68 results on '"van Eeden FJ"'
Search Results
2. HYPOXIA-INDUCIBLE FACTOR 2 alpha REGULATES NEUTROPHILIC INFLAMMATION IN HUMANS, MICE AND ZEBRAFISH
- Author
-
Thompson, AAR, Elks, PM, Marriott, HM, Higgins, KR, Parmar, S, Shaw, G, Eamsamarng, S, McGrath, EE, Formenti, F, Van Eeden, FJ, Kinnula, VL, Pugh, CW, Sabroe, I, Dockrell, DH, Chilvers, ER, Robbins, PA, Simon, MC, Johnson, RS, Renshaw, SA, Whyte, MKB, and Walmsley, SR
- Published
- 2016
- Full Text
- View/download PDF
3. Mutations in LRRC50 Predispose Zebrafish and Humans to Seminomas
- Author
-
Basten, SG, Davis, EE, Gillis, Ad, van Rooijen, E, Stoop, Hans, Babala, N, Logister, I, Heath, ZG, Jonges, TN, Katsanis, N, Voest, EE, van Eeden, FJ, Medema, RH, Ketting, RF, Schulte-Merker, S, Looijenga, LHJ (Leendert), Giles, RH, Basten, SG, Davis, EE, Gillis, Ad, van Rooijen, E, Stoop, Hans, Babala, N, Logister, I, Heath, ZG, Jonges, TN, Katsanis, N, Voest, EE, van Eeden, FJ, Medema, RH, Ketting, RF, Schulte-Merker, S, Looijenga, LHJ (Leendert), and Giles, RH
- Published
- 2013
4. Second generation lethality in RNAseH2a knockout zebrafish.
- Author
-
Thomas RC, Zaksauskaite R, Al-Kandari NY, Hyde AC, Abugable AA, El-Khamisy SF, and van Eeden FJ
- Subjects
- Animals, Female, Male, Brain metabolism, Brain embryology, Disease Models, Animal, Gene Knockout Techniques, Nervous System Malformations genetics, Ribonucleotides metabolism, Testis metabolism, Ribonuclease H genetics, Ribonuclease H metabolism, Zebrafish genetics, Zebrafish embryology, Zebrafish Proteins genetics, Zebrafish Proteins metabolism
- Abstract
Removal of ribonucleotides from DNA by RNaseH2 is essential for genome stability, and its impacted function causes the neurodegenerative disease, Aicardi Goutières Syndrome. We have created a zebrafish rnaseh2a mutant to model this process. Surprisingly, RNaseH2a knockouts show little phenotypic abnormality at adulthood in the first generation, unlike mouse knockout models, which are early embryonic lethal. However, the second generation offspring show reduced development, increased ribonucleotide incorporation and upregulation of key inflammatory markers, resulting in both maternal and paternal embryonic lethality. Thus, neither fathers or mothers can generate viable offspring even when crossed to wild-type partners. Despite their survival, rnaseh2a-/- adults show an accumulation of ribonucleotides in both the brain and testes that is not present in early development. Our data suggest that homozygotes possess RNaseH2 independent compensatory mechanisms that are inactive or overwhelmed by the inherited ribonucleotides in their offspring, or that zebrafish have a yet unknown tolerance mechanism. Additionally, we identify ribodysgenesis, the rapid removal of rNMPs and subsequently lethal fragmentation of DNA as responsible for maternal and paternal embryonic lethality., (© The Author(s) 2024. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2024
- Full Text
- View/download PDF
5. Asymmetric Hapln1a drives regionalized cardiac ECM expansion and promotes heart morphogenesis in zebrafish development.
- Author
-
Derrick CJ, Sánchez-Posada J, Hussein F, Tessadori F, Pollitt EJG, Savage AM, Wilkinson RN, Chico TJ, van Eeden FJ, Bakkers J, and Noël ES
- Subjects
- Animals, Animals, Genetically Modified, Body Patterning, Extracellular Matrix genetics, Extracellular Matrix Proteins genetics, Gene Expression Regulation, Developmental, Hyaluronic Acid metabolism, Mutation, Proteoglycans genetics, Signal Transduction, Transcriptome, Zebrafish embryology, Zebrafish genetics, Zebrafish Proteins genetics, Extracellular Matrix metabolism, Extracellular Matrix Proteins metabolism, Heart embryology, Morphogenesis, Myocardium metabolism, Proteoglycans metabolism, Zebrafish metabolism, Zebrafish Proteins metabolism
- Abstract
Aims: Vertebrate heart development requires the complex morphogenesis of a linear tube to form the mature organ, a process essential for correct cardiac form and function, requiring coordination of embryonic laterality, cardiac growth, and regionalized cellular changes. While previous studies have demonstrated broad requirements for extracellular matrix (ECM) components in cardiac morphogenesis, we hypothesized that ECM regionalization may fine tune cardiac shape during heart development., Methods and Results: Using live in vivo light sheet imaging of zebrafish embryos, we describe a left-sided expansion of the ECM between the myocardium and endocardium prior to the onset of heart looping and chamber ballooning. Analysis using an ECM sensor revealed the cardiac ECM is further regionalized along the atrioventricular axis. Spatial transcriptomic analysis of gene expression in the heart tube identified candidate genes that may drive ECM expansion. This approach identified regionalized expression of hapln1a, encoding an ECM cross-linking protein. Validation of transcriptomic data by in situ hybridization confirmed regionalized hapln1a expression in the heart, with highest levels of expression in the future atrium and on the left side of the tube, overlapping with the observed ECM expansion. Analysis of CRISPR-Cas9-generated hapln1a mutants revealed a reduction in atrial size and reduced chamber ballooning. Loss-of-function analysis demonstrated that ECM expansion is dependent upon Hapln1a, together supporting a role for Hapln1a in regionalized ECM modulation and cardiac morphogenesis. Analysis of hapln1a expression in zebrafish mutants with randomized or absent embryonic left-right asymmetry revealed that laterality cues position hapln1a-expressing cells asymmetrically in the left side of the heart tube., Conclusion: We identify a regionalized ECM expansion in the heart tube which promotes correct heart development, and propose a novel model whereby embryonic laterality cues orient the axis of ECM asymmetry in the heart, suggesting these two pathways interact to promote robust cardiac morphogenesis., (© The Author(s) 2021. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2022
- Full Text
- View/download PDF
6. Loss of Deacetylation Enzymes Hdac6 and Sirt2 Promotes Acetylation of Cytoplasmic Tubulin, but Suppresses Axonemal Acetylation in Zebrafish Cilia.
- Author
-
Łysyganicz PK, Pooranachandran N, Liu X, Adamson KI, Zielonka K, Elworthy S, van Eeden FJ, Grierson AJ, and Malicki JJ
- Abstract
Cilia are evolutionarily highly conserved organelles with important functions in many organs. The extracellular component of the cilium protruding from the plasma membrane comprises an axoneme composed of microtubule doublets, arranged in a 9 + 0 conformation in primary cilia or 9 + 2 in motile cilia. These microtubules facilitate transport of intraflagellar cargoes along the axoneme. They also provide structural stability to the cilium, which may play an important role in sensory cilia, where signals are received from the movement of extracellular fluid. Post-translational modification of microtubules in cilia is a well-studied phenomenon, and acetylation on lysine 40 (K40) of alpha tubulin is prominent in cilia. It is believed that this modification contributes to the stabilization of cilia. Two classes of enzymes, histone acetyltransferases and histone deacetylases, mediate regulation of tubulin acetylation. Here we use a genetic approach, immunocytochemistry and behavioral tests to investigate the function of tubulin deacetylases in cilia in a zebrafish model. By mutating three histone deacetylase genes ( Sirt2 , Hdac6 , and Hdac10 ), we identify an unforeseen role for Hdac6 and Sirt2 in cilia. As expected, mutation of these genes leads to increased acetylation of cytoplasmic tubulin, however, surprisingly it caused decreased tubulin acetylation in cilia in the developing eye, ear, brain and kidney. Cilia in the ear and eye showed elevated levels of mono-glycylated tubulin suggesting a compensatory mechanism. These changes did not affect the length or morphology of cilia, however, functional defects in balance was observed, suggesting that the level of tubulin acetylation may affect function of the cilium., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Łysyganicz, Pooranachandran, Liu, Adamson, Zielonka, Elworthy, van Eeden, Grierson and Malicki.)
- Published
- 2021
- Full Text
- View/download PDF
7. Investigation of the role of VHL-HIF signaling in DNA repair and apoptosis in zebrafish.
- Author
-
Kim HR, Santhakumar K, Markham E, Baldera D, Greenald D, Bryant HE, El-Khamisy SF, and van Eeden FJ
- Abstract
pVHL is a tumor suppressor. The lack of its function leads to various tumors, among which ccRCC (clear cell renal cell carcinoma) has the most serious outcome due to its resistance to chemotherapies and radiotherapies. Although HIF promotes the progression of ccRCC, the precise mechanism by which the loss of VHL leads to tumor initiation remains unclear. We exploited two zebrafish vhl mutants, vhl and vll , and Tg (phd3:: EGFP)
i144 fish to identify crucial functions of Vhl in tumor initiation. Through the mutant analysis, we found that the role of pVHL in DNA repair is conserved in zebrafish Vll. Interestingly, we also discovered that Hif activation strongly suppressed genotoxic stress induced DNA repair defects and apoptosis in vll and brca2 mutants and in embryos lacking ATM activity. These results suggest the potential of HIF as a clinical modulator that can protect cells from accumulating DNA damage and apoptosis which can lead to cancers and neurodegenerative disorders., Competing Interests: CONFLICTS OF INTEREST None.- Published
- 2020
- Full Text
- View/download PDF
8. Euthanizing zebrafish legally in Europe: Are the approved methods of euthanizing zebrafish appropriate to research reality and animal welfare?
- Author
-
Valentim AM, van Eeden FJ, Strähle U, and Olsson IA
- Subjects
- Animals, Ethics, Research, Europe, Euthanasia, Animal ethics, Euthanasia, Animal methods, Animal Welfare legislation & jurisprudence, Euthanasia, Animal legislation & jurisprudence, Research, Zebrafish
- Published
- 2016
- Full Text
- View/download PDF
9. Genome-wide mapping of Hif-1α binding sites in zebrafish.
- Author
-
Greenald D, Jeyakani J, Pelster B, Sealy I, Mathavan S, and van Eeden FJ
- Subjects
- Animals, Chromatin Immunoprecipitation, Computational Biology, Gene Expression Regulation, High-Throughput Nucleotide Sequencing, Humans, Mutation, Nucleotide Motifs, Protein Binding, Real-Time Polymerase Chain Reaction, Reproducibility of Results, Response Elements, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, Zebrafish Proteins genetics, Zebrafish Proteins metabolism, Binding Sites, Genome, Genome-Wide Association Study, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Zebrafish genetics, Zebrafish metabolism
- Abstract
Background: Hypoxia Inducible Factor (HIF) regulates a cascade of transcriptional events in response to decreased oxygenation, acting from the cellular to the physiological level. This response is evolutionarily conserved, allowing the use of zebrafish (Danio rerio) as a model for studying the hypoxic response. Activation of the hypoxic response can be achieved in zebrafish by homozygous null mutation of the von Hippel-Lindau (vhl) tumour suppressor gene. Previous work from our lab has focused on the phenotypic characterisation of this mutant, establishing the links between vhl mutation, the hypoxic response and cancer. To further develop fish as a model for studying hypoxic signalling, we examine the transcriptional profile of the vhl mutant with respect to Hif-1α. As our approach uses embryos consisting of many cell types, it has the potential to uncover additional HIF regulated genes that have escaped detection in analogous mammalian cell culture studies., Results: We performed high-density oligonucleotide microarray analysis of the gene expression changes in von Hippel-Lindau mutant zebrafish, which identified up-regulation of well-known hypoxia response genes and down-regulation of genes primarily involved in lipid processing. To identify the dependency of these transcriptional changes on HIF, we undertook Chromatin Immunoprecipitation linked next generation sequencing (ChIP-seq) for the transcription factor Hypoxia Inducible Factor 1α (HIF-1α). We identified HIF-1α binding sites across the genome, with binding sites showing enrichment for an RCGTG motif, showing conservation with the mammalian hypoxia response element., Conclusions: Transcriptome analysis of vhl mutant embryos detected activation of key hypoxia response genes seen in human cell models of hypoxia, but also suppression of many genes primarily involved in lipid processing. ChIP-seq analysis of Hif-1α binding sites unveiled an unprecedented number of loci, with a high proportion containing a canonical hypoxia response element. Whether these sites are functional remains unknown, nevertheless their frequent location near transcriptional start sites suggests functionality, and will allow for investigation into the potential hypoxic regulation of genes in their vicinity. We expect that our data will be an excellent starting point for analysis of both fish and mammalian gene regulation by HIF.
- Published
- 2015
- Full Text
- View/download PDF
10. Exploring the HIFs, buts and maybes of hypoxia signalling in disease: lessons from zebrafish models.
- Author
-
Elks PM, Renshaw SA, Meijer AH, Walmsley SR, and van Eeden FJ
- Subjects
- Animals, Aryl Hydrocarbon Receptor Nuclear Translocator genetics, Aryl Hydrocarbon Receptor Nuclear Translocator metabolism, Basic Helix-Loop-Helix Transcription Factors genetics, Cell Hypoxia, Drug Design, Humans, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Models, Animal, Molecular Targeted Therapy, Species Specificity, Transcription Factors genetics, Zebrafish genetics, Zebrafish Proteins genetics, Basic Helix-Loop-Helix Transcription Factors metabolism, Cellular Microenvironment, Oxygen metabolism, Signal Transduction drug effects, Transcription Factors metabolism, Zebrafish metabolism, Zebrafish Proteins metabolism
- Abstract
A low level of tissue oxygen (hypoxia) is a physiological feature of a wide range of diseases, from cancer to infection. Cellular hypoxia is sensed by oxygen-sensitive hydroxylase enzymes, which regulate the protein stability of hypoxia-inducible factor α (HIF-α) transcription factors. When stabilised, HIF-α binds with its cofactors to HIF-responsive elements (HREs) in the promoters of target genes to coordinate a wide-ranging transcriptional programme in response to the hypoxic environment. This year marks the 20th anniversary of the discovery of the HIF-1α transcription factor, and in recent years the HIF-mediated hypoxia response is being increasingly recognised as an important process in determining the outcome of diseases such as cancer, inflammatory disease and bacterial infections. Animal models have shed light on the roles of HIF in disease and have uncovered intricate control mechanisms that involve multiple cell types, observations that might have been missed in simpler in vitro systems. These findings highlight the need for new whole-organism models of disease to elucidate these complex regulatory mechanisms. In this Review, we discuss recent advances in our understanding of hypoxia and HIFs in disease that have emerged from studies of zebrafish disease models. Findings from such models identify HIF as an integral player in the disease processes. They also highlight HIF pathway components and their targets as potential therapeutic targets against conditions that range from cancers to infectious disease., (© 2015. Published by The Company of Biologists Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
11. klf2ash317 Mutant Zebrafish Do Not Recapitulate Morpholino-Induced Vascular and Haematopoietic Phenotypes.
- Author
-
Novodvorsky P, Watson O, Gray C, Wilkinson RN, Reeve S, Smythe C, Beniston R, Plant K, Maguire R, M K Rothman A, Elworthy S, van Eeden FJ, and Chico TJ
- Subjects
- Animals, Gene Expression Regulation, Developmental, Genotype, Humans, Kruppel-Like Transcription Factors antagonists & inhibitors, Kruppel-Like Transcription Factors biosynthesis, Morpholinos genetics, Mutation, Signal Transduction, Zebrafish genetics, Zebrafish growth & development, Zebrafish Proteins antagonists & inhibitors, Zebrafish Proteins biosynthesis, Cardiovascular System growth & development, Hematopoietic System growth & development, Kruppel-Like Transcription Factors genetics, Morphogenesis genetics, Zebrafish Proteins genetics
- Abstract
Introduction and Objectives: The zinc-finger transcription factor Krϋppel-like factor 2 (KLF2) transduces blood flow into molecular signals responsible for a wide range of responses within the vasculature. KLF2 maintains a healthy, quiescent endothelial phenotype. Previous studies report a range of phenotypes following morpholino antisense oligonucleotide-induced klf2a knockdown in zebrafish. Targeted genome editing is an increasingly applied method for functional assessment of candidate genes. We therefore generated a stable klf2a mutant zebrafish and characterised its cardiovascular and haematopoietic development., Methods and Results: Using Transcription Activator-Like Effector Nucleases (TALEN) we generated a klf2a mutant (klf2ash317) with a 14bp deletion leading to a premature stop codon in exon 2. Western blotting confirmed loss of wild type Klf2a protein and the presence of a truncated protein in klf2ash317 mutants. Homozygous klf2ash317 mutants exhibit no defects in vascular patterning, survive to adulthood and are fertile, without displaying previously described morphant phenotypes such as high-output cardiac failure, reduced haematopoetic stem cell (HSC) development or impaired formation of the 5th accessory aortic arch. Homozygous klf2ash317 mutation did not reduce angiogenesis in zebrafish with homozygous mutations in von Hippel Lindau (vhl), a form of angiogenesis that is dependent on blood flow. We examined expression of three klf family members in wildtype and klf2ash317 zebrafish. We detected vascular expression of klf2b (but not klf4a or biklf/klf4b/klf17) in wildtypes but found no differences in expression that might account for the lack of phenotype in klf2ash317 mutants. klf2b morpholino knockdown did not affect heart rate or impair formation of the 5th accessory aortic arch in either wildtypes or klf2ash317 mutants., Conclusions: The klf2ash317 mutation produces a truncated Klf2a protein but, unlike morpholino induced klf2a knockdown, does not affect cardiovascular development.
- Published
- 2015
- Full Text
- View/download PDF
12. Hypoxia-inducible factor 2α regulates key neutrophil functions in humans, mice, and zebrafish.
- Author
-
Thompson AA, Elks PM, Marriott HM, Eamsamarng S, Higgins KR, Lewis A, Williams L, Parmar S, Shaw G, McGrath EE, Formenti F, Van Eeden FJ, Kinnula VL, Pugh CW, Sabroe I, Dockrell DH, Chilvers ER, Robbins PA, Percy MJ, Simon MC, Johnson RS, Renshaw SA, Whyte MK, and Walmsley SR
- Subjects
- Animals, Apoptosis, Cell Hypoxia, Green Fluorescent Proteins metabolism, Humans, Immunohistochemistry, Mice, Mice, Inbred C57BL, Muramidase, Neutrophils cytology, Phagocytosis, Phenotype, RNA metabolism, Respiratory Burst, Zebrafish, Basic Helix-Loop-Helix Transcription Factors metabolism, Gene Expression Regulation, Inflammation, Neutrophils metabolism
- Abstract
Neutrophil lifespan and function are regulated by hypoxia via components of the hypoxia inducible factor (HIF)/von Hippel Lindau/hydroxylase pathway, including specific roles for HIF-1α and prolyl hydroxylase-3. HIF-2α has both distinct and overlapping biological roles with HIF-1α and has not previously been studied in the context of neutrophil biology. We investigated the role of HIF-2α in regulating key neutrophil functions. Human and murine peripheral blood neutrophils expressed HIF-2α, with expression up-regulated by acute and chronic inflammatory stimuli and in disease-associated inflammatory neutrophil. HIF2A gain-of-function mutations resulted in a reduction in neutrophil apoptosis both ex vivo, through the study of patient cells, and in vivo in a zebrafish tail injury model. In contrast, HIF-2α-deficient murine inflammatory neutrophils displayed increased sensitivity to nitrosative stress induced apoptosis ex vivo and increased neutrophil apoptosis in vivo, resulting in a reduction in neutrophilic inflammation and reduced tissue injury. Expression of HIF-2α was temporally dissociated from HIF-1α in vivo and predominated in the resolution phase of inflammation. These data support a critical and selective role for HIF-2α in persistence of neutrophilic inflammation and provide a platform to dissect the therapeutic utility of targeting HIF-2α in chronic inflammatory diseases.
- Published
- 2014
- Full Text
- View/download PDF
13. Zebrafish as a model of cardiac disease.
- Author
-
Wilkinson RN, Jopling C, and van Eeden FJ
- Subjects
- Animals, Electrophysiological Phenomena, Heart embryology, Heart physiology, Regeneration physiology, Zebrafish embryology, Disease Models, Animal, Heart Diseases pathology, Zebrafish physiology
- Abstract
The zebrafish has been rapidly adopted as a model for cardiac development and disease. The transparency of the embryo, its limited requirement for active oxygen delivery, and ease of use in genetic manipulations and chemical exposure have made it a powerful alternative to rodents. Novel technologies like TALEN/CRISPR-mediated genome engineering and advanced imaging methods will only accelerate its use. Here, we give an overview of heart development and function in the fish and highlight a number of areas where it is most actively contributing to the understanding of cardiac development and disease. We also review the current state of research on a feature that we only could wish to be conserved between fish and human; cardiac regeneration., (© 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
14. The zebrafish as a model of vascular development and disease.
- Author
-
Wilkinson RN and van Eeden FJ
- Subjects
- Animals, Blood Vessels pathology, Disease Models, Animal, Humans, Lymphangiogenesis, Muscle Development, Blood Vessels embryology, Vascular Diseases pathology, Zebrafish embryology, Zebrafish physiology
- Abstract
The zebrafish has recently emerged as an important animal model to study the formation of the vertebrate vascular network. The small size, optical translucency, and genetic tractability of the zebrafish embryo, in combination with an abundance of fluorescent transgenic lines which permit direct visualization of in vivo vessel formation, have greatly advanced our understanding of vascular biology. Widespread adoption of this powerful system has led to many important discoveries in relation to the mechanisms that underlie blood vessel formation. This review highlights the contribution of the zebrafish system to the current understanding of blood vessel formation and the use of zebrafish to model human vascular disease., (© 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
15. A method for high-throughput PCR-based genotyping of larval zebrafish tail biopsies.
- Author
-
Wilkinson RN, Elworthy S, Ingham PW, and van Eeden FJ
- Subjects
- Animals, Biopsy veterinary, High-Throughput Nucleotide Sequencing, Larva genetics, Polymerase Chain Reaction methods, Tail pathology, Zebrafish growth & development, Genotyping Techniques, Zebrafish genetics
- Abstract
Here we describe a method for high-throughput genotyping of live larval zebrafish as early as 72 h post-fertilization (hpf). Importantly, this technique allows rapid and cost-effective PCR-based genotyping from very small fin biopsies, which regenerate as the embryo develops, thereby allowing researchers to select embryos with desired genotypes to be raised to adulthood.
- Published
- 2013
- Full Text
- View/download PDF
16. Blood flow suppresses vascular Notch signalling via dll4 and is required for angiogenesis in response to hypoxic signalling.
- Author
-
Watson O, Novodvorsky P, Gray C, Rothman AM, Lawrie A, Crossman DC, Haase A, McMahon K, Gering M, Van Eeden FJ, and Chico TJ
- Subjects
- Animals, Diacetyl analogs & derivatives, Diacetyl pharmacology, Intracellular Signaling Peptides and Proteins genetics, Membrane Proteins genetics, Tumor Suppressor Proteins physiology, Vascular Endothelial Growth Factor Receptor-2 physiology, Zebrafish embryology, Zebrafish Proteins physiology, Blood Circulation physiology, Hypoxia physiopathology, Intracellular Signaling Peptides and Proteins physiology, Membrane Proteins physiology, Neovascularization, Physiologic, Receptors, Notch physiology, Signal Transduction physiology
- Abstract
Aims: The contribution of blood flow to angiogenesis is incompletely understood. We examined the effect of blood flow on Notch signalling in the vasculature of zebrafish embryos, and whether blood flow regulates angiogenesis in zebrafish with constitutively up-regulated hypoxic signalling., Methods and Results: Developing zebrafish (Danio rerio) embryos survive via diffusion in the absence of circulation induced by knockdown of cardiac troponin T2 or chemical cardiac cessation. The absence of blood flow increased vascular Notch signalling in 48 h post-fertilization old embryos via up-regulation of the Notch ligand dll4. Despite this, patterning of the intersegmental vessels is not affected by absent blood flow. We therefore examined homozygous vhl mutant zebrafish that have constitutively up-regulated hypoxic signalling. These display excessive and aberrant angiogenesis from 72 h post-fertilization, with significantly increased endothelial number, vessel diameter, and length. The absence of blood flow abolished these effects, though normal vessel patterning was preserved., Conclusion: We show that blood flow suppresses vascular Notch signalling via down-regulation of dll4. We have also shown that blood flow is required for angiogenesis in response to hypoxic signalling but is not required for normal vessel patterning. These data indicate important differences in hypoxia-driven vs. developmental angiogenesis.
- Published
- 2013
- Full Text
- View/download PDF
17. Mutations in LRRC50 predispose zebrafish and humans to seminomas.
- Author
-
Basten SG, Davis EE, Gillis AJ, van Rooijen E, Stoop H, Babala N, Logister I, Heath ZG, Jonges TN, Katsanis N, Voest EE, van Eeden FJ, Medema RH, Ketting RF, Schulte-Merker S, Looijenga LH, and Giles RH
- Subjects
- Animals, Genes, Tumor Suppressor, Genotype, Humans, Mutation, Seminoma, Zebrafish genetics
- Abstract
Seminoma is a subclass of human testicular germ cell tumors (TGCT), the most frequently observed cancer in young men with a rising incidence. Here we describe the identification of a novel gene predisposing specifically to seminoma formation in a vertebrate model organism. Zebrafish carrying a heterozygous nonsense mutation in Leucine-Rich Repeat Containing protein 50 (lrrc50 also called dnaaf1), associated previously with ciliary function, are found to be highly susceptible to the formation of seminomas. Genotyping of these zebrafish tumors shows loss of heterozygosity (LOH) of the wild-type lrrc50 allele in 44.4% of tumor samples, correlating with tumor progression. In humans we identified heterozygous germline LRRC50 mutations in two different pedigrees with a family history of seminomas, resulting in a nonsense Arg488* change and a missense Thr590Met change, which show reduced expression of the wild-type allele in seminomas. Zebrafish in vivo complementation studies indicate the Thr590Met to be a loss-of-function mutation. Moreover, we show that a pathogenic Gln307Glu change is significantly enriched in individuals with seminoma tumors (13% of our cohort). Together, our study introduces an animal model for seminoma and suggests LRRC50 to be a novel tumor suppressor implicated in human seminoma pathogenesis., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2013
- Full Text
- View/download PDF
18. Positive and negative regulation of Gli activity by Kif7 in the zebrafish embryo.
- Author
-
Maurya AK, Ben J, Zhao Z, Lee RT, Niah W, Ng AS, Iyu A, Yu W, Elworthy S, van Eeden FJ, and Ingham PW
- Subjects
- Abnormalities, Multiple, Animals, Cerebellar Diseases genetics, Cerebellar Diseases pathology, Cerebellum abnormalities, Embryo, Nonmammalian metabolism, Extremities growth & development, Eye Abnormalities genetics, Eye Abnormalities pathology, Gene Expression Regulation, Developmental, Humans, Kidney Diseases, Cystic genetics, Kidney Diseases, Cystic pathology, Kinesins metabolism, Mice, Neural Tube growth & development, Oncogene Proteins metabolism, Repressor Proteins genetics, Repressor Proteins metabolism, Retina abnormalities, Retina pathology, Trans-Activators metabolism, Transcription Factors metabolism, Zebrafish genetics, Zebrafish growth & development, Zebrafish Proteins metabolism, Zinc Finger Protein GLI1, Zinc Finger Protein Gli2, Cilia genetics, Kinesins genetics, Oncogene Proteins genetics, Trans-Activators genetics, Transcription Factors genetics, Zebrafish Proteins genetics
- Abstract
Loss of function mutations of Kif7, the vertebrate orthologue of the Drosophila Hh pathway component Costal2, cause defects in the limbs and neural tubes of mice, attributable to ectopic expression of Hh target genes. While this implies a functional conservation of Cos2 and Kif7 between flies and vertebrates, the association of Kif7 with the primary cilium, an organelle absent from most Drosophila cells, suggests their mechanisms of action may have diverged. Here, using mutant alleles induced by Zinc Finger Nuclease-mediated targeted mutagenesis, we show that in zebrafish, Kif7 acts principally to suppress the activity of the Gli1 transcription factor. Notably, we find that endogenous Kif7 protein accumulates not only in the primary cilium, as previously observed in mammalian cells, but also in cytoplasmic puncta that disperse in response to Hh pathway activation. Moreover, we show that Drosophila Costal2 can substitute for Kif7, suggesting a conserved mode of action of the two proteins. We show that Kif7 interacts with both Gli1 and Gli2a and suggest that it functions to sequester Gli proteins in the cytoplasm, in a manner analogous to the regulation of Ci by Cos2 in Drosophila. We also show that zebrafish Kif7 potentiates Gli2a activity by promoting its dissociation from the Suppressor of Fused (Sufu) protein and present evidence that it mediates a Smo dependent modification of the full length form of Gli2a. Surprisingly, the function of Kif7 in the zebrafish embryo appears restricted principally to mesodermal derivatives, its inactivation having little effect on neural tube patterning, even when Sufu protein levels are depleted. Remarkably, zebrafish lacking all Kif7 function are viable, in contrast to the peri-natal lethality of mouse kif7 mutants but similar to some Acrocallosal or Joubert syndrome patients who are homozygous for loss of function KIF7 alleles., Competing Interests: AKM, JB and PWI are named inventors on a Singapore Patent Application relating to the potential use of the Kif7 mutant in screens for Hh pathway antagonists. PWI has a minor shareholding in Curis Inc.
- Published
- 2013
- Full Text
- View/download PDF
19. Hypoxia inducible factor signaling modulates susceptibility to mycobacterial infection via a nitric oxide dependent mechanism.
- Author
-
Elks PM, Brizee S, van der Vaart M, Walmsley SR, van Eeden FJ, Renshaw SA, and Meijer AH
- Subjects
- Animals, Animals, Genetically Modified, Basic Helix-Loop-Helix Transcription Factors physiology, Cells, Cultured, Disease Models, Animal, Genetic Predisposition to Disease, Mycobacterium Infections, Nontuberculous genetics, Mycobacterium Infections, Nontuberculous microbiology, Neutrophils metabolism, Nitric Oxide Synthase Type II physiology, Nitrosation, Signal Transduction genetics, Zebrafish, Hypoxia-Inducible Factor 1, alpha Subunit physiology, Mycobacterium Infections, Nontuberculous immunology, Mycobacterium marinum, Nitric Oxide metabolism
- Abstract
Tuberculosis is a current major world-health problem, exacerbated by the causative pathogen, Mycobacterium tuberculosis (Mtb), becoming increasingly resistant to conventional antibiotic treatment. Mtb is able to counteract the bactericidal mechanisms of leukocytes to survive intracellularly and develop a niche permissive for proliferation and dissemination. Understanding of the pathogenesis of mycobacterial infections such as tuberculosis (TB) remains limited, especially for early infection and for reactivation of latent infection. Signaling via hypoxia inducible factor α (HIF-α) transcription factors has previously been implicated in leukocyte activation and host defence. We have previously shown that hypoxic signaling via stabilization of Hif-1α prolongs the functionality of leukocytes in the innate immune response to injury. We sought to manipulate Hif-α signaling in a well-established Mycobacterium marinum (Mm) zebrafish model of TB to investigate effects on the host's ability to combat mycobacterial infection. Stabilization of host Hif-1α, both pharmacologically and genetically, at early stages of Mm infection was able to reduce the bacterial burden of infected larvae. Increasing Hif-1α signaling enhanced levels of reactive nitrogen species (RNS) in neutrophils prior to infection and was able to reduce larval mycobacterial burden. Conversely, decreasing Hif-2α signaling enhanced RNS levels and reduced bacterial burden, demonstrating that Hif-1α and Hif-2α have opposing effects on host susceptibility to mycobacterial infection. The antimicrobial effect of Hif-1α stabilization, and Hif-2α reduction, were demonstrated to be dependent on inducible nitric oxide synthase (iNOS) signaling at early stages of infection. Our findings indicate that induction of leukocyte iNOS by stabilizing Hif-1α, or reducing Hif-2α, aids the host during early stages of Mm infection. Stabilization of Hif-1α therefore represents a potential target for therapeutic intervention against tuberculosis.
- Published
- 2013
- Full Text
- View/download PDF
20. A zebrafish model to study and therapeutically manipulate hypoxia signaling in tumorigenesis.
- Author
-
Santhakumar K, Judson EC, Elks PM, McKee S, Elworthy S, van Rooijen E, Walmsley SS, Renshaw SA, Cross SS, and van Eeden FJ
- Subjects
- 9,10-Dimethyl-1,2-benzanthracene, Animals, Cell Hypoxia drug effects, Cell Hypoxia physiology, Cell Transformation, Neoplastic chemically induced, Hypoxia-Inducible Factor 1 metabolism, Intestinal Neoplasms chemically induced, Liver Neoplasms, Experimental chemically induced, Signal Transduction, Tumor Suppressor Proteins metabolism, Zebrafish Proteins metabolism, Cell Transformation, Neoplastic metabolism, Disease Models, Animal, Intestinal Neoplasms metabolism, Liver Neoplasms, Experimental metabolism
- Abstract
Hypoxic signaling is a central modulator of cellular physiology in cancer. Core members of oxygen-sensing pathway including the von Hippel-Lindau tumor suppressor protein (pVHL) and the hypoxia inducible factor (HIF) transcription factors have been intensively studied, but improved organismal models might speed advances for both pathobiologic understanding and therapeutic modulation. To study HIF signaling during tumorigenesis and development in zebrafish, we developed a unique in vivo reporter for hypoxia, expressing EGFP driven by prolyl hydroxylase 3 (phd3) promoter/regulatory elements. Modulation of HIF pathway in Tg(phd3::EGFP) embryos showed a specific role for hypoxic signaling in the transgene activation. Zebrafish vhl mutants display a systemic hypoxia response, reflected by strong and ubiquitous transgene expression. In contrast to human VHL patients, heterozygous Vhl mice and vhl zebrafish are not predisposed to cancer. However, upon exposure to dimethylbenzanthracene (DMBA), the vhl heterozygous fish showed an increase in the occurrence of hepatic and intestinal tumors, a subset of which exhibited strong transgene expression, suggesting loss of Vhl function in these tumor cells. Compared with control fish, DMBA-treated vhl heterozygous fish also showed an increase in proliferating cell nuclear antigen-positive renal tubules. Taken together, our findings establish Vhl as a genuine tumor suppressor in zebrafish and offer this model as a tool to noninvasively study VHL and HIF signaling during tumorigenesis and development., (©2012 AACR.)
- Published
- 2012
- Full Text
- View/download PDF
21. Hedgehog signaling via a calcitonin receptor-like receptor can induce arterial differentiation independently of VEGF signaling in zebrafish.
- Author
-
Wilkinson RN, Koudijs MJ, Patient RK, Ingham PW, Schulte-Merker S, and van Eeden FJ
- Subjects
- Animals, Animals, Genetically Modified, Arteries embryology, Arteries metabolism, Core Binding Factor Alpha 2 Subunit metabolism, Gene Expression Regulation, Developmental, Gene Knockdown Techniques, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells metabolism, Membrane Proteins, Mutation, Patched Receptors, Patched-1 Receptor, Receptors, Cell Surface genetics, Receptors, Cell Surface metabolism, Receptors, Notch metabolism, Signal Transduction, Zebrafish genetics, Calcitonin Receptor-Like Protein metabolism, Hedgehog Proteins metabolism, Vascular Endothelial Growth Factor A metabolism, Zebrafish embryology, Zebrafish metabolism, Zebrafish Proteins metabolism
- Abstract
Multiple signaling pathways control the specification of endothelial cells (ECs) to become arteries or veins during vertebrate embryogenesis. Current models propose that a cascade of Hedgehog (Hh), vascular endothelial growth factor (VEGF), and Notch signaling acts instructively on ECs to control the choice between arterial or venous fate. Differences in the phenotypes induced by Hh, VEGF, or Notch inhibition suggest that not all of the effects of Hh on arteriovenous specification are mediated by VEGF. We establish that full derepression of the Hh pathway in ptc1;ptc2 mutants converts the posterior cardinal vein into a second arterial vessel that manifests intact arterial gene expression, intersegmental vessel sprouting, and HSC gene expression. Importantly, although VEGF was thought to be absolutely essential for arterial fates, we find that normal and ectopic arterial differentiation can occur without VEGF signaling in ptc1;ptc2 mutants. Furthermore, Hh is able to bypass VEGF to induce arterial differentiation in ECs via the calcitonin receptor-like receptor, thus revealing a surprising complexity in the interplay between Hh and VEGF signaling during arteriovenous specification. Finally, our experiments establish a dual function of Hh during induction of runx1(+) HSCs.
- Published
- 2012
- Full Text
- View/download PDF
22. Activation of hypoxia-inducible factor-1α (Hif-1α) delays inflammation resolution by reducing neutrophil apoptosis and reverse migration in a zebrafish inflammation model.
- Author
-
Elks PM, van Eeden FJ, Dixon G, Wang X, Reyes-Aldasoro CC, Ingham PW, Whyte MK, Walmsley SR, and Renshaw SA
- Subjects
- Amino Acid Sequence, Animals, Animals, Genetically Modified, Aryl Hydrocarbon Receptor Nuclear Translocator genetics, Aryl Hydrocarbon Receptor Nuclear Translocator immunology, Aryl Hydrocarbon Receptor Nuclear Translocator metabolism, Cell Movement immunology, Disease Models, Animal, Embryo, Nonmammalian immunology, Female, Genetic Variation, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Male, Molecular Sequence Data, Mutagenesis, Site-Directed, Zebrafish, Zebrafish Proteins genetics, Zebrafish Proteins immunology, Zebrafish Proteins metabolism, Apoptosis immunology, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Hypoxia-Inducible Factor 1, alpha Subunit immunology, Inflammation immunology, Neutrophils immunology
- Abstract
The oxygen-sensing transcription factor hypoxia-inducible factor-1α (HIF-1α) plays a critical role in the regulation of myeloid cell function. The mechanisms of regulation are not well understood, nor are the phenotypic consequences of HIF modulation in the context of neutrophilic inflammation. Species conservation across higher metazoans enables the use of the genetically tractable and transparent zebrafish (Danio rerio) embryo to study in vivo resolution of the inflammatory response. Using both a pharmacologic approach known to lead to stabilization of HIF-1α, and selective genetic manipulation of zebrafish HIF-1α homologs, we sought to determine the roles of HIF-1α in inflammation resolution. Both approaches reveal that activated Hif-1α delays resolution of inflammation after tail transection in zebrafish larvae. This delay can be replicated by neutrophil-specific Hif activation and is a consequence of both reduced neutrophil apoptosis and increased retention of neutrophils at the site of tissue injury. Hif-activated neutrophils continue to patrol the injury site during the resolution phase, when neutrophils would normally migrate away. Site-directed mutagenesis of Hif in vivo reveals that hydroxylation of Hif-1α by prolyl hydroxylases critically regulates the Hif pathway in zebrafish neutrophils. Our data demonstrate that Hif-1α regulates neutrophil function in complex ways during inflammation resolution in vivo.
- Published
- 2011
- Full Text
- View/download PDF
23. von Hippel-Lindau tumor suppressor mutants faithfully model pathological hypoxia-driven angiogenesis and vascular retinopathies in zebrafish.
- Author
-
van Rooijen E, Voest EE, Logister I, Bussmann J, Korving J, van Eeden FJ, Giles RH, and Schulte-Merker S
- Subjects
- Animals, Disease Models, Animal, Edema complications, Edema pathology, Humans, Macula Lutea metabolism, Macula Lutea pathology, Mutant Proteins metabolism, Receptors, Vascular Endothelial Growth Factor antagonists & inhibitors, Receptors, Vascular Endothelial Growth Factor metabolism, Retinal Detachment complications, Retinal Detachment pathology, Retinal Neovascularization enzymology, Signal Transduction, Tumor Suppressor Proteins genetics, Zebrafish Proteins genetics, Hypoxia complications, Hypoxia pathology, Mutation genetics, Retinal Neovascularization complications, Retinal Neovascularization pathology, Tumor Suppressor Proteins metabolism, Zebrafish metabolism, Zebrafish Proteins metabolism
- Abstract
Biallelic inactivation of the von Hippel-Lindau (VHL) tumor suppressor gene predisposes human patients to the development of highly vascularized neoplasms in multiple organ systems. We show that zebrafish vhl mutants display a marked increase in blood vessel formation throughout the embryo, starting at 2 days post-fertilization. The most severe neovascularization is observed in distinct areas that overlap with high vegfa mRNA expression, including the vhl mutant brain and eye. Real-time quantitative PCR revealed increased expression of the duplicated VEGFA orthologs vegfaa and vegfab, and of vegfb and its receptors flt1, kdr and kdr-like, indicating increased vascular endothelial growth factor (Vegf) signaling in vhl mutants. Similar to VHL-associated retinal neoplasms, diabetic retinopathy and age-related macular degeneration, we show, by tetramethyl rhodamine-dextran angiography, that vascular abnormalities in the vhl(-/-) retina lead to vascular leakage, severe macular edema and retinal detachment. Significantly, vessels in the brain and eye express cxcr4a, a marker gene expressed by tumor and vascular cells in VHL-associated hemangioblastomas and renal cell carcinomas. VEGF receptor (VEGFR) tyrosine kinase inhibition (through exposure to sunitinib and 676475) blocked vhl(-/-)-induced angiogenesis in all affected tissues, demonstrating that Vegfaa, Vegfab and Vegfb are key effectors of the vhl(-/-) angiogenic phenotype through Flt1, Kdr and Kdr-like signaling. Since we show that the vhl(-/-) angiogenic phenotype shares distinct characteristics with VHL-associated vascular neoplasms, zebrafish vhl mutants provide a valuable in vivo vertebrate model to elucidate underlying mechanisms contributing to the development of these lesions. Furthermore, vhl mutant zebrafish embryos carrying blood vessel-specific transgenes represent a unique and clinically relevant model for tissue-specific, hypoxia-induced pathological angiogenesis and vascular retinopathies. Importantly, they will allow for a cost-effective, non-invasive and efficient way to screen for novel pharmacological agents and combinatorial treatments.
- Published
- 2010
- Full Text
- View/download PDF
24. Repression of Hedgehog signalling is required for the acquisition of dorsolateral cell fates in the zebrafish otic vesicle.
- Author
-
Hammond KL, van Eeden FJ, and Whitfield TT
- Subjects
- Animals, Antibodies analysis, Chickens, Collagen genetics, Collagen immunology, DNA Primers, Ear, Inner anatomy & histology, Ear, Inner physiology, Embryo, Nonmammalian immunology, Embryo, Nonmammalian physiology, Hemoglobins genetics, Hemoglobins physiology, Mice, Mutation, Phenotype, Polymerase Chain Reaction, Semicircular Canals anatomy & histology, Semicircular Canals physiology, Species Specificity, Zebrafish genetics, Hearing physiology, Signal Transduction physiology, Zebrafish physiology
- Abstract
In zebrafish, Hedgehog (Hh) signalling from ventral midline structures is necessary and sufficient to specify posterior otic identity. Loss of Hh signalling gives rise to mirror symmetric ears with double anterior character, whereas severe upregulation of Hh signalling leads to double posterior ears. By contrast, in mouse and chick, Hh is predominantly required for dorsoventral otic patterning. Whereas a loss of Hh function in zebrafish does not affect dorsoventral and mediolateral otic patterning, we now show that a gain of Hh signalling activity causes ventromedial otic territories to expand at the expense of dorsolateral domains. In a panel of lines carrying mutations in Hh inhibitor genes, Hh pathway activity is increased throughout the embryo, and dorsolateral otic structures are lost or reduced. Even a modest increase in Hh signalling has consequences for patterning the ear. In ptc1(-/-) and ptc2(-/-) mutant embryos, in which Hh signalling is maximal throughout the embryo, the inner ear is severely ventralised and medialised, in addition to displaying the previously reported double posterior character. Transplantation experiments suggest that the effects of the loss of Hh pathway inhibition on the ear are mediated directly. These new data suggest that Hh signalling must be kept tightly repressed for the correct acquisition of dorsolateral cell fates in the zebrafish otic vesicle, revealing distinct similarities between the roles of Hh signalling in zebrafish and amniote inner ear patterning.
- Published
- 2010
- Full Text
- View/download PDF
25. Zebrafish mutants in the von Hippel-Lindau tumor suppressor display a hypoxic response and recapitulate key aspects of Chuvash polycythemia.
- Author
-
van Rooijen E, Voest EE, Logister I, Korving J, Schwerte T, Schulte-Merker S, Giles RH, and van Eeden FJ
- Subjects
- Amino Acid Sequence, Animals, Basic Helix-Loop-Helix Transcription Factors biosynthesis, Basic Helix-Loop-Helix Transcription Factors genetics, Cell Lineage, Conserved Sequence, Gene Knockout Techniques, Germ-Line Mutation, Hematopoiesis genetics, Humans, Hypoxia physiopathology, Molecular Sequence Data, Point Mutation, Polycythemia physiopathology, RNA, Messenger administration & dosage, RNA, Messenger genetics, RNA, Messenger pharmacology, Recombinant Fusion Proteins physiology, Sequence Alignment, Sequence Homology, Amino Acid, Synteny, Tumor Suppressor Proteins deficiency, Tumor Suppressor Proteins genetics, Von Hippel-Lindau Tumor Suppressor Protein chemistry, Von Hippel-Lindau Tumor Suppressor Protein genetics, Von Hippel-Lindau Tumor Suppressor Protein physiology, Zebrafish, Zebrafish Proteins deficiency, Zebrafish Proteins genetics, Disease Models, Animal, Hypoxia genetics, Polycythemia genetics, Tumor Suppressor Proteins physiology, Zebrafish Proteins physiology
- Abstract
We have generated 2 zebrafish lines carrying inactivating germline mutations in the von Hippel-Lindau (VHL) tumor suppressor gene ortholog vhl. Mutant embryos display a general systemic hypoxic response, including the up-regulation of hypoxia-induced genes by 1 day after fertilization and a severe hyperventilation and cardiophysiologic response. The vhl mutants develop polycythemia with concomitantly increased epo/epor mRNA levels and erythropoietin signaling. In situ hybridizations reveal global up-regulation of both red and white hematopoietic lineages. Hematopoietic tissues are highly proliferative, with enlarged populations of c-myb(+) hematopoietic stem cells and circulating erythroid precursors. Chemical activation of hypoxia-inducible factor signaling recapitulated aspects of the vhl(-/-) phenotype. Furthermore, microarray expression analysis confirms the hypoxic response and hematopoietic phenotype observed in vhl(-/-) embryos. We conclude that VHL participates in regulating hematopoiesis and erythroid differentiation. Injections with human VHLp30 and R200W mutant mRNA demonstrate functional conservation of VHL between mammals and zebrafish at the amino acid level, indicating that vhl mutants are a powerful new tool to study genotype-phenotype correlations in human disease. Zebrafish vhl mutants are the first congenital embryonic viable systemic vertebrate animal model for VHL, representing the most accurate model for VHL-associated polycythemia to date. They will contribute to our understanding of hypoxic signaling, hematopoiesis, and VHL-associated disease progression.
- Published
- 2009
- Full Text
- View/download PDF
26. Hedgehog signaling plays a cell-autonomous role in maximizing cardiac developmental potential.
- Author
-
Thomas NA, Koudijs M, van Eeden FJ, Joyner AL, and Yelon D
- Subjects
- Animals, Cell Differentiation, Cell Proliferation, Endothelial Cells cytology, Gene Expression Regulation, Developmental, Hedgehog Proteins genetics, Mice, Myocardium cytology, Stem Cells cytology, Stem Cells metabolism, Zebrafish embryology, Zebrafish genetics, Zebrafish metabolism, Heart embryology, Hedgehog Proteins metabolism, Myocardium metabolism, Signal Transduction
- Abstract
Elucidation of the complete roster of signals required for myocardial specification is crucial to the future of cardiac regenerative medicine. Prior studies have implicated the Hedgehog (Hh) signaling pathway in the regulation of multiple aspects of heart development. However, our understanding of the contribution of Hh signaling to the initial specification of myocardial progenitor cells remains incomplete. Here, we show that Hh signaling promotes cardiomyocyte formation in zebrafish. Reduced Hh signaling creates a cardiomyocyte deficit, and increased Hh signaling creates a surplus. Through fate-mapping, we find that Hh signaling is required at early stages to ensure specification of the proper number of myocardial progenitors. Genetic inducible fate mapping in mouse indicates that myocardial progenitors respond directly to Hh signals, and transplantation experiments in zebrafish demonstrate that Hh signaling acts cell autonomously to promote the contribution of cells to the myocardium. Thus, Hh signaling plays an essential early role in defining the optimal number of cardiomyocytes, making it an attractive target for manipulation of multipotent progenitor cells.
- Published
- 2008
- Full Text
- View/download PDF
27. LRRC50, a conserved ciliary protein implicated in polycystic kidney disease.
- Author
-
van Rooijen E, Giles RH, Voest EE, van Rooijen C, Schulte-Merker S, and van Eeden FJ
- Subjects
- Animals, Humans, Leucine-Rich Repeat Proteins, Mice, Zebrafish, Cilia, Mutation, Polycystic Kidney Diseases etiology, Proteins genetics
- Abstract
Cilia perform essential motile and sensory functions central to many developmental and physiological processes. Disruption of their structure or function can have profound phenotypic consequences, and has been linked to left-right patterning and polycystic kidney disease. In a forward genetic screen for mutations affecting ciliary motility, we isolated zebrafish mutant hu255H. The mutation was found to disrupt an ortholog of the uncharacterized highly conserved human SDS22-like leucine-rich repeat(LRR)-containing protein LRRC50 (16q24.1) and Chlamydomonas Oda7p. Zebrafish lrrc50 is specifically expressed in all ciliated tissues. lrrc50(hu255H) mutants develop pronephric cysts with an increased proliferative index, severely reduced brush border, and disorganized pronephric cilia manifesting impaired localized fluid flow consistent with ciliary dysfunction. Electron microscopy analysis revealed ultrastructural irregularities of the dynein arms and misalignments of the outer-doublet microtubules on the ciliary axonemes, suggesting instability of the ciliary architecture in lrrc50(hu255H) mutants. TheSDS22-like leucine-rich repeats present in Lrrc50 are necessary for proper protein function, since injection of a deletion construct of the first LRR did not rescue the zebrafish mutant phenotype. Subcellular distribution of human LRRC50-EGFP in MDCK and HEK293T cells is diffusely cytoplasmic and concentrated at the mitotic spindle poles and cilium. LRRC50 RNAi knock-down in human proximal tubule HK-2 cells thoroughly recapitulated the zebrafish brush border and cilia phenotype, suggesting conservation of LRRC50 function between both species. In summary, we present the first genetic vertebrate model for lrrc50 function and propose LRRC50 to be a novel candidate gene for human cystic kidney disease, involved in regulation of microtubule-based cilia and actin-based brush border microvilli.
- Published
- 2008
- Full Text
- View/download PDF
28. Genetic analysis of the two zebrafish patched homologues identifies novel roles for the hedgehog signaling pathway.
- Author
-
Koudijs MJ, den Broeder MJ, Groot E, and van Eeden FJ
- Subjects
- Animal Structures embryology, Animals, Base Sequence, Body Patterning drug effects, DNA Mutational Analysis, Embryo, Nonmammalian abnormalities, Eye Abnormalities genetics, Gene Expression Regulation, Developmental drug effects, Hedgehog Proteins genetics, Molecular Sequence Data, Mutant Proteins metabolism, Mutation genetics, Patched Receptors, Patched-1 Receptor, Phenotype, RNA Splice Sites genetics, Sequence Homology, Amino Acid, Somites abnormalities, Somites drug effects, Tretinoin pharmacology, Veratrum Alkaloids pharmacology, Hedgehog Proteins metabolism, Membrane Proteins genetics, Receptors, Cell Surface genetics, Signal Transduction drug effects, Zebrafish embryology, Zebrafish Proteins genetics
- Abstract
Background: Aberrant activation of the Hedgehog (Hh) signaling pathway in different organisms has shown the importance of this family of morphogens during development. Genetic screens in zebrafish have assigned specific roles for Hh in proliferation, differentiation and patterning, but mainly as a result of a loss of its activity. We attempted to fully activate the Hh pathway by removing both receptors for the Hh proteins, called Patched1 and 2, which are functioning as negative regulators in this pathway., Results: Here we describe a splice-donor mutation in Ptc1, called ptc1hu1602, which in a homozygous state results in a subtle eye and somite phenotype. Since we recently positionally cloned a ptc2 mutant, a ptc1;ptc2 double mutant was generated, showing severely increased levels of ptc1, gli1 and nkx2.2a, confirming an aberrant activation of Hh signaling. As a consequence, a number of phenotypes were observed that have not been reported previously using Shh mRNA overexpression. Somites of ptc1;ptc2 double mutants do not express anteroposterior polarity markers, however initial segmentation of the somites itself is not affected. This is the first evidence that segmentation and anterior/posterior (A/P) patterning of the somites are genetically uncoupled processes. Furthermore, a novel negative function of Hh signaling is observed in the induction of the fin field, acting well before any of the previously reported function of Shh in fin formation and in a way that is different from the proposed early role of Gli3 in limb/fin bud patterning., Conclusion: The generation and characterization of the ptc1;ptc2 double mutant assigned novel and unexpected functions to the Hh signaling pathway. Additionally, these mutants will provide a useful system to further investigate the consequences of constitutively activated Hh signaling during vertebrate development.
- Published
- 2008
- Full Text
- View/download PDF
29. Laminin-alpha4 and integrin-linked kinase mutations cause human cardiomyopathy via simultaneous defects in cardiomyocytes and endothelial cells.
- Author
-
Knöll R, Postel R, Wang J, Krätzner R, Hennecke G, Vacaru AM, Vakeel P, Schubert C, Murthy K, Rana BK, Kube D, Knöll G, Schäfer K, Hayashi T, Holm T, Kimura A, Schork N, Toliat MR, Nürnberg P, Schultheiss HP, Schaper W, Schaper J, Bos E, Den Hertog J, van Eeden FJ, Peters PJ, Hasenfuss G, Chien KR, and Bakkers J
- Subjects
- Adult, Amino Acid Substitution, Animals, COS Cells, Cardiomyopathy, Dilated metabolism, Cardiomyopathy, Dilated pathology, Cell Adhesion, Chlorocebus aethiops, Chromosome Mapping, Codon, Nonsense, DNA Mutational Analysis, Embryo, Nonmammalian pathology, Epigenesis, Genetic, Extracellular Matrix metabolism, Extracellular Matrix pathology, Female, Heart embryology, Heart Failure etiology, Heart Failure pathology, Humans, Integrins metabolism, Laminin physiology, Male, Middle Aged, Models, Molecular, Myocardium pathology, Oligonucleotides, Antisense toxicity, Pedigree, Protein Binding, Protein Conformation, Protein Interaction Mapping, Protein Serine-Threonine Kinases physiology, Protein Structure, Tertiary, Transfection, Zebrafish embryology, Zebrafish genetics, Zebrafish Proteins genetics, Zebrafish Proteins physiology, Cardiomyopathy, Dilated genetics, Endothelial Cells pathology, Laminin genetics, Mutation, Missense, Myocytes, Cardiac pathology, Point Mutation, Protein Serine-Threonine Kinases genetics
- Abstract
Background: Extracellular matrix proteins, such as laminins, and endothelial cells are known to influence cardiomyocyte performance; however, the underlying molecular mechanisms remain poorly understood., Methods and Results: We used a forward genetic screen in zebrafish to identify novel genes required for myocardial function and were able to identify the lost-contact (loc) mutant, which encodes a nonsense mutation in the integrin-linked kinase (ilk) gene. This loc/ilk mutant is associated with a severe defect in cardiomyocytes and endothelial cells that leads to severe myocardial dysfunction. Additional experiments revealed the epistatic regulation between laminin-alpha4 (Lama4), integrin, and Ilk, which led us to screen for mutations in the human ILK and LAMA4 genes in patients with severe dilated cardiomyopathy. We identified 2 novel amino acid residue-altering mutations (2828C>T [Pro943Leu] and 3217C>T [Arg1073X]) in the integrin-interacting domain of the LAMA4 gene and 1 mutation (785C>T [Ala262Val]) in the ILK gene. Biacore quantitative protein/protein interaction data, which have been used to determine the equilibrium dissociation constants, point to the loss of integrin-binding capacity in case of the Pro943Leu (Kd=5+/-3 micromol/L) and Arg1073X LAMA4 (Kd=1+/-0.2 micromol/L) mutants compared with the wild-type LAMA4 protein (Kd=440+/-20 nmol/L). Additional functional data point to the loss of endothelial cells in affected patients as a direct consequence of the mutant genes, which ultimately leads to heart failure., Conclusions: This is the first report on mutations in the laminin, integrin, and ILK system in human cardiomyopathy, which has consequences for endothelial cells as well as for cardiomyocytes, thus providing a new genetic basis for dilated cardiomyopathy in humans.
- Published
- 2007
- Full Text
- View/download PDF
30. Genetic variation in the zebrafish.
- Author
-
Guryev V, Koudijs MJ, Berezikov E, Johnson SL, Plasterk RH, van Eeden FJ, and Cuppen E
- Subjects
- Animals, Chromosome Mapping methods, Genetic Linkage genetics, Genetic Markers genetics, Species Specificity, Genome genetics, Polymorphism, Single Nucleotide, RNA Editing genetics, Zebrafish genetics
- Abstract
Although zebrafish was introduced as a laboratory model organism several decades ago and now serves as a primary model for developmental biology, there is only limited data on its genetic variation. An establishment of a dense polymorphism map becomes a requirement for effective linkage analysis and cloning approaches in zebrafish. By comparing ESTs to whole-genome shotgun data, we predicted >50,000 high-quality candidate SNPs covering the zebrafish genome with average resolution of 41 kbp. We experimentally validated approximately 65% of a randomly sampled subset by genotyping 16 samples from seven commonly used zebrafish strains. The analysis reveals very high nucleotide diversity between zebrafish isolates. Even with the limited number of samples that we genotyped, zebrafish isolates revealed considerable interstrain variation, ranging from 7% (inbred) to 37% (wild-derived) of polymorphic sites being heterozygous. The increased proportion of polymorphic over monomorphic sites results in five times more frequent observation of a three allelic variant compared with human or mouse. Phylogenetic analysis shows that comparisons between even the least divergent strains used in our analysis may provide one informative marker approximately every 500 nucleotides. Furthermore, the number of haplotypes per locus is relatively large, reflecting independent establishment of the different lines from wild isolates. Finally, our results suggest the presence of prominent C-to-U and A-to-I RNA editing events in zebrafish. Overall, the levels and organization of genetic variation between and within commonly used zebrafish strains are markedly different from other laboratory model organisms, which may affect experimental design and interpretation.
- Published
- 2006
- Full Text
- View/download PDF
31. The zebrafish mutants dre, uki, and lep encode negative regulators of the hedgehog signaling pathway.
- Author
-
Koudijs MJ, den Broeder MJ, Keijser A, Wienholds E, Houwing S, van Rooijen EM, Geisler R, and van Eeden FJ
- Subjects
- Animals, Body Size, Cell Proliferation, Dwarfism, Embryo, Nonmammalian, Hedgehog Proteins, Proliferating Cell Nuclear Antigen genetics, Zebrafish genetics, Zebrafish growth & development, Mutation, Signal Transduction genetics, Trans-Activators genetics
- Abstract
Proliferation is one of the basic processes that control embryogenesis. To identify factors involved in the regulation of proliferation, we performed a zebrafish genetic screen in which we used proliferating cell nuclear antigen (PCNA) expression as a readout. Two mutants, hu418B and hu540A, show increased PCNA expression. Morphologically both mutants resembled the dre (dreumes), uki (ukkie), and lep (leprechaun) mutant class and both are shown to be additional uki alleles. Surprisingly, although an increased size is detected of multiple structures in these mutant embryos, adults become dwarfs. We show that these mutations disrupt repressors of the Hedgehog (Hh) signaling pathway. The dre, uki, and lep loci encode Su(fu) (suppressor of fused), Hip (Hedgehog interacting protein), and Ptc2 (Patched2) proteins, respectively. This class of mutants is therefore unique compared to previously described Hh mutants from zebrafish genetic screens, which mainly show loss of Hh signaling. Furthermore, su(fu) and ptc2 mutants have not been described in vertebrate model systems before. Inhibiting Hh activity by cyclopamine rescues uki and lep mutants and confirms the overactivation of the Hh signaling pathway in these mutants. Triple uki/dre/lep mutants show neither an additive increase in PCNA expression nor enhanced embryonic phenotypes, suggesting that other negative regulators, possibly Ptc1, prevent further activation of the Hh signaling pathway. The effects of increased Hh signaling resulting from the genetic alterations in the uki, dre, and lep mutants differ from phenotypes described as a result of Hh overexpression and therefore provide additional insight into the role of Hh signaling during vertebrate development., Competing Interests: Competing interests. The authors have declared that no competing interests exist.
- Published
- 2005
- Full Text
- View/download PDF
32. The microRNA-producing enzyme Dicer1 is essential for zebrafish development.
- Author
-
Wienholds E, Koudijs MJ, van Eeden FJ, Cuppen E, and Plasterk RH
- Subjects
- Animals, DEAD-box RNA Helicases, Endoribonucleases genetics, Gene Silencing, Mutation, RNA Helicases genetics, Ribonuclease III, Endoribonucleases physiology, MicroRNAs biosynthesis, RNA Helicases physiology, Zebrafish embryology
- Abstract
MicroRNAs (miRNAs) are produced by the Dicer1 enzyme; the role of Dicer1 in vertebrate development is unknown. Here we report target-selected inactivation of the dicer1 gene in zebrafish. We observed an initial build-up of miRNA levels, produced by maternal Dicer1, in homozygous dicer1 mutants, but miRNA accumulation stopped after a few days. This resulted in developmental arrest around day 10. These results indicate that miRNA-producing Dicer1 is essential for vertebrate development.
- Published
- 2003
- Full Text
- View/download PDF
33. Zebrafish embryos as a model host for the real time analysis of Salmonella typhimurium infections.
- Author
-
van der Sar AM, Musters RJ, van Eeden FJ, Appelmelk BJ, Vandenbroucke-Grauls CM, and Bitter W
- Subjects
- Animals, Colony Count, Microbial, Embryo, Nonmammalian metabolism, Escherichia coli physiology, Escherichia coli Infections microbiology, Macrophages cytology, Macrophages microbiology, Salmonella typhimurium cytology, Salmonella typhimurium genetics, Salmonella typhimurium pathogenicity, Time Factors, Embryo, Nonmammalian microbiology, Salmonella Infections, Animal microbiology, Salmonella typhimurium physiology, Zebrafish embryology
- Abstract
Bacterial virulence is best studied in animal models. However, the lack of possibilities for real time analysis and the need for laborious and invasive sample analysis limit the use of experimental animals. In the present study 28 h-old zebrafish embryos were infected with DsRed-labelled cells of Salmonella typhimurium. Using multidimensional digital imaging microscopy we were able to determine the exact location and fate of these bacterial pathogens in a living vertebrate host during three days. A low dose of wild-type S. typhimurium resulted in a lethal infection with bacteria residing and multiplying both in macrophage-like cells and at the epithelium of blood vessels. Lipopolysaccharide (LPS) mutants of S. typhimurium, known to be attenuated in the murine model, proved to be non-pathogenic in the zebrafish embryos and were partially lysed in the bloodstream or degraded in macrophage-like cells. However, injection of LPS mutants in the yolk of the embryo resulted in uncontrolled bacterial proliferation. Heat-killed, wild-type bacteria were completely lysed extracellularly within minutes after injection, which shows that the blood of these zebrafish embryos does already contain lytic activity. In conclusion, the zebrafish embryo model allows for rapid, non-invasive and real time analysis of bacterial infections in a vertebrate host.
- Published
- 2003
- Full Text
- View/download PDF
34. Barentsz is essential for the posterior localization of oskar mRNA and colocalizes with it to the posterior pole.
- Author
-
van Eeden FJ, Palacios IM, Petronczki M, Weston MJ, and St Johnston D
- Subjects
- Animals, Cell Polarity physiology, Cloning, Molecular, Drosophila, Female, Insect Proteins analysis, Male, Microtubules physiology, Molecular Sequence Data, Mutation physiology, Oocytes cytology, Oocytes physiology, Oogenesis physiology, Phenotype, Polymorphism, Restriction Fragment Length, RNA, Messenger metabolism, Recombination, Genetic physiology, Sequence Homology, Amino Acid, Drosophila Proteins, Insect Proteins genetics, Insect Proteins metabolism, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism
- Abstract
The localization of Oskar at the posterior pole of the Drosophila oocyte induces the assembly of the pole plasm and therefore defines where the abdomen and germ cells form in the embryo. This localization is achieved by the targeting of oskar mRNA to the posterior and the localized activation of its translation. oskar mRNA seems likely to be actively transported along microtubules, since its localization requires both an intact microtubule cytoskeleton and the plus end-directed motor kinesin I, but nothing is known about how the RNA is coupled to the motor. Here, we describe barentsz, a novel gene required for the localization of oskar mRNA. In contrast to all other mutations that disrupt this process, barentsz-null mutants completely block the posterior localization of oskar mRNA without affecting bicoid and gurken mRNA localization, the organization of the microtubules, or subsequent steps in pole plasm assembly. Surprisingly, most mutant embryos still form an abdomen, indicating that oskar mRNA localization is partially redundant with the translational control. Barentsz protein colocalizes to the posterior with oskar mRNA, and this localization is oskar mRNA dependent. Thus, Barentsz is essential for the posterior localization of oskar mRNA and behaves as a specific component of the oskar RNA transport complex.
- Published
- 2001
- Full Text
- View/download PDF
35. Two distinct cell populations in the floor plate of the zebrafish are induced by different pathways.
- Author
-
Odenthal J, van Eeden FJ, Haffter P, Ingham PW, and Nüsslein-Volhard C
- Subjects
- Animals, Body Patterning genetics, Gene Expression Regulation, Developmental, Hedgehog Proteins, In Situ Hybridization, Mice, Motor Neurons cytology, Mutation, Notochord cytology, Notochord embryology, Proteins genetics, Species Specificity, Zebrafish genetics, Spinal Cord cytology, Spinal Cord embryology, Trans-Activators, Zebrafish embryology
- Abstract
The floor plate is a morphologically distinct structure of epithelial cells situated along the midline of the ventral spinal cord in vertebrates. It is a source of guidance molecules directing the growth of axons along and across the midline of the neural tube. In the zebrafish, the floor plate is about three cells wide and composed of cuboidal cells. Two cell populations can be distinguished by the expression patterns of several marker genes, including sonic hedgehog (shh) and the fork head-domain gene fkd4: a single row of medial floor plate (MFP) cells, expressing both shh and fkd4, is flanked by rows of lateral floor plate (LFP) cells that express fkd4 but not shh. Systematic mutant searches in zebrafish embryos have identified a number of genes, mutations in which visibly reduce the floor plate. In these mutants either the MFP or the LFP cells are absent, as revealed by the analysis of the shh and fkd4 expression patterns. MFP cells are absent, but LFP cells are present, in mutants of cyclops, one-eyed pinhead, and schmalspur, whose development of midline structures is affected. LFP cells are absent, but MFP cells are present, in mutants of four genes, sonic you, you, you-too, and chameleon, collectively called the you-type genes. This group of mutants also shows defects in patterning of the paraxial mesoderm, causing U- instead of V-shaped somites. One of the you-type genes, sonic you, was recently shown to encode the zebrafish Shh protein, suggesting that the you-type genes encode components of the Shh signaling pathway. It has been shown previously that in the zebrafish shh is required for the induction of LFP cells, but not for the development of MFP cells. This conclusion is supported by the finding that injection of shh RNA causes an increase in the number of LFP, but not MFP cells. Embryos mutant for iguana, detour, and umleitung share the lack of LFP cells with you-type mutants while somite patterning is not severely affected. In mutants that fail to develop a notochord, MFP cells may be present, but are always surrounded by LFP cells. These data indicate that shh, expressed in the notochord and/or the MFP cells, induces the formation of LFP cells. In embryos doubly mutant for cyclops (cyc) and sonic you (syu) both LFP and MFP cells are deleted. The number of primary motor neurons is strongly reduced in cyc;syu double mutants, while almost normal in single mutants, suggesting that the two different pathways have overlapping functions in the induction of primary motor neurons., (Copyright 2000 Academic Press.)
- Published
- 2000
- Full Text
- View/download PDF
36. Developmental mutant screens in the zebrafish.
- Author
-
van Eeden FJ, Granato M, Odenthal J, and Haffter P
- Subjects
- Alleles, Animals, Breeding, Humans, Genetic Testing methods, Mutagenesis, Zebrafish genetics
- Published
- 1999
- Full Text
- View/download PDF
37. Role of sonic hedgehog in branchiomotor neuron induction in zebrafish.
- Author
-
Chandrasekhar A, Warren JT Jr, Takahashi K, Schauerte HE, van Eeden FJ, Haffter P, and Kuwada JY
- Subjects
- Animals, Branchial Region innervation, Cell Nucleus pathology, Embryo, Nonmammalian, Embryonic Induction, Hedgehog Proteins, Intracellular Signaling Peptides and Proteins, Motor Neurons physiology, Mutation, Nervous System embryology, Proteins metabolism, Rhombencephalon embryology, Rhombencephalon pathology, Spinal Cord, Transforming Growth Factor beta genetics, Zebrafish Proteins, Gene Expression Regulation, Developmental, Neurons physiology, Proteins genetics, Trans-Activators, Zebrafish embryology
- Abstract
The role of zebrafish hedgehog genes in branchiomotor neuron development was analyzed by examining mutations that affect the expression of the hedgehog genes and by overexpressing these genes in embryos. In cyclops mutants, reduction in sonic hedgehog (shh) expression, and elimination of tiggy-winkle hedgehog (twhh) expression, correlated with reductions in branchiomotor neuron populations. Furthermore, branchiomotor neurons were restored in cyclops mutants when shh or twhh was overexpressed. These results suggest that Shh and/or Twhh play an important role in the induction of branchiomotor neurons in vivo. In sonic-you (syu) mutants, where Shh activity was reduced or eliminated due to mutations in shh, branchiomotor neurons were reduced in number in a rhombomere-specific fashion, but never eliminated. Similarly, spinal motor neurons were reduced, but not eliminated, in syu mutants. These results demonstrate that Shh is not solely responsible for inducing branchiomotor and spinal motor neurons, and suggest that Shh and Twhh may function as partially redundant signals for motor neuron induction in zebrafish., (Copyright 1998 Elsevier Science Ireland Ltd. All rights reserved.)
- Published
- 1998
- Full Text
- View/download PDF
38. Sonic hedgehog is not required for the induction of medial floor plate cells in the zebrafish.
- Author
-
Schauerte HE, van Eeden FJ, Fricke C, Odenthal J, Strähle U, and Haffter P
- Subjects
- Animals, Axons, Body Patterning, Cyclic AMP-Dependent Protein Kinases metabolism, DNA-Binding Proteins biosynthesis, Eye embryology, Eye Proteins, Genetic Complementation Test, Hedgehog Proteins, Homeodomain Proteins biosynthesis, Muscles cytology, Mutation, Nervous System Malformations, PAX2 Transcription Factor, PAX6 Transcription Factor, Paired Box Transcription Factors, Protein Sorting Signals genetics, RNA Splicing, Repressor Proteins, Retinal Ganglion Cells, Sequence Analysis, DNA, Signal Transduction, Stem Cells, Transcription Factors biosynthesis, Zebrafish Proteins, Embryonic Induction, Nervous System embryology, Proteins metabolism, Somites, Trans-Activators, Zebrafish embryology
- Abstract
Sonic hedgehog (Shh) is a secreted protein that is involved in the organization and patterning of several tissues in vertebrates. We show that the zebrafish sonic-you (syu) gene, a member of a group of five genes required for somite patterning, is encoding Shh. Embryos mutant for a deletion of syu display defects in patterning of the somites, the lateral floor plate cells, the pectoral fins, the axons of motorneurons and the retinal ganglion cells. In contrast to mouse embryos lacking Shh activity, syu mutant embryos do form medial floor plate cells and motorneurons. Since ectopic overexpression of shh in zebrafish embryos does not induce ectopic medial floor plate cells, we conclude that shh is neither required nor sufficient to induce this cell type in the zebrafish.
- Published
- 1998
- Full Text
- View/download PDF
39. Regulation of netrin-1a expression by hedgehog proteins.
- Author
-
Lauderdale JD, Pasquali SK, Fazel R, van Eeden FJ, Schauerte HE, Haffter P, and Kuwada JY
- Subjects
- Animals, Blastomeres metabolism, Central Nervous System embryology, Embryo, Nonmammalian abnormalities, Embryo, Nonmammalian metabolism, Embryo, Nonmammalian ultrastructure, Embryonic Development, Head abnormalities, Head embryology, Hedgehog Proteins, In Situ Hybridization, Morphogenesis genetics, Nerve Growth Factors genetics, Netrin-1, Notochord physiology, Proteins genetics, Proteins physiology, Recombinant Fusion Proteins metabolism, Signal Transduction, Tail abnormalities, Tail embryology, Tumor Suppressor Proteins, Zebrafish embryology, Zebrafish genetics, Zebrafish Proteins, Central Nervous System metabolism, Gene Expression Regulation, Developmental, Nerve Growth Factors biosynthesis, Nerve Growth Factors physiology, Somites metabolism, Trans-Activators
- Abstract
Netrins, a family of growth cone guidance molecules, are expressed both in the ventral neural tube and in subsets of mesodermal cells. In an effort to better understand the regulation of netrins, we examined the expression of netrin-1a in mutant cyclops, no tail, and floating head zebrafish embryos, in which axial midline structures are perturbed. Netrin-1a expression requires signals present in notochord and floor plate cells. In the myotome, but not the neural tube, netrin-1a expression requires sonic hedgehog. In embryos lacking sonic hedgehog, the sonic-you locus, netrin-1a expression is reduced or absent in the myotomes but present in the neural tube. Embryos lacking sonic hedgehog express tiggy-winkle hedgehog in the floor plate, suggesting that, in the neural tube, tiggy-winkle hedgehog can compensate for the lack of sonic hedgehog in inducing netrin-1a expression. Ectopic expression of sonic hedgehog, tiggy-winkle hedgehog, or echidna hedgehog induces ectopic netrin-1a expression in the neural tube, and ectopic expression of sonic hedgehog or tiggy-winkle hedgehog, but not echidna hedgehog, induces ectopic netrin-1a expression in somites. These data demonstrate that in vertebrates netrin expression is regulated by Hedgehog signaling., (Copyright 1998 Academic Press.)
- Published
- 1998
- Full Text
- View/download PDF
40. Zebrafish segmentation and pair-rule patterning.
- Author
-
van Eeden FJ, Holley SA, Haffter P, and Nüsslein-Volhard C
- Subjects
- Animals, Drosophila embryology, Drosophila genetics, Gene Expression Regulation, Developmental, Genes, Insect, Genetic Linkage, Mutation, Phenotype, Somites cytology, Species Specificity, Body Patterning genetics, Zebrafish embryology, Zebrafish genetics
- Abstract
Segmentation in the vertebrate embryo is evident within the paraxial mesoderm in the form of somites, which are repeated structures that give rise to the vertebrae and muscle of the trunk and tail. In the zebrafish, our genetic screen identified two groups of mutants that affect somite formation and pattern. Mutations of one class, the fss-type mutants, disrupt the formation of the anterior-posterior somite boundaries during somitogenesis. However, segmentation within the paraxial mesoderm is not completely eliminated in these mutants. Irregular somite boundaries form later during embryogenesis and, strikingly, the vertebrae are not fused. Here, we show that formation of the irregular somite boundaries in these mutants is dependent upon the activity of a second group of genes, the you-type genes, which include sonic you, the zebrafish homologue of the Drosophila segment polarity gene, sonic hedgehog. Further to characterize the defects caused by the fss-type mutations, we examined their effects on the expression of her1, a zebrafish homologue of the Drosophila pair-rule gene hairy. In wild-type embryos, her1 is expressed in a dynamic, repeating pattern, remarkably similar to that of its Drosophila and Tribolium counterparts, suggesting that a pair-rule mechanism also functions in the segmentation of the vertebrate paraxial mesoderm. We have found that the fss-type mutants have abnormal pair-rule patterning. Although a her1 mutant could not be identified, analysis of a double mutant that abolishes most her1 expression suggests that a her1 mutant may not display a pair-rule phenotype analogous to the hairy phenotype observed in Drosophila. Cumulatively, our data indicate that zebrafish homologues of both the Drosophila segment polarity genes and pair-rule genes are involved in segmenting the paraxial mesoderm. However, both the relationship between these two groups of genes within the genetic heirarchy governing segmentation and the precise roles that they play during segmentation likely differ significantly between the two organisms.
- Published
- 1998
- Full Text
- View/download PDF
41. Left-right pattern of cardiac BMP4 may drive asymmetry of the heart in zebrafish.
- Author
-
Chen JN, van Eeden FJ, Warren KS, Chin A, Nüsslein-Volhard C, Haffter P, and Fishman MC
- Subjects
- Animals, Bone Morphogenetic Protein 4, Bone Morphogenetic Proteins genetics, Embryo, Nonmammalian, Gene Expression Regulation, Developmental, Mutation, Myocardium metabolism, Signal Transduction, Zebrafish genetics, Zebrafish Proteins, Bone Morphogenetic Proteins metabolism, Heart embryology, Zebrafish embryology
- Abstract
The first evident break in left-right symmetry of the primitive zebrafish heart tube is the shift in pattern of BMP4 expression from radially symmetric to left-predominant. The midline heart tube then 'jogs' to the left and subsequently loops to the right. We examined 279 mutations, affecting more than 200 genes, and found 21 mutations that perturb this process. Some cause BMP4 to remain radially symmetric. Others randomize the asymmetric BMP4 pattern. Retention of BMP4 symmetry is associated with failure to jog: right-predominance of the BMP4 pattern is associated with reversal of the direction of jogging and looping. Raising BMP4 diffusely throughout the heart, via sonic hedgehog injection, or the blocking of its action by injection of a dominant negative BMP4 receptor, prevent directional jogging or looping. The genes crucial to directing cardiac asymmetry include a subset of those needed for patterning the dorsoventral axis and for notochord and ventral spinal cord development. Thus, the pattern of cardiac BMP4 appears to be in the pathway by which the heart interprets lateralizing signals from the midline.
- Published
- 1997
- Full Text
- View/download PDF
42. Mutations affecting development of the zebrafish inner ear and lateral line.
- Author
-
Whitfield TT, Granato M, van Eeden FJ, Schach U, Brand M, Furutani-Seiki M, Haffter P, Hammerschmidt M, Heisenberg CP, Jiang YJ, Kane DA, Kelsh RN, Mullins MC, Odenthal J, and Nüsslein-Volhard C
- Subjects
- Animals, Chromatophores physiology, Genes, Jaw Abnormalities genetics, Otolithic Membrane embryology, Phenotype, Postural Balance physiology, Semicircular Canals abnormalities, Zebrafish anatomy & histology, Mutagenesis, Semicircular Canals embryology, Sense Organs embryology, Zebrafish embryology, Zebrafish genetics
- Abstract
Mutations giving rise to anatomical defects in the inner ear have been isolated in a large scale screen for mutations causing visible abnormalities in the zebrafish embryo (Haffter, P., Granato, M., Brand, M. et al. (1996) Development 123, 1-36). 58 mutants have been classified as having a primary ear phenotype; these fall into several phenotypic classes, affecting presence or size of the otoliths, size and shape of the otic vesicle and formation of the semicircular canals, and define at least 20 complementation groups. Mutations in seven genes cause loss of one or both otoliths, but do not appear to affect development of other structures within the ear. Mutations in seven genes affect morphology and patterning of the inner ear epithelium, including formation of the semicircular canals and, in some, development of sensory patches (maculae and cristae). Within this class, dog-eared mutants show abnormal development of semicircular canals and lack cristae within the ear, while in van gogh, semicircular canals fail to form altogether, resulting in a tiny otic vesicle containing a single sensory patch. Both these mutants show defects in the expression of homeobox genes within the otic vesicle. In a further class of mutants, ear size is affected while patterning appears to be relatively normal; mutations in three genes cause expansion of the otic vesicle, while in little ears and microtic, the ear is abnormally small, but still contains all five sensory patches, as in the wild type. Many of the ear and otolith mutants show an expected behavioural phenotype: embryos fail to balance correctly, and may swim on their sides, upside down, or in circles. Several mutants with similar balance defects have also been isolated that have no obvious structural ear defect, but that may include mutants with vestibular dysfunction of the inner ear (Granato, M., van Eeden, F. J. M., Schach, U. et al. (1996) Development, 123, 399-413,). Mutations in 19 genes causing primary defects in other structures also show an ear defect. In particular, ear phenotypes are often found in conjunction with defects of neural crest derivatives (pigment cells and/or cartilaginous elements of the jaw). At least one mutant, dog-eared, shows defects in both the ear and another placodally derived sensory system, the lateral line, while hypersensitive mutants have additional trunk lateral line organs.
- Published
- 1996
- Full Text
- View/download PDF
43. Mutations affecting somite formation and patterning in the zebrafish, Danio rerio.
- Author
-
van Eeden FJ, Granato M, Schach U, Brand M, Furutani-Seiki M, Haffter P, Hammerschmidt M, Heisenberg CP, Jiang YJ, Kane DA, Kelsh RN, Mullins MC, Odenthal J, Warga RM, Allende ML, Weinberg ES, and Nüsslein-Volhard C
- Subjects
- Animals, Gene Expression Regulation, Developmental, Mesoderm physiology, Motor Neurons pathology, Muscles cytology, Muscles embryology, Somites cytology, Zebrafish anatomy & histology, Body Patterning genetics, Mutation, Somites physiology, Zebrafish embryology, Zebrafish genetics
- Abstract
Somitogenesis is the basis of segmentation of the mesoderm in the trunk and tail of vertebrate embryos. Two groups of mutants with defects in this patterning process have been isolated in our screen for zygotic mutations affecting the embryonic development of the zebrafish (Danio rerio). In mutants of the first group, boundaries between individual somites are invisible early on, although the paraxial mesoderm is present. Later, irregular boundaries between somites are present. Mutations in fused somites (fss) and beamter (bea) affect all somites, whereas mutations in deadly seven (des), after eight (aei) and white tail (wit) only affect the more posterior somites. Mutants of all genes but wit are homozygous viable and fertile. Skeletal stainings and the expression pattern of myoD and snail1 suggest that anteroposterior patterning within individual somites is abnormal. In the second group of mutants, formation of the horizontal myoseptum, which separates the dorsal and ventral part of the myotome, is reduced. Six genes have been defined in this group (you-type genes). you-too mutants show the most severe phenotype; in these the adaxial cells, muscle pioneers and the primary motoneurons are affected, in addition to the horizontal myoseptum. The horizontal myoseptum is also missing in mutants that lack a notochord. The similarity of the somite phenotype in mutants lacking the notochord and in the you-type mutants suggests that the genes mutated in these two groups are involved in a signaling pathway from the notochord, important for patterning of the somites.
- Published
- 1996
- Full Text
- View/download PDF
44. Genetic analysis of fin formation in the zebrafish, Danio rerio.
- Author
-
van Eeden FJ, Granato M, Schach U, Brand M, Furutani-Seiki M, Haffter P, Hammerschmidt M, Heisenberg CP, Jiang YJ, Kane DA, Kelsh RN, Mullins MC, Odenthal J, Warga RM, and Nüsslein-Volhard C
- Subjects
- Animals, Extremities growth & development, Larva genetics, Larva growth & development, Pectoralis Muscles, Phenotype, Skin embryology, Tail embryology, Zebrafish growth & development, Extremities embryology, Mutation, Zebrafish embryology, Zebrafish genetics
- Abstract
In the zebrafish, Danio rerio, a caudal and pectoral fin fold develop during embryogenesis. At larval stages the caudal fin fold is replaced by four different fins, the unpaired anal, dorsal and tail fins. In addition the paired pelvic fins are formed. We have identified a total of 118 mutations affecting larval fin formation. Mutations in 11 genes lead to abnormal morphology or degeneration of both caudal and pectoral fin folds. Most mutants survive to adulthood and form a surprisingly normal complement of adult fins. Mutations in nine genes result in an increased or reduced size of the pectoral fins. Interestingly, in mutants of one of these genes, dackel (dak), pectoral fin buds form initially, but later the fin epithelium fails to expand. Expression of sonic hedgehog mRNA in the posterior mesenchyme of the pectoral fin bud is initiated in dak embryos, but not maintained. Mutations in five other genes affect adult fin but not larval fin development. Two mutants, longfin (lof) and another longfin (alf) have generally longer fins. Stein und bein (sub) has reduced dorsal and pelvic fins, whereas finless (fls) and wanda (wan) mutants affect all adult fins. Finally, mutations in four genes causing defects in embryonic skin formation will be briefly reported.
- Published
- 1996
- Full Text
- View/download PDF
45. The identification of genes with unique and essential functions in the development of the zebrafish, Danio rerio.
- Author
-
Haffter P, Granato M, Brand M, Mullins MC, Hammerschmidt M, Kane DA, Odenthal J, van Eeden FJ, Jiang YJ, Heisenberg CP, Kelsh RN, Furutani-Seiki M, Vogelsang E, Beuchle D, Schach U, Fabian C, and Nüsslein-Volhard C
- Subjects
- Animals, Crosses, Genetic, Embryonic Development, Gene Expression Regulation, Developmental, Genetic Complementation Test, Male, Mutagenesis, Phenotype, Zebrafish growth & development, Genes, Zebrafish embryology, Zebrafish genetics
- Abstract
In a large-scale screen, we isolated mutants displaying a specific visible phenotype in embryos or early larvae of the zebrafish, Danio rerio. Males were mutagenized with ethylnitrosourea (ENU) and F2 families of single pair matings between sibling F1 fish, heterozygous for a mutagenized genome, were raised. Egg lays were obtained from several crosses between F2 siblings, resulting in scoring of 3857 mutagenized genomes. F3 progeny were scored at the second, third and sixth day of development, using a stereomicroscope. In a subsequent screen, fixed embryos were analyzed for correct retinotectal projection. A total of 4264 mutants were identified. Two thirds of the mutants displaying rather general abnormalities were eventually discarded. We kept and characterized 1163 mutants. In complementation crosses performed between mutants with similar phenotypes, 894 mutants have been assigned to 372 genes. The average allele frequency is 2.4. We identified genes involved in early development, notochord, brain, spinal cord, somites, muscles, heart, circulation, blood, skin, fin, eye, otic vesicle, jaw and branchial arches, pigment pattern, pigment formation, gut, liver, motility and touch response. Our collection contains alleles of almost all previously described zebrafish mutants. From the allele frequencies and other considerations we estimate that the 372 genes defined by the mutants probably represent more than half of all genes that could have been discovered using the criteria of our screen. Here we give an overview of the spectrum of mutant phenotypes obtained, and discuss the limits and the potentials of a genetic saturation screen in the zebrafish.
- Published
- 1996
- Full Text
- View/download PDF
46. Mutations affecting the cardiovascular system and other internal organs in zebrafish.
- Author
-
Chen JN, Haffter P, Odenthal J, Vogelsang E, Brand M, van Eeden FJ, Furutani-Seiki M, Granato M, Hammerschmidt M, Heisenberg CP, Jiang YJ, Kane DA, Kelsh RN, Mullins MC, and Nüsslein-Volhard C
- Subjects
- Animals, Body Patterning genetics, Cardiomegaly embryology, Cardiomegaly genetics, Embryonic Development, Heart Atria abnormalities, Heart Atria embryology, Heart Defects, Congenital embryology, Heart Defects, Congenital genetics, Heart Rate genetics, Heart Ventricles abnormalities, Heart Ventricles embryology, Intestines abnormalities, Intestines embryology, Kidney abnormalities, Kidney embryology, Liver abnormalities, Liver embryology, Myocardial Contraction genetics, Phenotype, Cardiovascular System embryology, Mutation, Zebrafish embryology, Zebrafish genetics
- Abstract
In a screen for early developmental mutants of the zebrafish, we have identified mutations specifically affecting the internal organs. We identified 53 mutations affecting the cardiovascular system. Nine of them affect specific landmarks of heart morphogenesis. Mutations in four genes cause a failure in the fusion of the bilateral heart primordia, resulting in cardia bifida. In lonely atrium, no heart venticle is visible and the atrium is directly fused to the outflow tract. In the overlooped mutant, the relative position of the two heart chambers is distorted. The heart is enormously enlarged in the santa mutant. In two mutants, scotch tape and superglue, the cardiac jelly between the two layers of the heart is significantly reduced. We also identified a number of mutations affecting the function of the heart. The mutations affecting heart function can be subdivided into two groups, one affecting heart contraction and another affecting the rhythm of the heart beat. Among the contractility group of mutants are 5 with no heart beat at all and 15 with a reduced heart beat of one or both chambers. 6 mutations are in the rhythmicity group and specifically affect the beating pattern of the heart. Mutations in two genes, bypass and kurzschluss, cause specific defects in the circulatory system. In addition to the heart mutants, we identified 23 mutations affecting the integrity of the liver, the intestine or the kidney. In this report, we demonstrate that it is feasible to screen for genes specific for the patterning or function of certain internal organs in the zebrafish. The mutations presented here could serve as an entry point to the establishment of a genetic hierarchy underlying organogenesis.
- Published
- 1996
- Full Text
- View/download PDF
47. Genes involved in forebrain development in the zebrafish, Danio rerio.
- Author
-
Heisenberg CP, Brand M, Jiang YJ, Warga RM, Beuchle D, van Eeden FJ, Furutani-Seiki M, Granato M, Haffter P, Hammerschmidt M, Kane DA, Kelsh RN, Mullins MC, Odenthal J, and Nusslein-Volhard C
- Subjects
- Animals, Body Patterning genetics, Cerebral Ventricles embryology, Ectoderm cytology, Ectoderm physiology, Gastrula physiology, Genetic Linkage, Mice, Mutagenesis, Nervous System embryology, Phenotype, Telencephalon embryology, Genes, Prosencephalon embryology, Zebrafish embryology, Zebrafish genetics
- Abstract
We identified four zebrafish mutants with defects in forebrain induction and patterning during embryogenesis. The four mutants define three genes: masterblind (mbl), silberblick (slb), and knollnase (kas). In mbl embryos, the anterior forebrain acquires posterior forebrain characteristics: anterior structures such as the eyes, olfactory placodes and the telencephalon are missing, whereas the epiphysis located in the posterior forebrain is expanded. In slb embryos, the extension of the embryonic axis is initially delayed and eventually followed by a partial fusion of the eyes. Finally, in kas embryos, separation of the telencephalic primordia is incomplete and dorsal midline cells fail to form a differentiated roof plate. Analysis of the mutant phenotypes indicates that we have identified genes essential for the specification of the anterior forebrain (mbl), positioning of the eyes (slb) and differentiation of the roof plate (kas). In an appendix to this study we list mutants showing alterations in the size of the eyes and abnormal differentiation of the lenses.
- Published
- 1996
- Full Text
- View/download PDF
48. Jaw and branchial arch mutants in zebrafish II: anterior arches and cartilage differentiation.
- Author
-
Piotrowski T, Schilling TF, Brand M, Jiang YJ, Heisenberg CP, Beuchle D, Grandel H, van Eeden FJ, Furutani-Seiki M, Granato M, Haffter P, Hammerschmidt M, Kane DA, Kelsh RN, Mullins MC, Odenthal J, Warga RM, and Nüsslein-Volhard C
- Subjects
- Animals, Branchial Region abnormalities, Cartilage abnormalities, Cartilage pathology, Cell Division genetics, Extracellular Matrix pathology, Head and Neck Neoplasms embryology, Head and Neck Neoplasms genetics, Head and Neck Neoplasms pathology, Larva, Phenotype, Skull embryology, Branchial Region embryology, Cartilage embryology, Jaw embryology, Mutation, Zebrafish embryology, Zebrafish genetics
- Abstract
In a large scale screen for mutants that affect the early development of the zebrafish, 109 mutants were found that cause defects in the formation of the jaw and the more posterior pharyngeal arches. Here we present the phenotypic description and results of the complementation analysis of mutants belonging to two major classes: (1) mutants with defects in the mandibular and hyoid arches and (2) mutants with defects in cartilage differentiation and growth in all arches. Mutations in four of the genes identified during the screen show specific defects in the first two arches and leave the more posterior pharyngeal arches largely unaffected (schmerle, sucker, hoover and sturgeon). In these mutants ventral components of the mandibular and hyoid arches are reduced (Meckel's cartilage and ceratohyal cartilage) whereas dorsal structures (palatoquadrate and hyosymplectic cartilages) are of normal size or enlarged. Thus, mutations in single genes cause defects in the formation of first and second arch structures but also differentially affect development of the dorsal and ventral structures within one arch. In 27 mutants that define at least 8 genes, the differentiation of cartilage and growth is affected. In hammerhead mutants particularly the mesodermally derived cartilages are reduced, whereas jellyfish mutant larvae are characterized by a severe reduction of all cartilaginous elements, leaving only two pieces in the position of the ceratohyal cartilages. In all other mutant larvae all skeletal elements are present, but consist of smaller and disorganized chondrocytes. These mutants also exhibit shortened heads and reduced pectoral fins. In homozygous knorrig embryos, tumor-like outgrowths of chondrocytes occur along the edges of all cartilaginous elements. The mutants presented here may be valuable tools for elucidating the genetic mechanisms that underlie the development of the mandibular and the hyoid arches, as well as the process of cartilage differentiation.
- Published
- 1996
- Full Text
- View/download PDF
49. Mutations affecting development of the midline and general body shape during zebrafish embryogenesis.
- Author
-
Brand M, Heisenberg CP, Warga RM, Pelegri F, Karlstrom RO, Beuchle D, Picker A, Jiang YJ, Furutani-Seiki M, van Eeden FJ, Granato M, Haffter P, Hammerschmidt M, Kane DA, Kelsh RN, Mullins MC, Odenthal J, and Nüsslein-Volhard C
- Subjects
- Animals, Axons physiology, Brain embryology, Brain pathology, Embryo, Nonmammalian anatomy & histology, Embryonic Development, Genetic Complementation Test, Mesoderm pathology, Motor Neurons pathology, Nervous System embryology, Zebrafish genetics, Body Patterning genetics, Gene Expression Regulation, Developmental, Mutation, Zebrafish anatomy & histology, Zebrafish embryology
- Abstract
Tissues of the dorsal midline of vertebrate embryos, such as notochord and floor plate, have been implicated in inductive interactions that pattern the neural tube and somites. In our screen for embryonic visible mutations in the zebrafish we found 113 mutations in more than 27 genes with altered body shape, often with additional defects in CNS development. We concentrated on a subgroup of mutations in ten genes (the midline-group) that cause defective development of the floor plate. By using floor plate markers, such as the signaling molecule sonic hedgehog, we show that the schmalspur (sur) gene is needed for early floor plate development, similar to one-eyed-pinhead (oep) and the previously described cyclops (cyc) gene. In contrast to oep and cyc, sur embryos show deletions of ventral CNS tissue restricted to the mid- and hindbrain, whereas the forebrain appears largely unaffected. In the underlying mesendodermal tissue of the head, sur is needed only for development of the posterior prechordal plate, whereas oep and cyc are required for both anterior and posterior prechordal plate development. Our analysis of sur mutants suggests that defects within the posterior prechordal plate may cause aberrant development of ventral CNS structures in the mid- and hindbrain. Later development of the floor plate is affected in mutant chameleon, you-too, sonic-you, iguana, detour, schmalhans and monorail embryos; these mutants often show additional defects in tissues that are known to depend on signals from notochord and floor plate. For example, sur, con and yot mutants show reduction of motor neurons; median deletions of brain tissue are seen in sur, con and yot embryos; and cyc, con, yot, igu and dtr mutants often show no or abnormal formation of the optic chiasm. We also find fusions of the ventral neurocranium for all midline mutants tested, which may reveal a hitherto unrecognized function of the midline in influencing differentiation of neural crest cells at their destination. As a working hypothesis, we propose that midline-group genes may act to maintain proper structure and inductive function of zebrafish midline tissues.
- Published
- 1996
- Full Text
- View/download PDF
50. Zebrafish pigmentation mutations and the processes of neural crest development.
- Author
-
Kelsh RN, Brand M, Jiang YJ, Heisenberg CP, Lin S, Haffter P, Odenthal J, Mullins MC, van Eeden FJ, Furutani-Seiki M, Granato M, Hammerschmidt M, Kane DA, Warga RM, Beuchle D, Vogelsang L, and Nüsslein-Volhard C
- Subjects
- Adaptation, Physiological genetics, Animals, Body Patterning genetics, Cell Count, Cell Differentiation genetics, Chromatophores metabolism, Chromatophores pathology, Chromatophores physiology, Larva, Melanins biosynthesis, Melanins genetics, Melanophores pathology, Pigments, Biological genetics, Mutation, Neural Crest embryology, Pigmentation genetics, Zebrafish embryology, Zebrafish genetics
- Abstract
Neural crest development involves cell-fate specification, proliferation, patterned cell migration, survival and differentiation. Zebrafish neural crest derivatives include three distinct chromatophores, which are well-suited to genetic analysis of their development. As part of a large-scale mutagenesis screen for embryonic/early larval mutations, we have isolated 285 mutations affecting all aspects of zebrafish larval pigmentation. By complementation analysis, we define 94 genes. We show here that comparison of their phenotypes permits classification of these mutations according to the types of defects they cause, and these suggest which process of neural crest development is probably affected. Mutations in eight genes affect the number of chromatophores: these include strong candidates for genes necessary for the processes of pigment cell specification and proliferation. Mutations in five genes remove part of the wild-type pigment pattern, and suggest a role in larval pigment pattern formation. Mutations in five genes show ectopic chromatophores in distinct sites, and may have implications for chromatophore patterning and proliferation. 76 genes affect pigment or morphology of one or more chromatophore types: these mutations include strong candidates for genes important in various aspects of chromatophore differentiation and survival. In combination with the embryological advantages of zebrafish, these mutations should permit cellular and molecular dissection of many aspects of neural crest development.
- Published
- 1996
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.