Xia B, Dorsman JC, Ameziane N, de Vries Y, Rooimans MA, Sheng Q, Pals G, Errami A, Gluckman E, Llera J, Wang W, Livingston DM, Joenje H, and de Winter JP
- Subjects
- Fanconi Anemia Complementation Group N Protein, Fanconi Anemia Complementation Group Proteins genetics, Genetic Predisposition to Disease, Humans, Mutation, Nuclear Proteins genetics, Tumor Suppressor Proteins genetics, BRCA2 Protein physiology, Breast Neoplasms genetics, Fanconi Anemia genetics, Nuclear Proteins physiology, Tumor Suppressor Proteins physiology
- Abstract
The Fanconi anemia and BRCA networks are considered interconnected, as BRCA2 gene defects have been discovered in individuals with Fanconi anemia subtype D1. Here we show that a defect in the BRCA2-interacting protein PALB2 is associated with Fanconi anemia in an individual with a new subtype. PALB2-deficient cells showed hypersensitivity to cross-linking agents and lacked chromatin-bound BRCA2; these defects were corrected upon ectopic expression of PALB2 or by spontaneous reversion.
- Published
- 2007
- Full Text
- View/download PDF
41. Identification of the Fanconi anemia complementation group I gene, FANCI.
- Author
-
Dorsman JC, Levitus M, Rockx D, Rooimans MA, Oostra AB, Haitjema A, Bakker ST, Steltenpool J, Schuler D, Mohan S, Schindler D, Arwert F, Pals G, Mathew CG, Waisfisz Q, de Winter JP, and Joenje H
- Subjects
- Adolescent, Adult, Base Sequence, Cell Line, Child, Chromosomal Instability genetics, Fanconi Anemia Complementation Group D2 Protein metabolism, Female, Genome, Human genetics, HeLa Cells, Humans, Male, Molecular Sequence Data, Mutation genetics, Pedigree, Phenotype, Ubiquitin metabolism, Fanconi Anemia Complementation Group Proteins genetics
- Abstract
To identify the gene underlying Fanconi anemia (FA) complementation group I we studied informative FA-I families by a genome-wide linkage analysis, which resulted in 4 candidate regions together encompassing 351 genes. Candidates were selected via bioinformatics and data mining on the basis of their resemblance to other FA genes/proteins acting in the FA pathway, such as: degree of evolutionary conservation, presence of nuclear localization signals and pattern of tissue-dependent expression. We found a candidate, KIAA1794 on chromosome 15q25-26, to be mutated in 8 affected individuals previously assigned to complementation group I. Western blots of endogenous FANCI indicated that functionally active KIAA1794 protein is lacking in FA-I individuals. Knock-down of KIAA1794 expression by siRNA in HeLa cells caused excessive chromosomal breakage induced by mitomycin C, a hallmark of FA cells. Furthermore, phenotypic reversion of a patient-derived cell line was associated with a secondary genetic alteration at the KIAA1794 locus. These data add up to two conclusions. First, KIAA1794 is a FA gene. Second, this gene is identical to FANCI, since the patient cell lines found mutated in this study included the reference cell line for group I, EUFA592.
- Published
- 2007
- Full Text
- View/download PDF
42. BRCA1 and p53 protein expression in cultured ovarian surface epithelial cells derived from women with and without a BRCA1 germline mutation.
- Author
-
Piek JM, Dorsman JC, Massuger LF, Ansink AC, Weegenaar J, Shvarts A, Kenemans P, and Verheijen RH
- Subjects
- Adult, BRCA1 Protein genetics, Blotting, Western, Cell Line, Tumor, Female, Germ-Line Mutation, Humans, Immunohistochemistry, Middle Aged, Ovarian Neoplasms genetics, Tumor Suppressor Protein p53 genetics, BRCA1 Protein metabolism, Ki-67 Antigen metabolism, Ovarian Neoplasms metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
Aim: Mutations in the BRCA1 and TP53 genes are early genetic events leading to (hereditary) ovarian carcinoma. The human ovarian surface epithelium (OSE) is considered the tissue of origin of at least a subset of these tumours. Therefore, OSE cell cultures derived from women harbouring BRCA1 germline mutations can be a potential model to study hereditary ovarian carcinogenesis. In fact, previous in vitro studies indicate phenotypical differences between OSE from women with and without such germline mutations. Therefore, we have assessed whether differences in the expression of BRCA1 and p53 proteins in cultured OSE cells could contribute to these observations., Study Design: Thirty-two OSE cultures derived from women harbouring a BRCA1 mutation (Predisposed OSE [POSE]) and ten cultures from women without a cancer predisposition (Non predisposed OSE [NPOSE]) were grown under standard conditions. Immunocytochemistry was performed to assess the expression of the BRCA1- and p53 proteins. Ki67 immunocytochemical expression was assessed to determine possible differences in cell cycle status between the two groups. In addition, to study whether wild type p53 was expressed, induction of p53 by cis-platinum was assessed by Western blot., Results: On the basis of Ki67 expression, three different groups were analyzed. In the group with all cultures that expressed Ki67 no significant difference was observed in BRCA1 (P = 0.19) and p53 expression (P = 0.09). In the group with moderate to high Ki67 expression no difference in BRCA1 expression (P = 0.50) was observed. However, p53 expression was significantly lower in the case group (P = 0.01). The same observation for p53 was made in the group with only high Ki67 expression (P = 0.02). Furthermore, the expression of both BRCA1 and p53 positively correlates with Ki67 expression. In POSE and NPOSE, p53 was induced by cis-platinum to a similar extent., Conclusion: Our study indicates differences in the expression of p53, but not in the expression of BRCA1 between POSE and NPOSE. In addition, our findings do suggest the absence of losses of the wild type BRCA1 and p53 genes in the studied OSE cultures. This indicates that losses in these genes cannot account for observed differences in phenotypical traits between POSE and NPOSE, but that differences in levels of p53 might contribute.
- Published
- 2006
- Full Text
- View/download PDF
43. Small N-terminal mutant huntingtin fragments, but not wild type, are mainly present in monomeric form: Implications for pathogenesis.
- Author
-
Cong SY, Pepers BA, Roos RA, van Ommen GJ, and Dorsman JC
- Subjects
- Animals, Blotting, Western methods, Chromatography, Gel methods, Electrophoresis, Polyacrylamide Gel methods, Gene Expression physiology, Green Fluorescent Proteins biosynthesis, Humans, Huntingtin Protein, Huntington Disease genetics, Molecular Weight, Nerve Tissue Proteins genetics, Nuclear Proteins genetics, PC12 Cells, Peptide Fragments genetics, Peptide Fragments metabolism, Peptides chemistry, Rats, Time Factors, Transfection methods, Mutagenesis physiology, Nerve Tissue Proteins chemistry, Nuclear Proteins chemistry, Peptides genetics, Peptides metabolism
- Abstract
N-terminal fragments of huntingtin containing an expanded polyglutamine stretch play an important role in the molecular pathogenesis of Huntington's disease. Their ultimate accumulation in insoluble protein aggregates constitutes an important pathological hallmark of Huntington's disease. We report on systematic biochemical comparison studies of soluble wild type and mutant N-terminal huntingtin fragments. The results show that soluble wild type exon 1 fragments are predominantly present in higher molecular weight complexes with a molecular size of approximately 300 kDa, while their mutant counterparts are mainly present in their monomeric form. In contrast, longer N-terminal fragments corresponding to peptides produced by caspase cleavage do not display these differential properties. These findings suggest that especially an increased amount of monomeric form of small N-terminal mutant huntingtin fragments may facilitate aberrant interactions both with itself via the polyglutamine stretch and with other proteins and thereby contribute to molecular pathogenesis.
- Published
- 2006
- Full Text
- View/download PDF
44. Serous borderline tumor of the ovary presenting with cervical lymph node involvement: a report of 3 cases.
- Author
-
Verbruggen MB, Verheijen RH, van de Goot FR, van Beurden M, Dorsman JC, and van Diest PJ
- Subjects
- Adenocarcinoma pathology, Adult, Arm pathology, Biopsy, Fine-Needle, Cystadenoma, Serous metabolism, Diagnosis, Differential, Female, Humans, Immunohistochemistry, Inclusion Bodies pathology, Lymphatic Metastasis physiopathology, Ovarian Neoplasms metabolism, Thrombosis etiology, Cystadenoma, Serous pathology, Lymphatic Diseases etiology, Lymphatic Metastasis pathology, Ovarian Neoplasms pathology
- Abstract
Supradiaphragmatic lymhadenopathy is extremely rare in patients with a serous borderline ovarian tumor (BOT), and clinically difficult to recognize. We describe 3 cases of serous BOT that primarily presented with arm thrombosis due to supradiaphragmatic lymphadenopathy. In all the 3 cases, fine needle aspiration cytology initially indicated metastatic adenocarcinoma. The primary tumor was not immediately apparent, and multiple diagnostic examinations had to be done before the definitive diagnosis of serous BOT, International Federation of Gynecology and Obstetrics stage IV could be made. In the meanwhile, erroneous therapies had been given in 1 case. After surgical removal of the adnexal masses and full surgical staging, all the 3 patients remained free of disease after a follow-up period of 48 to 84 months. In conclusion, supradiaphragmatic lymph node involvement can be present in patients with serous BOTs, and can even be the presenting symptom. When fine needle aspiration cytology of such a lymph node is compatible with adenocarcinoma of unknown primary, serous BOT should be included in the differential diagnosis and pelvic examination should be performed.
- Published
- 2006
- Full Text
- View/download PDF
45. Mutant huntingtin represses CBP, but not p300, by binding and protein degradation.
- Author
-
Cong SY, Pepers BA, Evert BO, Rubinsztein DC, Roos RA, van Ommen GJ, and Dorsman JC
- Subjects
- Animals, Brain physiopathology, Histone Acetyltransferases metabolism, Huntingtin Protein, Huntington Disease genetics, Huntington Disease metabolism, Huntington Disease physiopathology, Machado-Joseph Disease genetics, Machado-Joseph Disease metabolism, Nerve Tissue Proteins genetics, Nuclear Proteins genetics, PC12 Cells, Proteasome Endopeptidase Complex metabolism, Protein Binding physiology, Rats, Regulatory Elements, Transcriptional physiology, Repressor Proteins genetics, Time Factors, p300-CBP Transcription Factors metabolism, Brain metabolism, CREB-Binding Protein metabolism, Down-Regulation physiology, Mutation physiology, Nerve Tissue Proteins metabolism, Nuclear Proteins metabolism, Repressor Proteins metabolism
- Abstract
Huntington's disease can be used as a model to study neurodegenerative disorders caused by aggregation-prone proteins. It has been proposed that the entrapment of transcription factors in aggregates plays an important role in pathogenesis. We now report that the transcriptional activity of CBP is already repressed in the early time points by soluble mutant huntingtin, whereas the histone acetylase activity of CBP/p300 is gradually diminished over time. Mutant huntingtin bound much stronger to CBP than normal huntingtin, possibly contributing to repression. Especially at the later time points, CBP protein level was gradually reduced via the proteasome pathway. In sharp contrast, p300 was unaffected by mutant huntingtin. This selective degradation of CBP was absent in spinocerebellar ataxia 3. Thus, mutant huntingtin specifically affects CBP and not p300 both at the early and later time points, via multiple mechanisms. In addition to the reduction of CBP, also the altered ratio of these closely related histone acetyl transferases may affect chromatin structure and transcription and thus contribute to neurodegeneration.
- Published
- 2005
46. Epitope mapping of monoclonal antibody 4C8 recognizing the protein huntingtin.
- Author
-
Cong SY, Pepers BA, Roos RA, Van Ommen GJ, and Dorsman JC
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Monoclonal isolation & purification, Cells, Cultured, Glutamic Acid genetics, Humans, Huntingtin Protein, Mice, Molecular Sequence Data, Mutation, Nerve Tissue Proteins metabolism, Nuclear Proteins metabolism, Protein Structure, Tertiary, Antibodies, Monoclonal immunology, Epitope Mapping, Nerve Tissue Proteins immunology, Nuclear Proteins immunology
- Abstract
Huntington's disease is a dominantly inherited, devastating neurodegenerative disorder, caused by a polyglutamine expansion (>37) in the N-terminal region of huntingtin, a protein of unknown function. In patients and normal individuals, N-terminal fragments of huntingtin are found, and the N-terminal fragments of mutant huntingtin are cytotoxic. The functions of wild-type huntingtin and the mechanisms underlying the toxic effects of mutant huntingtin are still ill defined. To get more insight into these topics, monoclonal antibodies (MAbs) are indispensable tools. Antibodies raised against the N-terminus are especially important. Among these, the 4C8 mouse MAb has been extensively used in various approaches. In this study, we have mapped the epitope of 4C8 to a 15-amino acid (aa) region spanning from aa 443 to 457 of the human protein, and found that mutation of three consecutive glutamic acids present in this region disrupts the recognition by 4C8. These results allow a more accurate interpretation of the results obtained by usage of the 4C8 antibody and broaden the utility of this antibody.
- Published
- 2005
- Full Text
- View/download PDF
47. Cultures of ovarian surface epithelium from women with and without a hereditary predisposition to develop female adnexal carcinoma.
- Author
-
Piek JM, Dorsman JC, Shvarts A, Ansink AC, Massuger LF, Scholten P, van Diest PJ, Dijkstra JC, Weegenaar J, Kenemans P, and Verheijen RH
- Subjects
- Adnexa Uteri pathology, Adult, Aged, Biomarkers, Tumor biosynthesis, CA-125 Antigen biosynthesis, Calbindin 2, Cell Culture Techniques, Collagen Type IV biosynthesis, Epithelial Cells cytology, Epithelial Cells metabolism, Epithelial Cells physiology, Female, Genes, BRCA1, Genes, BRCA2, Genetic Predisposition to Disease, Humans, Keratin-7, Keratins biosynthesis, Middle Aged, Mutation, Ovary metabolism, Ovary physiology, S100 Calcium Binding Protein G biosynthesis, Uterine Neoplasms metabolism, Ovary cytology, Uterine Neoplasms genetics, Uterine Neoplasms pathology
- Abstract
Aim: Conflicting evidence exists on whether in vivo morphological characteristics can distinguish Ovarian Surface Epithelium (OSE) of ovaries obtained from women with and without a predisposition to develop female adnexal (ovarian and fallopian tube) carcinoma. This study aims to detect differences in growth potential and morphology that are maintained or specifically expressed in vitro., Study Design: Ovarian surfaces were scraped to retrieve OSE cells from 56 women at hereditary high risk for female adnexal carcinoma, of whom 33 are BRCA1 and four are BRCA2 mutation carriers (Predisposed OSE, POSE) and from 26 women without such risk (Non Predisposed OSE, NPOSE). Number of passages and total cell yield until last passage, as well as morphology was compared between both groups. To confirm morphology, the expression of epithelial, mesothelial, and fibroblast markers was assessed., Results: Both POSE and NPOSE cultures displayed similar growth potential and morphology. The expression of epithelial markers cyto-keratins 7 and 8 was similar between both groups. Only in cultures in which cells did not uniformly exhibit these markers, the percentage of cells expressing these markers was significantly lower at last passage when compared to the initial culture. In these latter cultures, cells that were morphologically indistinguishable from fibroblasts were observed. Mesothelial marker calretinin was expressed in 75% of cells of both POSE and NPOSE cultures and correlates with cyto-keratins 7 and 8 expression. CA 125 expression was equally low in POSE and NPOSE cultures (4.3%). Fibroblast markers FSM and vimentin were expressed in 100% and collagen IV was expressed in 16% of cells in all cultures., Conclusion: OSE cells derived from women with a hereditary predisposition to develop female adnexal cancer possess similar in vitro characteristics as OSE from women without this predisposition. On basis of our results, it seems advisable to study only 100% cyto-keratins 7 and 8 positive OSE cultures, since contamination of fibroblasts in some primary OSE cultures cannot be ruled out.
- Published
- 2004
- Full Text
- View/download PDF
48. Genome-wide-array-based comparative genomic hybridization reveals genetic homogeneity and frequent copy number increases encompassing CCNE1 in fallopian tube carcinoma.
- Author
-
Snijders AM, Nowee ME, Fridlyand J, Piek JM, Dorsman JC, Jain AN, Pinkel D, van Diest PJ, Verheijen RH, and Albertson DG
- Subjects
- Carcinoma metabolism, Chromosome Mapping, Cyclin E, DNA metabolism, Fallopian Tube Neoplasms metabolism, Female, Genome, Humans, Models, Genetic, Mutation, Nucleic Acid Hybridization, Oncogene Proteins biosynthesis, Carcinoma genetics, Fallopian Tube Neoplasms genetics, Gene Expression Regulation, Neoplastic, Oligonucleotide Array Sequence Analysis methods, Oncogene Proteins genetics, Oncogenes genetics
- Abstract
Fallopian tube carcinoma (FTC) is a rare, poorly studied and aggressive cancer, associated with poor survival. Since tumorigenesis is related to the acquisition of genetic changes, we used genome-wide array comparative genomic hybridization to analyse copy number aberrations occurring in FTC in order to obtain a better understanding of FTC carcinogenesis and to identify prognostic events and targets for therapy. We used arrays of 2464 genomic clones, providing approximately 1.4 Mb resolution across the genome to map genomic DNA copy number aberrations quantitatively from 14 FTC onto the human genome sequence. All tumors showed a high frequency of copy number aberrations with recurrent gains on 3q, 6p, 7q, 8q, 12p, 17q, 19 and 20q, and losses involving chromosomes 4, 5q, 8p, 16q, 17p, 18q and X. Recurrent regions of amplification included 1p34, 8p11-q11, 8q24, 12p, 17p13, 17q12-q21, 19p13, 19q12-q13 and 19q13. Candidate, known oncogenes mapping to these amplicons included CMYC (8q24), CCNE1 (19q12-q21) and AKT2 (19q13), whereas PIK3CA and KRAS, previously suggested to be candidate driver genes for amplification, mapped outside copy number maxima on 3q and 12p, respectively. The FTC were remarkably homogeneous, with some recurrent aberrations occurring in more than 70% of samples, which suggests a stereotyped pattern of tumor evolution.
- Published
- 2003
- Full Text
- View/download PDF
49. Women harboring BRCA1/2 germline mutations are at risk for breast and female adnexal carcinoma.
- Author
-
Piek JM, Dorsman JC, Zweemer RP, Verheijen RH, van Diest PJ, and Colgan TJ
- Subjects
- Female, Genetic Predisposition to Disease, Humans, Breast Neoplasms genetics, Fallopian Tube Neoplasms genetics, Genes, BRCA1, Genes, BRCA2, Mutation, Ovarian Neoplasms genetics
- Published
- 2003
- Full Text
- View/download PDF
50. Interruption of perfect CAG repeats by CAA triplets improves the stability of glutamine-encoding repeat sequences.
- Author
-
Dorsman JC, Bremmer-Bout M, Pepers B, van Ommen GJ, and Den Dunnen JT
- Subjects
- Cloning, Molecular, Codon, DNA, Recombinant genetics, Deoxyribonucleases, Type II Site-Specific metabolism, Escherichia coli metabolism, Gene Targeting methods, Genetic Code, Glutamine, Oligodeoxyribonucleotides chemical synthesis, Oligodeoxyribonucleotides genetics, Substrate Specificity, Mutagenesis, Site-Directed, Trinucleotide Repeats
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.