Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Leukemia ; 38(3): 621-629, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38184753

ABSTRACT

MYC translocation occurs in 8-14% of diffuse large B-cell lymphoma (DLBCL), and may concur with BCL2 and/or BCL6 translocation, known as double-hit (DH) or triple-hit (TH). DLBCL-MYC/BCL2-DH/TH are largely germinal centre B-cell like subtype, but show variable clinical outcome, with IG::MYC fusion significantly associated with inferior survival. While DLBCL-MYC/BCL6-DH are variable in their cell-of-origin subtypes and clinical outcome. Intriguingly, only 40-50% of DLBCL with MYC translocation show high MYC protein expression (>70%). We studied 186 DLBCLs with MYC translocation including 32 MYC/BCL2/BCL6-TH, 75 MYC/BCL2-DH and 26 MYC/BCL6-DH. FISH revealed a MYC/BCL6 fusion in 59% of DLBCL-MYC/BCL2/BCL6-TH and 27% of DLBCL-MYC/BCL6-DH. Targeted NGS showed a similar mutation profile and LymphGen genetic subtype between DLBCL-MYC/BCL2/BCL6-TH and DLBCL-MYC/BCL2-DH, but variable LymphGen subtypes among DLBCL-MYC/BCL6-DH. MYC protein expression is uniformly high in DLBCL with IG::MYC, but variable in those with non-IG::MYC including MYC/BCL6-fusion. Translocation breakpoint analyses of 8 cases by TLC-based NGS showed no obvious genomic configuration that enables MYC transactivation in 3 of the 4 cases with non-IG::MYC, while a typical promoter substitution or IGH super enhancer juxtaposition in the remaining cases. The findings potentially explain variable MYC expression in DLBCL with MYC translocation, and also bear practical implications in its routine assessment.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Humans , Transcriptional Activation , Proto-Oncogene Proteins c-bcl-6/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Translocation, Genetic , Genomics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism
2.
J Mol Diagn ; 25(10): 758-770, 2023 10.
Article in English | MEDLINE | ID: mdl-37517473

ABSTRACT

Chromosomal rearrangements are important drivers in cancer, and their robust detection is essential for diagnosis, prognosis, and treatment selection, particularly for bone and soft tissue tumors. Current diagnostic methods are hindered by limitations, including difficulties with multiplexing targets and poor quality of RNA. A novel targeted DNA-based next-generation sequencing method, formalin-fixed, paraffin-embedded-targeted locus capture (FFPE-TLC), has shown advantages over current diagnostic methods when applied on FFPE lymphomas, including the ability to detect novel rearrangements. We evaluated the utility of FFPE-TLC in bone and soft tissue tumor diagnostics. FFPE-TLC sequencing was successfully applied on noncalcified and decalcified FFPE samples (n = 44) and control samples (n = 19). In total, 58 rearrangements were identified in 40 FFPE tumor samples, including three previously negative samples, and none was identified in the FFPE control samples. In all five discordant cases, FFPE-TLC could identify gene fusions where other methods had failed due to either detection limits or poor sample quality. FFPE-TLC achieved a high specificity and sensitivity (no false positives and negatives). These results indicate that FFPE-TLC is applicable in cancer diagnostics to simultaneously analyze many genes for their involvement in gene fusions. Similar to the observation in lymphomas, FFPE-TLC is a good DNA-based alternative to the conventional methods for detection of rearrangements in bone and soft tissue tumors.


Subject(s)
High-Throughput Nucleotide Sequencing , Soft Tissue Neoplasms , Humans , Paraffin Embedding/methods , High-Throughput Nucleotide Sequencing/methods , DNA/genetics , Formaldehyde , Soft Tissue Neoplasms/diagnosis , Soft Tissue Neoplasms/genetics , Gene Fusion , Technology , Tissue Fixation
3.
Front Oncol ; 13: 1124737, 2023.
Article in English | MEDLINE | ID: mdl-37152023

ABSTRACT

Background: Liquid biopsies combine minimally invasive sample collection with sensitive detection of residual disease. Pediatric malignancies harbor tumor-driving copy number alterations or fusion genes, rather than recurrent point mutations. These regions contain tumor-specific DNA breakpoint sequences. We investigated the feasibility to use these breakpoints to design patient-specific markers to detect tumor-derived cell-free DNA (cfDNA) in plasma from patients with pediatric solid tumors. Materials and methods: Regions of interest (ROI) were identified through standard clinical diagnostic pipelines, using SNP array for CNAs, and FISH or RT-qPCR for fusion genes. Using targeted locus amplification (TLA) on tumor organoids grown from tumor material or targeted locus capture (TLC) on FFPE material, ROI-specific primers and probes were designed, which were used to design droplet digital PCR (ddPCR) assays. cfDNA from patient plasma at diagnosis and during therapy was analyzed. Results: TLA was performed on material from 2 rhabdomyosarcoma, 1 Ewing sarcoma and 3 neuroblastoma. FFPE-TLC was performed on 8 neuroblastoma tumors. For all patients, at least one patient-specific ddPCR was successfully designed and in all diagnostic plasma samples the patient-specific markers were detected. In the rhabdomyosarcoma and Ewing sarcoma patients, all samples after start of therapy were negative. In neuroblastoma patients, presence of patient-specific markers in cfDNA tracked tumor burden, decreasing during induction therapy, disappearing at complete remission and re-appearing at relapse. Conclusion: We demonstrate the feasibility to determine tumor-specific breakpoints using TLA/TLC in different pediatric solid tumors and use these for analysis of cfDNA from plasma. Considering the high prevalence of CNAs and fusion genes in pediatric solid tumors, this approach holds great promise and deserves further study in a larger cohort with standardized plasma sampling protocols.

4.
Virchows Arch ; 482(6): 983-992, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37067589

ABSTRACT

Tropomyosin receptor kinase (TRK) inhibitors have been approved for metastatic solid tumors harboring NTRK fusions, but the detection of NTRK fusions is challenging. International guidelines recommend pan-TRK immunohistochemistry (IHC) screening followed by next generation sequencing (NGS) in tumor types with low prevalence of NTRK fusions, including metastatic colorectal cancer (mCRC). RNA-based NGS is preferred, but is expensive, time-consuming, and extracting good-quality RNA from FFPE tissue is challenging. Alternatives in daily clinical practice are warranted. We assessed the diagnostic performance of RNA-NGS, FFPE-targeted locus capture (FFPE-TLC), fluorescence in situ hybridization (FISH), and the 5'/3' imbalance quantitative RT-PCR (qRT-PCR) after IHC screening in 268 patients with microsatellite-instability-high mCRC, the subgroup in which NTRK fusions are most prevalent (1-5%). A consensus result was determined after review of all assay results. In 16 IHC positive tumors, 10 NTRK fusions were detected. In 33 IHC negative samples, no additional transcribed NTRK fusions were found, underscoring the high sensitivity of IHC. Sensitivity of RNA-NGS, FFPE-TLC, FISH, and qRT-PCR was 90%, 90%, 78%, and 100%, respectively. Specificity was 100% for all assays. Robustness, defined as the percentage of samples that provided an interpretable result in the first run, was 100% for FFPE-TLC, yet more limited for RNA-NGS (85%), FISH (70%), and qRT-PCR (70%). Overall, we do not recommend FISH for the detection of NTRK fusions in mCRC due to its low sensitivity and limited robustness. We conclude that RNA-NGS, FFPE-TLC, and qRT-PCR are appropriate assays for NTRK fusion detection, after enrichment with pan-TRK IHC, in routine clinical practice.


Subject(s)
Colonic Neoplasms , Neoplasms , Humans , Receptor, trkA/genetics , In Situ Hybridization, Fluorescence , Neoplasms/genetics , Colonic Neoplasms/genetics , Microsatellite Repeats , Oncogene Proteins, Fusion/genetics , Gene Fusion
5.
Br J Haematol ; 194(5): 888-892, 2021 09.
Article in English | MEDLINE | ID: mdl-34337744

ABSTRACT

Minimal residual disease (MRD) diagnostics are implemented in most clinical protocols for patients with acute lymphoblastic leukaemia (ALL) and are mostly performed using rearranged immunoglobulin (IG) and/or T-cell receptor (TR) gene rearrangements as molecular polymerase chain reaction targets. Unfortunately, in 5-10% of patients no or no sensitive IG/TR targets are available, and patients therefore cannot be stratified appropriately. In the present study, we used fusion genes and genomic deletions as alternative MRD targets in these patients, which retrospectively revealed appropriate MDR stratification in 79% of patients with no (sensitive) IG/TR target, and a different risk group stratification in more than half of the cases.


Subject(s)
Neoplasm, Residual/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Child , Gene Deletion , Humans , Neoplasm, Residual/genetics , Oncogene Fusion , Polymerase Chain Reaction , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics
6.
Nat Commun ; 12(1): 3361, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099699

ABSTRACT

In routine diagnostic pathology, cancer biopsies are preserved by formalin-fixed, paraffin-embedding (FFPE) procedures for examination of (intra-) cellular morphology. Such procedures inadvertently induce DNA fragmentation, which compromises sequencing-based analyses of chromosomal rearrangements. Yet, rearrangements drive many types of hematolymphoid malignancies and solid tumors, and their manifestation is instructive for diagnosis, prognosis, and treatment. Here, we present FFPE-targeted locus capture (FFPE-TLC) for targeted sequencing of proximity-ligation products formed in FFPE tissue blocks, and PLIER, a computational framework that allows automated identification and characterization of rearrangements involving selected, clinically relevant, loci. FFPE-TLC, blindly applied to 149 lymphoma and control FFPE samples, identifies the known and previously uncharacterized rearrangement partners. It outperforms fluorescence in situ hybridization (FISH) in sensitivity and specificity, and shows clear advantages over standard capture-NGS methods, finding rearrangements involving repetitive sequences which they typically miss. FFPE-TLC is therefore a powerful clinical diagnostics tool for accurate targeted rearrangement detection in FFPE specimens.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Lymphoma, B-Cell/genetics , Lymphoma, Non-Hodgkin/genetics , Paraffin Embedding/methods , Tissue Fixation/methods , Translocation, Genetic , Computational Biology/methods , Gene Rearrangement , Genes, bcl-2/genetics , Genes, myc/genetics , Humans , In Situ Hybridization, Fluorescence/methods , Lymphoma, B-Cell/diagnosis , Lymphoma, Non-Hodgkin/diagnosis , Proto-Oncogene Proteins c-bcl-6/genetics , Reproducibility of Results , Retrospective Studies , Sensitivity and Specificity
7.
Biosens Bioelectron ; 29(1): 18-22, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21868213

ABSTRACT

It is very challenging to perform sample enrichment for protein biomarkers because proteins can easily change conformation and denature. In this paper we demonstrate protein enrichment suited for high-sensitivity integrated immuno-biosensing. The method enhances the concentration of the biomarkers and simultaneously removes matrix components that could interfere with the immunoassay. Biomarkers are captured using antibody coated magnetic particles and the biomarker antibody complexes are released by enzymatic elution. The eluted complexes are subsequently detected in a sandwich immunoassay biosensor. A scaling study of the enrichment process demonstrates an enrichment factor of 15 in buffer and plasma. We analyze the enrichment factor in terms of the three basic steps of the assay (capture, concentration, elution) and we quantify their respective efficiencies. The process is suited for integration into bio-analytical tools.


Subject(s)
Biosensing Techniques/methods , Immunoassay/methods , Proteins/isolation & purification , Antigen-Antibody Complex/isolation & purification , Biomarkers/analysis , Biomarkers, Tumor/blood , Epitopes/isolation & purification , Humans , Magnetics , Male , Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Prostatic Neoplasms/diagnosis , Proteins/immunology
8.
PLoS One ; 6(4): e19357, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21559415

ABSTRACT

BACKGROUND: Mutagenesis screens in the mouse have been proven useful for the identification of novel gene functions and generation of interesting mutant alleles. Here we describe a phenotype-based screen for recessive mutations affecting embryonic development. METHODOLOGY/PRINCIPAL FINDINGS: Mice were mutagenized with N-ethyl-N-nitrosourea (ENU) and following incrossing the offspring, embryos were analyzed at embryonic day 10.5. Mutant phenotypes that arose in our screen include cardiac and nuchal edema, neural tube defects, situs inversus of the heart, posterior truncation and the absence of limbs and lungs. We isolated amongst others novel mutant alleles for Dll1, Ptprb, Plexin-B2, Fgf10, Wnt3a, Ncx1, Scrib(Scrib, Scribbled homolog [Drosophila]) and Sec24b. We found both nonsense alleles leading to severe protein truncations and mutants with single-amino acid substitutions that are informative at a molecular level. Novel findings include an ectopic neural tube in our Dll1 mutant and lung defects in the planar cell polarity mutants for Sec24b and Scrib. CONCLUSIONS/SIGNIFICANCE: Using a forward genetics approach, we have generated a number of novel mutant alleles that are linked to disturbed morphogenesis during development.


Subject(s)
Ethylnitrosourea/toxicity , Mutagenicity Tests/methods , Mutagens , Alleles , Animals , Embryonic Development/genetics , Genes, Developmental , Genetic Techniques , Lung/drug effects , Mice , Models, Genetic , Mutation , Phenotype , Time Factors
9.
Development ; 137(7): 1067-73, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20215345

ABSTRACT

Among the cellular properties that are essential for the organization of tissues during animal development, the importance of cell polarity in the plane of epithelial sheets has become increasingly clear in the past decades. Planar cell polarity (PCP) signaling in vertebrates has indispensable roles in many aspects of their development, in particular, controlling alignment of various types of epithelial cells. Disrupted PCP has been linked to developmental defects in animals and to human pathology. Neural tube closure defects (NTD) and disorganization of the mechanosensory cells of the organ of Corti are commonly known consequences of disturbed PCP signaling in mammals. We report here a typical PCP phenotype in a mouse mutant for the Sec24b gene, including the severe NTD craniorachischisis, abnormal arrangement of outflow tract vessels and disturbed development of the cochlea. In addition, we observed genetic interaction between Sec24b and the known PCP gene, scribble. Sec24b is a component of the COPII coat protein complex that is part of the endoplasmic reticulum (ER)-derived transport vesicles. Sec24 isoforms are thought to be directly involved in cargo selection, and we present evidence that Sec24b deficiency specifically affects transport of the PCP core protein Vangl2, based on experiments in embryos and in cultured primary cells.


Subject(s)
Cell Polarity , Mutation , Nerve Tissue Proteins/metabolism , Neural Tube Defects/metabolism , Signal Transduction/physiology , Vesicular Transport Proteins/metabolism , Animals , Aorta, Thoracic/abnormalities , Cells, Cultured , Cochlea/abnormalities , Cochlea/anatomy & histology , Cochlea/embryology , Embryo, Mammalian/abnormalities , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Epithelial Cells/cytology , Epithelial Cells/physiology , Female , Fibroblasts/cytology , Fibroblasts/physiology , Heart Defects, Congenital , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Neural Tube Defects/genetics , Neural Tube Defects/pathology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Vesicular Transport Proteins/genetics
10.
Nucleic Acids Res ; 38(10): e116, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20164091

ABSTRACT

Microarray-based enrichment of selected genomic loci is a powerful method for genome complexity reduction for next-generation sequencing. Since the vast majority of exons in vertebrate genomes are smaller than 150 nt, we explored the use of short fragment libraries (85-110 bp) to achieve higher enrichment specificity by reducing carryover and adverse effects of flanking intronic sequences. High enrichment specificity (60-75%) was obtained with a relative even base coverage. Up to 98% of the target-sequence was covered more than 20x at an average coverage depth of about 200x. To verify the accuracy of SNP/mutation detection, we evaluated 384 known non-reference SNPs in the targeted regions. At approximately 200x average sequence coverage, we were able to survey 96.4% of 1.69 Mb of genomic sequence with only 4.2% false negative calls, mostly due to low coverage. Using the same settings, a total of 1197 novel candidate variants were detected. Verification experiments revealed only eight false positive calls, indicating an overall false positive rate of less than 1 per approximately 200,000 bp. Taken together, short fragment libraries provide highly efficient and flexible enrichment of exonic targets and yield relatively even base coverage, which facilitates accurate SNP and mutation detection. Raw sequencing data, alignment files and called SNPs have been submitted into GEO database http://www.ncbi.nlm.nih.gov/geo/ with accession number GSE18542.


Subject(s)
DNA Mutational Analysis/methods , Oligonucleotide Array Sequence Analysis/methods , Polymorphism, Single Nucleotide , Sequence Analysis, DNA/methods , Gene Library , Genome, Human , Humans , Mutation
11.
Int J Dev Biol ; 54(10): 1465-71, 2010.
Article in English | MEDLINE | ID: mdl-21302256

ABSTRACT

The biological role and structure-function relationship of the Na(+)Ca(2+) exchanger NCX1 have been the subject of much investigation. Subtle mutagenesis to study the function of a protein seems only feasible in in vitro systems, but genetic forward screens have the potential to provide in vivo models to study single amino acid substitutions. In a genetic screen in mouse, we have isolated a mutant line carrying a novel mutant allele of the mouse Ncx1 gene. In this allele, a point mutation causes the substitution of a highly conserved asparagine residue (N874) with lysine. Accepted models for NCX1 structure propose that the affected amino acid is located in one of the reentrant membrane loops and experiments in vitro have identified N874 as critical for the ion transport function of NCX1. We found severe circulation defects and defective placentation in homozygous Ncx1(N87K4) mutant embryos, making the phenotype essentially indistinguishable from those of previously described null mutants. By ex vivo analysis, we demonstrated intrinsic functional abnormalities of cardiomyocytes. Western blot analysis and immunohistochemistry demonstrated normal levels and subcellular localization of the altered protein, ruling out the possibility that the abnormalities are a mere consequence of a major disturbance of protein structure. This study confirms and extends studies in vitro indicating the significance of amino acid N874 for the function of the NCX1 protein. It provides an in vivo model for this mutation and demonstrates the potential of forward genetic screens in a mammalian system.


Subject(s)
Amino Acid Substitution , Heart Defects, Congenital/genetics , Myocytes, Cardiac/physiology , Sodium-Calcium Exchanger/genetics , Action Potentials , Animals , Calcium/blood , Embryo, Mammalian/metabolism , Female , Ion Transport , Mice , Mice, Transgenic , Myocardial Contraction , Myocytes, Cardiac/pathology , Phenotype , Placentation , Pregnancy , Sodium-Calcium Exchanger/chemistry , Structure-Activity Relationship
12.
Methods Mol Biol ; 546: 3-12, 2009.
Article in English | MEDLINE | ID: mdl-19378094

ABSTRACT

ENU (N-ethyl-N-nitrosourea) mutagenesis is a widely accepted and proven method to introduce random point mutations in the genome. Because there are no targeted knockout strategies available for zebrafish so far, random mutagenesis is currently the preferred method in both forward and reverse genetic approaches. To obtain high-density mutagenized zebrafish, six consecutive ENU treatments are applied at weekly intervals to adult male zebrafish by bathing them in ENU solution. With this procedure an average germ line mutation load of one mutation every 1.0 x 10(5)-1.5 x 10(5) basepairs is reached routinely in our lab.


Subject(s)
Ethylnitrosourea/administration & dosage , Mutagenesis/drug effects , Zebrafish/genetics , Alkylating Agents/administration & dosage , Animals , Animals, Genetically Modified , DNA/drug effects , Drug Administration Schedule , Female , Genetic Engineering/methods , Genome/drug effects , Germ-Line Mutation , Male
13.
Cancer Res ; 68(13): 5059-66, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18593904

ABSTRACT

Defective mismatch repair (MMR) in humans causes hereditary nonpolyposis colorectal cancer. This genetic predisposition to colon cancer is linked to heterozygous familial mutations, and loss-of-heterozygosity is necessary for tumor development. In contrast, the rare cases with biallelic MMR mutations are juvenile patients with brain tumors, skin neurofibromas, and café-au-lait spots, resembling the neurofibromatosis syndrome. Many of them also display lymphomas and leukemias, which phenotypically resembles the frequent lymphoma development in mouse MMR knockouts. Here, we describe the identification and characterization of novel knockout mutants of the three major MMR genes, mlh1, msh2, and msh6, in zebrafish and show that they develop tumors at low frequencies. Predominantly, neurofibromas/malignant peripheral nerve sheath tumors were observed; however, a range of other tumor types was also observed. Our findings indicate that zebrafish mimic distinct features of the human disease and are complementary to mouse models.


Subject(s)
DNA Mismatch Repair , DNA Repair Enzymes/genetics , Neoplasms/genetics , Neurofibromatoses/genetics , Zebrafish/genetics , Abdominal Neoplasms/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Base Sequence , Brain Neoplasms/genetics , DNA Mutational Analysis , DNA-Binding Proteins/genetics , Eye Neoplasms/genetics , Female , Hemangiosarcoma/genetics , Male , Microsatellite Instability , MutS Homolog 2 Protein/genetics , Mutation/physiology , Nerve Sheath Neoplasms/genetics
14.
Nucleic Acids Res ; 36(12): 4047-56, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18522974

ABSTRACT

S(N)1-type alkylating agents, like N-methyl-N-nitrosourea (MNU) and N-ethyl-N-nitrosourea (ENU), are potent mutagens. Exposure to alkylating agents gives rise to O(6)-alkylguanine, a modified base that is recognized by DNA mismatch repair (MMR) proteins but is not repairable, resulting in replication fork stalling and cell death. We used a somatic mutation detection assay to study the in vivo effects of alkylation damage on lethality and mutation frequency in developing zebrafish embryos. Consistent with the damage-sensing role of the MMR system, mutant embryos lacking the MMR enzyme MSH6 displayed lower lethality than wild-type embryos after exposure to ENU and MNU. In line with this, alkylation-induced somatic mutation frequencies were found to be higher in wild-type embryos than in the msh6 loss-of-function mutants. These mutations were found to be chromosomal aberrations that may be caused by chromosomal breaks that arise from stalled replication forks. As these chromosomal breaks arise at replication, they are not expected to be repaired by non-homologous end joining. Indeed, Ku70 loss-of-function mutants were found to be equally sensitive to ENU as wild-type embryos. Taken together, our results suggest that in vivo alkylation damage results in chromosomal instability and cell death due to aberrantly processed MMR-induced stalled replication forks.


Subject(s)
Alkylating Agents/toxicity , Chromosomal Instability , DNA Damage , DNA Mismatch Repair , Ethylnitrosourea/toxicity , Methylnitrosourea/toxicity , Mutagens/toxicity , Alkylation , Animals , Cell Death , Chromosome Aberrations , DNA Replication , DNA-Binding Proteins/genetics , Embryo, Nonmammalian/drug effects , Gene Deletion , Mutation , Zebrafish/embryology , Zebrafish/genetics
15.
Mutagenesis ; 23(4): 325-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18469325

ABSTRACT

S(N)1-type alkylating agents such as N-ethyl-N-nitrosourea (ENU) are very potent mutagens. They act by transferring their alkyl group to DNA bases, which, upon mispairing during replication, can cause single base pair mutations in the next replication cycle. As DNA mismatch repair (MMR) proteins are involved in the recognition of alkylation damage, we hypothesized that ENU-induced mutation rates could be increased in a MMR-deficient background, which would be beneficial for mutagenesis approaches. We applied a standard ENU mutagenesis protocol to adult zebrafish deficient in the MMR gene msh6 and heterozygous controls to study the effect of MMR on ENU-induced DNA damage. Dose-dependent lethality was found to be similar for homozygous and heterozygous mutants, indicating that there is no difference in ENU resistance. Mutation discovery by high-throughput dideoxy resequencing of genomic targets in outcrossed progeny of the mutagenized fish did also not reveal any differences in germ line mutation frequency. These results may indicate that the maximum mutation load for zebrafish has been reached with the currently used, highly optimized ENU mutagenesis protocol. Alternatively, the MMR system in the zebrafish germ line may be saturated very rapidly, thereby having a limited effect on high-dose ENU mutagenesis.


Subject(s)
DNA Repair-Deficiency Disorders/genetics , Ethylnitrosourea/toxicity , Germ Cells/drug effects , Mutagenesis/drug effects , Zebrafish/genetics , Animals , Animals, Genetically Modified , DNA Mismatch Repair/drug effects , DNA-Binding Proteins/genetics , Embryo, Nonmammalian , Fertilization/genetics , Gene Frequency , Germ-Line Mutation , Male , Mutagenesis/genetics , Mutagens/toxicity , Zebrafish/embryology
16.
Mol Cancer Res ; 6(5): 685-94, 2008 May.
Article in English | MEDLINE | ID: mdl-18505914

ABSTRACT

The zebrafish has developed into an important model organism for biomedical research over the last decades. Although the main focus of zebrafish research has traditionally been on developmental biology, keeping and observing zebrafish in the lab led to the identification of diseases similar to humans, such as cancer, which subsequently became a subject for study. As a result, about 50 articles have been published since 2000 in which zebrafish were used as a cancer model. Strategies used include carcinogenic treatments, transplantation of mammalian cancer cells, forward genetic screens for proliferation or genomic instability, reverse genetic target-selected mutagenesis to inactivate known tumor suppressor genes, and the generation of transgenics to express human oncogenes. Zebrafish have been found to develop almost any tumor type known from human, with similar morphology and, according to gene expression array studies, comparable signaling pathways. However, tumor incidences are relatively low, albeit highly comparable between different mutants, and tumors develop late in life. In addition, tumor spectra are sometimes different when compared with mice and humans. Nevertheless, the zebrafish model has created its own niche in cancer research, complementing existing models with its specific experimental advantages and characteristics. Examples of these are imaging of tumor progression in living fish by fluorescence, treatment with chemical compounds, and screening possibilities not only for chemical modifiers but also for genetic enhancers and suppressors. This review aims to provide a comprehensive overview of the state of the art of zebrafish as a model in cancer research. (Mol Cancer Res 2008;6(5):685-94).


Subject(s)
Disease Models, Animal , Gene Expression Regulation, Neoplastic , Mutation , Neoplasms/genetics , Neoplasms/therapy , Animals , Animals, Genetically Modified , Genome , Humans , Mice , Models, Genetic , Mutagenesis , Mutagens , Neoplasms/epidemiology , Neoplasms/metabolism , Signal Transduction , Zebrafish
17.
Cell Tissue Res ; 332(1): 133-9, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18247060

ABSTRACT

Mlh1 is a member of DNA mismatch repair (MMR) machinery and is also essential for the stabilization of crossovers during the first meiotic division. Recently, we have shown that zebrafish mlh1 mutant males are completely infertile because of a block in metaphase I, whereas females are fertile but have aneuploid progeny. When studying fertility in males in a two-fold more inbred background, we have however observed low numbers of fertilized eggs (approximately 0.4%). Histological examination of the testis has revealed that all spermatogenic stages prior to spermatids (spermatogonia, primary spermatocytes, and secondary spermatocytes) are significantly increased in the mutant, whereas the total weight of spermatids and spermatozoa is highly decreased (1.8 mg in wild-type vs. 0.1 mg in mutants), a result clearly different from our previous study in which outbred males lack secondary spermatocytes or postmeiotic cells. Thus, a delay of both meiotic divisions occurs rather than complete arrest during meiosis I in these males. Eggs fertilized with mutant sperm develop as malformed embryos and are aneuploid making this male phenotype much more similar to that previously described in the mutant females. Therefore, crossovers are still essential for proper meiosis, but meiotic cell divisions can progress without it, suggesting that this mutant is a suitable model for studying the cellular mechanisms of completing meiosis without crossover stabilization.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , DNA Mismatch Repair , Meiosis/genetics , Nuclear Proteins/genetics , Spermatogenesis/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Aneuploidy , Animals , Animals, Genetically Modified , Apoptosis/genetics , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/pathology , Female , Fertility/genetics , Histones/metabolism , In Situ Nick-End Labeling , Male , MutL Protein Homolog 1 , Organ Size , Phenotype , Point Mutation , Spermatozoa/metabolism , Spermatozoa/pathology , Testis/pathology , Zebrafish/embryology
18.
Circulation ; 115(14): 1830-8, 2007 Apr 10.
Article in English | MEDLINE | ID: mdl-17372176

ABSTRACT

BACKGROUND: Identifying molecular pathways regulating the development of pacemaking and coordinated heartbeat is crucial for a comprehensive mechanistic understanding of arrhythmia-related diseases. Elucidation of these pathways has been complicated mainly by an insufficient definition of the developmental structures involved in these processes and the unavailability of animal models specifically targeting the relevant tissues. Here, we report on a highly restricted expression pattern of the homeodomain transcription factor Shox2 in the sinus venosus myocardium, including the sinoatrial nodal region and the venous valves. METHODS AND RESULTS: To investigate its function in vivo, we have generated mouse lines carrying a targeted mutation of the Shox2 gene. Although heterozygous animals did not exhibit obvious defects, homozygosity of the targeted allele led to embryonic lethality at 11.5 to 13.5 dpc. Shox2-/- embryos exhibited severe hypoplasia of the sinus venosus myocardium in the posterior heart field, including the sinoatrial nodal region and venous valves. We furthermore demonstrate aberrant expression of connexin 40 and connexin 43 and the transcription factor Nkx2.5 in vivo specifically within the sinoatrial nodal region and show that Shox2 deficiency interferes with pacemaking function in zebrafish embryos. CONCLUSIONS: From these results, we postulate a critical function of Shox2 in the recruitment of sinus venosus myocardium comprising the sinoatrial nodal region.


Subject(s)
Gene Expression Regulation, Developmental , Heart/embryology , Homeodomain Proteins/physiology , Transcription Factors/physiology , Zebrafish Proteins/physiology , Animals , Bradycardia/embryology , Bradycardia/genetics , Connexin 43/analysis , Connexins/analysis , Embryonic Development/genetics , Fetal Heart/pathology , Gene Targeting , Genes, Lethal , Heart Conduction System/embryology , Heart Conduction System/physiopathology , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Heart Valves/embryology , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/analysis , Homeodomain Proteins/genetics , Mice/embryology , Mice, Inbred C57BL , Mice, Knockout , Myocardium/pathology , Myocytes, Cardiac/cytology , Phenotype , Sinoatrial Node/embryology , Transcription Factors/analysis , Transcription Factors/genetics , Zebrafish/embryology , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , Gap Junction alpha-5 Protein
19.
Genetics ; 175(4): 1561-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17237513

ABSTRACT

In most eukaryotes, recombination of homologous chromosomes during meiosis is necessary for proper chromosome pairing and subsequent segregation. The molecular mechanisms of meiosis are still relatively unknown, but numerous genes are known to be involved, among which are many mismatch repair genes. One of them, mlh1, colocalizes with presumptive sites of crossing over, but its exact action remains unclear. We studied meiotic processes in a knockout line for mlh1 in zebrafish. Male mlh1 mutants are sterile and display an arrest in spermatogenesis at metaphase I, resulting in increased testis weight due to accumulation of prophase I spermatocytes. In contrast, females are fully fertile, but their progeny shows high rates of dysmorphology and mortality within the first days of development. SNP-based chromosome analysis shows that this is caused by aneuploidy, resulting from meiosis I chromosomal missegregation. Surprisingly, the small percentage of progeny that develops normally has a complete triploid genome, consisting of both sets of maternal and one set of paternal chromosomes. As adults, these triploid fish are infertile males with wild-type appearance. The frequency of triploid progeny of mlh1 mutant females is much higher than could be expected for random chromosome segregation. Together, these results show that multiple solutions exist for meiotic crossover/segregation problems.


Subject(s)
DNA Repair Enzymes/deficiency , Infertility, Male/genetics , Infertility, Male/metabolism , Zebrafish Proteins/deficiency , Zebrafish/genetics , Zebrafish/metabolism , Aneuploidy , Animals , Base Sequence , Crossing Over, Genetic/genetics , DNA Mismatch Repair , DNA Primers/genetics , DNA Repair Enzymes/genetics , Female , Infertility, Male/pathology , Male , Meiosis/genetics , Mutation , Phenotype , Polyploidy , Seminiferous Tubules/pathology , Zebrafish Proteins/genetics
20.
Dev Biol ; 285(2): 533-44, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16039644

ABSTRACT

The role of the aristaless-related homeobox gene Alx4 in antero-posterior (AP-) patterning of the developing vertebrate limb has remained somewhat elusive. Polydactyly of Alx4 mutant mice is known to be accompanied by ectopic anterior expression of genes like Shh, Fgf4 and 5'Hoxd. We reported previously that polydactyly in Alx4 mutant mice requires SHH signaling, but we now show that in early Alx4-/- limb buds the anterior ectopic expression of Fgf4 and Hoxd13, and therefore disruption of AP-patterning, occurs independently of SHH signaling. To better understand how Alx4 functions in the pathways that regulate AP-patterning, we also studied genomic regulatory sequences that are capable of directing expression of a reporter gene in a pattern corresponding to endogenous Alx4 expression in anterior limb bud mesenchyme. We observed, as expected for authentic Alx4 expression, expansion of reporter construct expression in a Shh-/- background. Total lack of reporter expression in a Gli3-/- background confirms the existence of Gli3-dependent and -independent Alx4 expression in the limb bud. Apparently, these two modules of Alx4 expression are linked to dissimilar functions.


Subject(s)
Body Patterning/genetics , Extremities/embryology , Homeodomain Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction/genetics , Trans-Activators/metabolism , Animals , DNA Primers , Genomics/methods , Hedgehog Proteins , In Situ Hybridization , Mice , Mice, Knockout , Plasmids/genetics , Zinc Finger Protein Gli3 , beta-Galactosidase
SELECTION OF CITATIONS
SEARCH DETAIL
...