44 results on '"Fredericus J.M. van Eeden"'
Search Results
2. A method for high-throughput PCR-based genotyping of larval zebrafish tail biopsies
- Author
-
Robert N. Wilkinson, Stone Elworthy, Philip W. Ingham, and Fredericus J.M. van Eeden
- Subjects
zebrafish ,embryo ,genotyping ,PCR ,fin clip ,Biology (General) ,QH301-705.5 - 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
3. Data from A Zebrafish Model to Study and Therapeutically Manipulate Hypoxia Signaling in Tumorigenesis
- Author
-
Fredericus J.M. van Eeden, Simon S. Cross, Stephen A. Renshaw, Sarah S. Walmsley, Ellen van Rooijen, Stone Elworthy, Sarah McKee, Philip M. Elks, Emma C. Judson, and Kirankumar Santhakumar
- 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. Cancer Res; 72(16); 4017–27. ©2012 AACR.
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Figures 1-9 from A Zebrafish Model to Study and Therapeutically Manipulate Hypoxia Signaling in Tumorigenesis
- Author
-
Fredericus J.M. van Eeden, Simon S. Cross, Stephen A. Renshaw, Sarah S. Walmsley, Ellen van Rooijen, Stone Elworthy, Sarah McKee, Philip M. Elks, Emma C. Judson, and Kirankumar Santhakumar
- Abstract
PDF file - 817K, Figure S1 shows in vivo expression of phd3::EGFP in wild-type and vhl-/- Figure S2 shows that phd3::EGFP mRNA expression recapitulates endogenous phd3 expression. Figure S3 shows that EGFP expression in phd3::EGFP embryos is Hif dependent. Figure S4 shows that EGFP expression in phd3::EGFP embryos can be activated by inhibitors of PHD hydroxylases. Figure S5 shows the half life of the fluorescence of the phd3::EGFP line. Figure S6 shows that the phd3::EGFP transgene acts as an in vivo marker for detecting LOH events at the vhl locus in vhl+/- fish. Figure S7 shows morphology of gonadal tumors after DMBA treatment. Figure S8 shows the sequence analysis of the vhl locus in dissected tumors. Figure S9 shows that vhl mRNA levels are decreased in EGFP+ tumors in vhl+/- fish.
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Figure Legends 1-9 from A Zebrafish Model to Study and Therapeutically Manipulate Hypoxia Signaling in Tumorigenesis
- Author
-
Fredericus J.M. van Eeden, Simon S. Cross, Stephen A. Renshaw, Sarah S. Walmsley, Ellen van Rooijen, Stone Elworthy, Sarah McKee, Philip M. Elks, Emma C. Judson, and Kirankumar Santhakumar
- Abstract
PDF file - 438K
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Tables 1-4 from A Zebrafish Model to Study and Therapeutically Manipulate Hypoxia Signaling in Tumorigenesis
- Author
-
Fredericus J.M. van Eeden, Simon S. Cross, Stephen A. Renshaw, Sarah S. Walmsley, Ellen van Rooijen, Stone Elworthy, Sarah McKee, Philip M. Elks, Emma C. Judson, and Kirankumar Santhakumar
- Abstract
PDF file - 56K, Table S1, shows relative increase in EGFP fluorescence levels of phd3: EGFP in vhl-/- embryos vs wild-type. Table S2 shows the effect of DMBA treatment on the rates of LOH for the vhl locus in 2.5 month old wild-type and vhl+/- fish. Table S3 shows the increased number of macroscopic phd3: GFP positive areas induced by DMBA vhl-/+ adults as compared to wild-type, they are likely to correspond to vhl-/- tumors. Table S4 shows preferential induction of renal tubule proliferation in vhl heterozygotes.
- Published
- 2023
- Full Text
- View/download PDF
7. Asymmetric Hapln1a drives regionalized cardiac ECM expansion and promotes heart morphogenesis in zebrafish development
- Author
-
Farah Hussein, Timothy J. A. Chico, Robert N. Wilkinson, Eric J. G. Pollitt, Jeroen Bakkers, Emily S. Noël, Christopher J Derrick, Juliana Sánchez-Posada, Fredericus J.M. van Eeden, Aaron M Savage, Federico Tessadori, and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
Heart morphogenesis ,Physiology ,Heart malformation ,Morphogenesis ,Developmental Cardiology ,Heart development ,Extracellular matrix ,Animals, Genetically Modified ,Physiology (medical) ,symbols.heraldic_charge ,Animals ,Heart looping ,AcademicSubjects/MED00200 ,Hyaluronic Acid ,Zebrafish ,Body Patterning ,Extracellular Matrix Proteins ,biology ,Myocardium ,Heart shape ,Laterality ,Gene Expression Regulation, Developmental ,Heart ,Original Articles ,Zebrafish Proteins ,biology.organism_classification ,Cell biology ,Mutation ,symbols ,Proteoglycans ,Cardiology and Cardiovascular Medicine ,Transcriptome ,Signal Transduction - 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., Graphical Abstract
- Published
- 2021
8. Ciliopathy genes are required for apical secretion of Cochlin, an otolith crystallization factor
- Author
-
Zhou Zhu, Eleanor Markham, Fredericus J.M. van Eeden, Robert A. Hirst, Jarema Malicki, Xiaoming Fang, Eleni Leventea, and Yulia Nikolaeva
- Subjects
0301 basic medicine ,Ciliopathies ,03 medical and health sciences ,Otolithic Membrane ,0302 clinical medicine ,medicine ,Animals ,Secretion ,Amino Acid Sequence ,Cilia ,Bardet-Biedl Syndrome ,Secretory pathway ,Zebrafish ,Vestibular system ,Extracellular Matrix Proteins ,Multidisciplinary ,Base Sequence ,Chemistry ,Cilium ,Homozygote ,Gene Expression Regulation, Developmental ,Epistasis, Genetic ,Biological Sciences ,Zebrafish Proteins ,medicine.disease ,Cell biology ,Transport protein ,Ciliopathy ,030104 developmental biology ,medicine.anatomical_structure ,Phenotype ,Mutation ,Basal lamina ,sense organs ,Crystallization ,030217 neurology & neurosurgery - Abstract
Here, we report that important regulators of cilia formation and ciliary compartment-directed protein transport function in secretion polarity. Mutations in cilia genes cep290 and bbs2, involved in human ciliopathies, affect apical secretion of Cochlin, a major otolith component and a determinant of calcium carbonate crystallization form. We show that Cochlin, defective in human auditory and vestibular disorder, DFNA9, is secreted from small specialized regions of vestibular system epithelia. Cells of these regions secrete Cochlin both apically into the ear lumen and basally into the basal lamina. Basally secreted Cochlin diffuses along the basal surface of vestibular epithelia, while apically secreted Cochlin is incorporated into the otolith. Mutations in a subset of ciliopathy genes lead to defects in Cochlin apical secretion, causing abnormal otolith crystallization and behavioral defects. This study reveals a class of ciliary proteins that are important for the polarity of secretion and delineate a secretory pathway that regulates biomineralization.
- Published
- 2021
9. Transgenesis, mutagenesis, knockdown, and genetic colony management
- Author
-
Fredericus J.M. van Eeden, Francesco Argenton, Natascia Tiso, and Claire E. Allen
- Subjects
animal structures ,Killifish ,Danio ,Mutagenesis (molecular biology technique) ,Techniques of genetic engineering ,Computational biology ,Biology ,Stickleback ,biology.organism_classification ,Genome ,Forward genetics ,Medaka ,Goldfish ,Swordtail Fish ,Zebrafish, Medaka, Killifish, Swordtail Fish, Cavefish, Stickleback, Goldfish, Danionella Translucida ,Cavefish ,Danionella Translucida ,Zebrafish ,Gene ,Genotyping - Abstract
Genetic engineering techniques have rapidly developed since the first large-scale forward genetics screen was developed in 1996, which generated a spectrum of phenotypic zebrafish lines with mutations in many essential genes for normal development. Management of this original collection was relatively straightforward and embryonic analysis was the focus. Adults were heterozygous in their genotype, and phenotypes were primarily within homozygous embryos. Homozygous fish were rarely viable and reporter lines were in their molecular infancy. Genotyping methods were not available since extensive molecular investigation was required to piece together the pathways involved. Mapping and genetic characterization of mutations was propelled forward in the early 2000s by the full sequencing of the Danio rerio genome. This chapter aims to provide a comprehensive overview of the ethical issues in zebrafish (Danio rerio) handling and the most recent practices for the generation and management of zebrafish transgenic, mutant, and mosaic lines. The discussion will take into account recent legislation, international publications, and standard guidelines applied in world reference centers such as the Zebrafish International Resource Center and the European Zebrafish Resource Center, as well as our personal experience in the management of national-level facilities. A set of schemes will be included to provide the reader with an immediate overview of the main operational phases in the different workflows.
- Published
- 2021
10. Identification of additional outer segment targeting signals in zebrafish rod opsin
- Author
-
Andrew A. Peden, Fredericus J.M. van Eeden, Jarema Malicki, and Xiaoming Fang
- Subjects
Opsin ,Rhodopsin ,genetic structures ,biology ,Cilium ,Xenopus ,Cell Biology ,biology.organism_classification ,Rod Cell Outer Segment ,eye diseases ,Opsin transport ,Transmembrane protein ,Cell biology ,Protein Transport ,biology.protein ,Compartment (development) ,Animals ,Humans ,Photoreceptor Cells ,sense organs ,Zebrafish ,Photoreceptor Cells, Vertebrate - Abstract
In vertebrate photoreceptors, opsins are highly concentrated in a morphologically distinct ciliary compartment known as the outer segment (OS). Opsin is synthesized in the cell body and transported to the OS at a remarkable rate of 100 to 1000 molecules per second. Opsin transport defects contribute to photoreceptor loss and blindness in human ciliopathies. Previous studies revealed that the rhodopsin C-terminal tail, of 44 amino acids, is sufficient to mediate OS targeting in Xenopus photoreceptors. Here, we show that, although the Xenopus C-terminus retains this function in zebrafish, the homologous zebrafish sequence is not sufficient to target opsin to the OS. This functional difference is largely caused by a change of a single amino acid present in Xenopus but not in other vertebrates examined. Furthermore, we find that sequences in the third intracellular cytoplasmic loop (IC3) and adjacent regions of transmembrane helices 6 and 7 are also necessary for opsin transport in zebrafish. Combined with the cytoplasmic tail, these sequences are sufficient to target opsin to the ciliary compartment.
- Published
- 2020
11. Asymmetric Hapln1a drives regionalised cardiac ECM expansion and promotes heart morphogenesis during zebrafish development
- Author
-
Juliana Sánchez-Posada, Farah Hussein, Aaron M Savage, Christopher J Derrick, Robert N. Wilkinson, Federico Tessadori, Eric J. G. Pollitt, Fredericus J.M. van Eeden, Timothy J. A. Chico, Jeroen Bakkers, and Emily S. Noël
- Subjects
Extracellular matrix ,Heart morphogenesis ,Proteoglycan ,biology ,Link protein ,Cardiac looping ,biology.protein ,biology.organism_classification ,Embryonic stem cell ,Zebrafish ,Endocardium ,Cell biology - Abstract
The mature vertebrate heart develops from a simple linear cardiac tube during early development through a series of highly asymmetric morphogenetic processes including cardiac looping and chamber ballooning. While the directionality of heart morphogenesis is partly controlled by embryonic laterality signals, previous studies have suggested that these extrinsic laterality cues interact with tissue-intrinsic signals in the heart to ensure robust asymmetric cardiac morphogenesis. Using livein vivoimaging of zebrafish embryos we describe a left-sided, chamber-specific expansion of the extracellular matrix (ECM) between the myocardium and endocardium at early stages of heart morphogenesis. We use Tomo-seq, a spatial transcriptomic approach, to identify transient and regionalised expression ofhyaluronan and proteoglycan link protein 1a(hapln1a),encoding an ECM cross-linking protein, in the heart tube prior to cardiac looping overlapping with regionalised ECM expansion. Loss- and gain-of-function experiments demonstrate that regionalised Hapln1a promotes heart morphogenesis through regional modulation of ECM thickness in the heart tube. Finally, we show that while induction of asymmetrichapln1aexpression is independent of embryonic left-right asymmetry, these laterality cues are required to orient thehapln1a-expressing cells asymmetrically along the left-right axis of the heart tube.Together, we propose a model whereby laterality cues positionhapln1aexpression on the left of the heart tube, and this asymmetric Hapln1a deposition drives ECM asymmetry and subsequently promotes robust asymmetric cardiac morphogenesis.
- Published
- 2019
- Full Text
- View/download PDF
12. Bidirectional crosstalk between Hypoxia-Inducible Factor and glucocorticoid signalling in zebrafish larvae
- Author
-
Vincent T. Cunliffe, Fredericus J.M. van Eeden, Kirankumar Santhakumar, Karl-Heinz Storbeck, Eleanor Markham, Nils Krone, Nan Li, and Davide Marchi
- Subjects
Life Cycles ,Embryology ,Cancer Research ,Embryo, Nonmammalian ,Pulmonology ,Gene Expression ,QH426-470 ,Biochemistry ,Cortisol ,Animals, Genetically Modified ,Larvae ,0302 clinical medicine ,Glucocorticoid receptor ,Mineralocorticoid receptor ,Cell Signaling ,Medicine and Health Sciences ,Lipid Hormones ,Animal Anatomy ,Receptor ,Zebrafish ,Genetics (clinical) ,Tails ,0303 health sciences ,Gene Expression Regulation, Developmental ,Eukaryota ,Animal Models ,3. Good health ,Cell biology ,Crosstalk (biology) ,Experimental Organism Systems ,Cellular Crosstalk ,Hypoxia-inducible factors ,Osteichthyes ,Larva ,Vertebrates ,Hypoxia-Inducible Factor 1 ,medicine.symptom ,Anatomy ,hormones, hormone substitutes, and hormone antagonists ,Glucocorticoid ,Signal Transduction ,Research Article ,medicine.drug ,Inflammation ,Biology ,Research and Analysis Methods ,03 medical and health sciences ,Receptors, Glucocorticoid ,Model Organisms ,Downregulation and upregulation ,In vivo ,Medical Hypoxia ,Genetics ,medicine ,Animals ,Glucocorticoids ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology ,Steroid Hormones ,Tumor Suppressor Proteins ,Aryl Hydrocarbon Receptor Nuclear Translocator ,Embryos ,Organisms ,Biology and Life Sciences ,Receptor Cross-Talk ,Cell Biology ,Zebrafish Proteins ,biology.organism_classification ,Hormones ,Fish ,Animal Studies ,Zoology ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
In the last decades in vitro studies highlighted the potential for crosstalk between Hypoxia-Inducible Factor-(HIF) and glucocorticoid-(GC) signalling pathways. However, how this interplay precisely occurs in vivo is still debated. Here, we use zebrafish larvae (Danio rerio) to elucidate how and to what degree hypoxic signalling affects the endogenous glucocorticoid pathway and vice versa, in vivo. Firstly, our results demonstrate that in the presence of upregulated HIF signalling, both glucocorticoid receptor (Gr) responsiveness and endogenous cortisol levels are repressed in 5 days post fertilisation larvae. In addition, despite HIF activity being low at normoxia, our data show that it already impedes both glucocorticoid activity and levels. Secondly, we further analysed the in vivo contribution of glucocorticoids to HIF activity. Interestingly, our results show that both glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) play a key role in enhancing it. Finally, we found indications that glucocorticoids promote HIF signalling via multiple routes. Cumulatively, our findings allowed us to suggest a model for how this crosstalk occurs in vivo., Author summary Hypoxia is a common pathophysiological condition to which cells must rapidly respond in order to prevent metabolic shutdown and subsequent death. This is achieved via the activity of Hypoxia-Inducible Factors (HIFs), which are key oxygen sensors that mediate the ability of the cell to cope with decreased oxygen levels. Although it aims to restore tissue oxygenation and perfusion, it can sometimes be maladaptive and contributes to a variety of pathological conditions including inflammation, tissue ischemia, stroke and growth of solid tumours. In this regard, synthetic glucocorticoids which are analogous to naturally occurring steroid hormones, have been used for decades as anti-inflammatory drugs for treating pathological conditions which are linked to hypoxia (i.e. asthma, rheumatoid arthritis, ischemic injury). Indeed, previous in vitro studies highlighted the presence of a crosstalk between HIF and glucocorticoids. However, how this interplay precisely occurs in an organism and what the molecular mechanism is behind it are questions that still remain unanswered. Here, we provide a thorough in vivo genetic analysis, which allowed us to propose a logical model of interaction between glucocorticoid and HIF signalling. In addition, our results are important because they suggest a new route to downregulate HIF for clinical purposes.
- Published
- 2019
- Full Text
- View/download PDF
13. TMEM33 regulates intracellular calcium homeostasis in renal tubular epithelial cells
- Author
-
Robert N. Wilkinson, Charbel El Boustany, Fabrice Duprat, Jonathan S. Marchant, Céline Moro, Ivana Y. Kuo, Barbara E. Ehrlich, Christophe Duranton, Aaron M Savage, Nicolas Picard, Gihan S. Gunaratne, Fredericus J.M. van Eeden, Amanda Patel, Isabelle Rubera, Dahui Li, Sophie Pagnotta, Hélène Duval, Sandra Lacas-Gervais, Malika Arhatte, Magali Plaisant, Anais Couvreux, Sophie Demolombe, Eric Honoré, Institut de pharmacologie moléculaire et cellulaire (IPMC), Université Nice Sophia Antipolis (... - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Centre National de la Recherche Scientifique (CNRS), University of Minnesota [Twin Cities] (UMN), University of Minnesota System, Centre National de la Recherche Scientifique (CNRS)-Université Nice Sophia Antipolis (... - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Université Côte d'Azur (UCA), Institut de signalisation, biologie du développement et cancer (ISBDC), Service d'anatomie et cytologie pathologiques [Rennes] = Anatomy and Cytopathology [Rennes], CHU Pontchaillou [Rennes], Laboratoire de Biologie Tissulaire et d'ingénierie Thérapeutique UMR 5305 (LBTI), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS), Laboratoire de PhysioMédecine Moléculaire (LP2M), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Centre National de la Recherche Scientifique (CNRS)-Université Côte d'Azur (UCA), Physiologie cellulaire et moléculaire des systèmes intégrés (PCMSI), Centre Commun de Microscopie Appliquée (CCMA), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA), University Medical Center Carl Gustav Carus, Dresden University of Technology Department of Conserva, COMUE Université Côte d'Azur (2015 - 2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015 - 2019) (COMUE UCA)-Centre National de la Recherche Scientifique (CNRS), COMUE Université Côte d'Azur (2015 - 2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015 - 2019) (COMUE UCA)-Université Côte d'Azur (UCA), Service d'anatomie et cytologie pathologiques [Rennes], Université de Rennes 1 (UR1), Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES)-Hôpital Pontchaillou-CHU Pontchaillou [Rennes], COMUE Université Côte d'Azur (2015 - 2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015 - 2019) (COMUE UCA)-Centre National de la Recherche Scientifique (CNRS)-Université Côte d'Azur (UCA), and COMUE Université Côte d'Azur (2015 - 2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015 - 2019) (COMUE UCA)
- Subjects
0301 basic medicine ,Embryo, Nonmammalian ,General Physics and Astronomy ,02 engineering and technology ,[SDV.BC.BC]Life Sciences [q-bio]/Cellular Biology/Subcellular Processes [q-bio.SC] ,Endoplasmic Reticulum ,urologic and male genital diseases ,[SDV.MHEP.UN]Life Sciences [q-bio]/Human health and pathology/Urology and Nephrology ,Calcium in biology ,Kidney Tubules, Proximal ,Mice ,RNA, Small Interfering ,lcsh:Science ,Zebrafish ,Mice, Knockout ,Multidisciplinary ,Chemistry ,Calcium signalling ,Acute kidney injury ,Acute Kidney Injury ,021001 nanoscience & nanotechnology ,Endoplasmic Reticulum Stress ,Polycystic Kidney, Autosomal Dominant ,3. Good health ,Cell biology ,Convoluted tubule ,Gene Knockdown Techniques ,0210 nano-technology ,endocrine system ,TRPP Cation Channels ,Science ,Autosomal dominant polycystic kidney disease ,chemistry.chemical_element ,Calcium ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Polycystic kidney disease ,medicine ,Animals ,Humans ,Calcium metabolism ,urogenital system ,Endoplasmic reticulum ,Cell Membrane ,Membrane Proteins ,Epithelial Cells ,General Chemistry ,Zebrafish Proteins ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,Mutation ,Unfolded protein response ,lcsh:Q ,Lysosomes ,HeLa Cells - Abstract
Mutations in the polycystins cause autosomal dominant polycystic kidney disease (ADPKD). Here we show that transmembrane protein 33 (TMEM33) interacts with the ion channel polycystin-2 (PC2) at the endoplasmic reticulum (ER) membrane, enhancing its opening over the whole physiological calcium range in ER liposomes fused to planar bilayers. Consequently, TMEM33 reduces intracellular calcium content in a PC2-dependent manner, impairs lysosomal calcium refilling, causes cathepsins translocation, inhibition of autophagic flux upon ER stress, as well as sensitization to apoptosis. Invalidation of TMEM33 in the mouse exerts a potent protection against renal ER stress. By contrast, TMEM33 does not influence pkd2-dependent renal cystogenesis in the zebrafish. Together, our results identify a key role for TMEM33 in the regulation of intracellular calcium homeostasis of renal proximal convoluted tubule cells and establish a causal link between TMEM33 and acute kidney injury., Polycystin-2 (PC2) is an ion channel commonly found mutated in autosomal dominant polycystic kidney disease. Here Arhatte et al. identify transmembrane protein 33 (TMEM33) as a regulator of PC2 function at the endoplasmic reticulum, and find that deletion of TMEM33 protects mice from acute kidney injury.
- Published
- 2019
- Full Text
- View/download PDF
14. foxc1a and foxc1b differentially regulate angiogenesis from arteries and veins by modulating Vascular Endothelial Growth Factor signalling
- Author
-
Teri Forey, Robert N. Wilkinson, Zhen Jiang, Fredericus J.M. van Eeden, Matthew Loose, Aaron M Savage, and Timothy J. A. Chico
- Subjects
0303 health sciences ,biology ,Angiogenesis ,Receptor expression ,030302 biochemistry & molecular biology ,Notch signaling pathway ,Hindbrain ,biology.organism_classification ,Cell biology ,Vascular endothelial growth factor ,03 medical and health sciences ,chemistry.chemical_compound ,chemistry ,Vascular endothelial growth factor C ,biology.protein ,FOXC2 ,Zebrafish ,030304 developmental biology - Abstract
The forkhead transcription factorsFoxc1andFoxc2are essential to establish intact vascular networks in mammals. How these genes interact with endothelial signalling pathways to exert their functions remains incompletely understood. We have generated novel zebrafish mutants infoxc1aandfoxc1b, the zebrafish orthologues of mammalianFoxc1, to determine their function during angiogenesis.foxc1amutants display abnormal formation of cranial veins including the primordial hindbrain channels (PHBC), reduced Vascular Endothelial Growth Factor (VEGF) receptor expression in these and loss of central arteries.foxc1bmutants are normal, whereasfoxc1a;foxc1bdouble mutants exhibit ectopic angiogenesis from trunk segmental arteries. Dll4/Notch signalling is reduced infoxc1a; foxc1bdouble mutant arteries and ectopic angiogenesis can be suppressed by induction of Notch or inhibition of Vegfc signalling. We conclude thatfoxc1aandfoxc1bplay compensatory and context-dependent roles to co-ordinate angiogenesis by promoting venous sprouting via induction of VEGF receptor expression whilst antagonising arterial sprouting by inducing Dll4/Notch signalling.foxc1a/bmediated induction of both pro- and anti-angiogenic axes of VEGF-Dll4/Notch negative feedback imparts competition to balance arterial and venous angiogenesis within developing vascular beds.Summary Statementfoxc1aandfoxc1bpromote angiogenesis from veins and suppress angiogenesis from arteries by promoting competing pro-angiogenic Vascular Endothelial Growth Factor signalling, and anti-angiogenic Dll4/Notch signalling in zebrafish embryos.
- Published
- 2018
- Full Text
- View/download PDF
15. Exploring the HIFs, buts and maybes of hypoxia signalling in disease: lessons from zebrafish models
- Author
-
Fredericus J.M. van Eeden, Stephen A. Renshaw, Sarah R. Walmsley, Philip M. Elks, and Annemarie H. Meijer
- Subjects
Aryl hydrocarbon receptor nuclear translocator ,Neuroscience (miscellaneous) ,lcsh:Medicine ,Medicine (miscellaneous) ,Review ,Disease ,Disease models ,Hypoxia-inducible factor ,Bioinformatics ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Species Specificity ,Immunology and Microbiology (miscellaneous) ,Basic Helix-Loop-Helix Transcription Factors ,lcsh:Pathology ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,Hypoxia ,Zebrafish ,Transcription factor ,030304 developmental biology ,0303 health sciences ,biology ,Aryl Hydrocarbon Receptor Nuclear Translocator ,lcsh:R ,Promoter ,Zebrafish Proteins ,Hypoxia (medical) ,Hypoxia-Inducible Factor 1, alpha Subunit ,biology.organism_classification ,Cell Hypoxia ,3. Good health ,Oxygen ,Cellular Microenvironment ,Hypoxia-inducible factors ,Infectious disease (medical specialty) ,Drug Design ,030220 oncology & carcinogenesis ,Models, Animal ,medicine.symptom ,Neuroscience ,Signal Transduction ,Transcription Factors ,lcsh:RB1-214 - 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., Summary: Hypoxia signalling, mediated by HIF, is a crucial pathway in many disease processes. Here, we review current knowledge of HIF signalling and disease, focusing on recent findings from zebrafish models.
- Published
- 2015
- Full Text
- View/download PDF
16. tmem33 is essential for VEGF-mediated endothelial calcium oscillations and angiogenesis
- Author
-
Angel L. Armesilla, Heather L. Wilson, Karishma Chhabria, Robert N. Wilkinson, Sathishkumar Kurusamy, Timothy J. A. Chico, Aaron M Savage, Ryan B. MacDonald, Fredericus J.M. van Eeden, Zhen Jiang, Hyejeong R. Kim, and Yan Chen
- Subjects
0301 basic medicine ,Embryo, Nonmammalian ,Angiogenesis ,Science ,Notch signaling pathway ,General Physics and Astronomy ,chemistry.chemical_element ,Neovascularization, Physiologic ,02 engineering and technology ,Calcium ,Endoplasmic Reticulum ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Animals ,Humans ,Calcium Signaling ,Phosphorylation ,lcsh:Science ,Extracellular Signal-Regulated MAP Kinases ,Zebrafish ,Calcium signaling ,Multidisciplinary ,biology ,Chemistry ,Vascular Endothelial Growth Factors ,Endoplasmic reticulum ,Endothelial Cells ,Membrane Proteins ,General Chemistry ,021001 nanoscience & nanotechnology ,biology.organism_classification ,Cell biology ,Vascular endothelial growth factor ,Vascular endothelial growth factor A ,030104 developmental biology ,Gene Knockdown Techniques ,Blood Vessels ,lcsh:Q ,0210 nano-technology - Abstract
Angiogenesis requires co-ordination of multiple signalling inputs to regulate the behaviour of endothelial cells (ECs) as they form vascular networks. Vascular endothelial growth factor (VEGF) is essential for angiogenesis and induces downstream signalling pathways including increased cytosolic calcium levels. Here we show that transmembrane protein 33 (tmem33), which has no known function in multicellular organisms, is essential to mediate effects of VEGF in both zebrafish and human ECs. We find that tmem33 localises to the endoplasmic reticulum in zebrafish ECs and is required for cytosolic calcium oscillations in response to Vegfa. tmem33-mediated endothelial calcium oscillations are critical for formation of endothelial tip cell filopodia and EC migration. Global or endothelial-cell-specific knockdown of tmem33 impairs multiple downstream effects of VEGF including ERK phosphorylation, Notch signalling and embryonic vascular development. These studies reveal a hitherto unsuspected role for tmem33 and calcium oscillations in the regulation of vascular development., Calcium signalling downstream of VEGF is essential for VEGF-induced angiogenesis. Here Savage et al. show that Transmembrane Protein 33 (TMEM33) is required for angiogenesis and the endothelial calcium response to VEGF, revealing a function for TMEM33 in multicellular organisms.
- Published
- 2017
17. Glucocorticoids promote Von Hippel Lindau degradation and Hif-1α stabilization
- Author
-
David Greenald, Francesco Argenton, Margherita Peron, Eleanor Markham, Mathavan Sinnakaruppan, Jane A. McKeating, Garrick K. Wilson, Laura Matthews, Andrea Vettori, Fredericus J.M. van Eeden, and Nicola Facchinello
- Subjects
0301 basic medicine ,medicine.medical_specialty ,von Hippel-Lindau Disease ,Ubiquitin-Protein Ligases ,Glucocorticoid signaling ,alpha Subunit ,Hypoxia-inducible factor ,Ligases ,Liver ,Metabolism ,Von Hippel Lindau ,Multidisciplinary ,03 medical and health sciences ,Glucocorticoid receptor ,Transcription (biology) ,Internal medicine ,medicine ,Glucose homeostasis ,Animals ,Humans ,Hypoxia ,Zebrafish ,Glucocorticoids ,Tissue homeostasis ,biology ,Biological Sciences ,biology.organism_classification ,Hypoxia-Inducible Factor 1, alpha Subunit ,Cell Hypoxia ,Cell biology ,030104 developmental biology ,Endocrinology ,Hypoxia-inducible factors ,Trans-Activators ,Hypoxia-Inducible Factor 1 ,Signal transduction ,Glucocorticoid ,hormones, hormone substitutes, and hormone antagonists ,medicine.drug ,glucocorticoid signaling ,hypoxia-inducible factor ,liver ,metabolism ,Protein Binding ,Signal Transduction - Abstract
Glucocorticoid (GC) and hypoxic transcriptional responses play a central role in tissue homeostasis and regulate the cellular response to stress and inflammation, highlighting the potential for cross-talk between these two signaling pathways. We present results from an unbiased in vivo chemical screen in zebrafish that identifies GCs as activators of hypoxia-inducible factors (HIFs) in the liver. GCs activated consensus hypoxia response element (HRE) reporters in a glucocorticoid receptor (GR)-dependent manner. Importantly, GCs activated HIF transcriptional responses in a zebrafish mutant line harboring a point mutation in the GR DNA-binding domain, suggesting a nontranscriptional route for GR to activate HIF signaling. We noted that GCs increase the transcription of several key regulators of glucose metabolism that contain HREs, suggesting a role for GC/HIF cross-talk in regulating glucose homeostasis. Importantly, we show that GCs stabilize HIF protein in intact human liver tissue and isolated hepatocytes. We find that GCs limit the expression of Von Hippel Lindau protein (pVHL), a negative regulator of HIF, and that treatment with the c-src inhibitor PP2 rescued this effect, suggesting a role for GCs in promoting c-src–mediated proteosomal degradation of pVHL. Our data support a model for GCs to stabilize HIF through activation of c-src and subsequent destabilization of pVHL.
- Published
- 2017
18. Repression of Hedgehog signalling is required for the acquisition of dorsolateral cell fates in the zebrafish otic vesicle
- Author
-
Tanya T. Whitfield, Fredericus J.M. van Eeden, and Katherine L. Hammond
- Subjects
Embryo, Nonmammalian ,animal structures ,Biology ,Polymerase Chain Reaction ,Antibodies ,Hemoglobins ,Mice ,Hearing ,Species Specificity ,Downregulation and upregulation ,medicine ,Animals ,Inner ear ,Molecular Biology ,Hedgehog ,Zebrafish ,Research Articles ,DNA Primers ,Embryo ,Anatomy ,biology.organism_classification ,Semicircular Canals ,Cell biology ,Transplantation ,Phenotype ,medicine.anatomical_structure ,Ear, Inner ,Mutation ,embryonic structures ,Collagen ,sense organs ,Otic vesicle ,Signal transduction ,Chickens ,Signal Transduction ,Developmental Biology - 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
19. Laminin-α4 and Integrin-Linked Kinase Mutations Cause Human Cardiomyopathy Via Simultaneous Defects in Cardiomyocytes and Endothelial Cells
- Author
-
Mohammad R. Toliat, Akinori Kimura, Nicholas J. Schork, Takeharu Hayashi, Peter Nürnberg, Jeroen Bakkers, Katrin Schäfer, Cornelia Schubert, Heinz-Peter Schultheiss, Jutta Schaper, Jeroen den Hertog, Gerd Hasenfuss, Ruben Postel, Dieter Kube, Ralph Knöll, Padmanabhan Vakeel, Kenton K. Murthy, Torbjorn Holm, Gudrun Knöll, Wolfgang Schaper, Brinda K. Rana, Gerrit Hennecke, Kenneth R. Chien, Erik Bos, Fredericus J.M. van Eeden, Andrei M. Vacaru, Peter J. Peters, Jianming Wang, and Ralph Krätzner
- Subjects
Male ,Models, Molecular ,Integrins ,Embryo, Nonmammalian ,Protein Conformation ,DNA Mutational Analysis ,Mutant ,medicine.disease_cause ,Epigenesis, Genetic ,Laminin ,Chlorocebus aethiops ,Protein Interaction Mapping ,Myocytes, Cardiac ,Zebrafish ,Genetics ,Mutation ,biology ,Chromosome Mapping ,Heart ,Middle Aged ,Extracellular Matrix ,Pedigree ,Cell biology ,Codon, Nonsense ,COS Cells ,Female ,Signal transduction ,Cardiology and Cardiovascular Medicine ,Protein Binding ,Adult ,Cardiomyopathy, Dilated ,Integrin ,Nonsense mutation ,Mutation, Missense ,Protein Serine-Threonine Kinases ,Transfection ,Physiology (medical) ,Cell Adhesion ,medicine ,Animals ,Humans ,Point Mutation ,Integrin-linked kinase ,Heart Failure ,Myocardium ,Endothelial Cells ,Oligonucleotides, Antisense ,Zebrafish Proteins ,Protein Structure, Tertiary ,Amino Acid Substitution ,biology.protein ,Genetic screen - 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-α4 (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 ( K d =5±3 μmol/L) and Arg1073X LAMA4 ( K d =1±0.2 μmol/L) mutants compared with the wild-type LAMA4 protein ( K d =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
20. klf2ash317 Mutant Zebrafish Do Not Recapitulate Morpholino-Induced Vascular and Haematopoietic Phenotypes
- Author
-
Richard Maguire, Carl Smythe, Robert N. Wilkinson, Richard Beniston, Timothy J. A. Chico, Stone Elworthy, Oliver Watson, Peter Novodvorsky, Caroline Gray, Scott Reeve, Alexander M.K. Rothman, Karen Plant, and Fredericus J.M. van Eeden
- Subjects
Morpholino ,Genotype ,Science ,Hematopoietic System ,Mutant ,Kruppel-Like Transcription Factors ,Biology ,Cardiovascular System ,Morpholinos ,03 medical and health sciences ,0302 clinical medicine ,Morphogenesis ,Animals ,Humans ,Zebrafish ,030304 developmental biology ,0303 health sciences ,Gene knockdown ,Multidisciplinary ,Wild type ,Gene Expression Regulation, Developmental ,Morphant ,Zebrafish Proteins ,biology.organism_classification ,Molecular biology ,Phenotype ,KLF2 ,Mutation ,Medicine ,030217 neurology & neurosurgery ,Signal Transduction ,Research Article - Abstract
Introduction and objectivesThe 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 resultsUsing 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.ConclusionsThe klf2ash317 mutation produces a truncated Klf2a protein but, unlike morpholino induced klf2a knockdown, does not affect cardiovascular development.
- Published
- 2015
21. Genetic variation in the zebrafish
- Author
-
Marco J. Koudijs, Ronald H.A. Plasterk, Victor Guryev, Eugene Berezikov, Fredericus J.M. van Eeden, Stephen L. Johnson, Edwin Cuppen, and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
Genetic Markers ,Genetics ,Genome ,Letter ,biology ,Genetic Linkage ,ved/biology ,ved/biology.organism_classification_rank.species ,Chromosome Mapping ,Locus (genetics) ,biology.organism_classification ,Polymorphism, Single Nucleotide ,Nucleotide diversity ,Species Specificity ,Genetic marker ,Genetic variation ,Animals ,RNA Editing ,Allele ,Model organism ,Genotyping ,Zebrafish ,Genetics (clinical) - 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 ∼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
22. Euthanizing zebrafish legally in Europe
- Author
-
Ana M. Valentim, Fredericus J.M. van Eeden, I. Anna S. Olsson, and Uwe Strähle
- Subjects
0301 basic medicine ,Internet privacy ,Fish species ,Legislation ,Biology ,Animal Welfare ,Biochemistry ,Ethics, Research ,03 medical and health sciences ,0302 clinical medicine ,Animal model ,Euthanasia, Animal ,Animal welfare ,Genetics ,Animals ,Science & Society ,Molecular Biology ,Zebrafish ,Research ethics ,business.industry ,Research ,biology.organism_classification ,Directive ,Biotechnology ,Europe ,030104 developmental biology ,Animal ethics ,business ,030217 neurology & neurosurgery - Abstract
Zebrafish have become an increasingly popular model organism in basic biological research. According to the European Commission's latest report on the number of animals used in research in the EU, the number of fish increased by 28.5% from 2008 to 2011 (310,307 more fish) [1] . The UK Home Office reported that fish were the second most used group of animals in research in 2015 in the UK (14%, 294,000 procedures), with zebrafish representing 50% of all fish species [2]. The growing use of fish in research is often regarded as a major achievement to replace mammalian model organisms, notably rodents [3]. However, the increasing relevance of zebrafish as an animal model creates an urgent need for techniques and methods that guarantee that any research with zebrafish can be carried out according to the same scientific and animal welfare standards as the laboratory rodents they are often replacing. ### Legal methods to euthanize fish From the moment zebrafish become free‐feeding larvae, any intervention on them is regulated by EU legislation. This means that any procedure done in these animals from about 5 days after fertilization must comply with the EU Directive 2010/63/EU on the protection of animals used for scientific purposes [4]. This includes euthanizing animals, which are no longer needed, whose suffering needs to be ended or whose organs are collected for further analysis. The European Directive lists only three methods for humanely killing fish: anesthetic overdose, concussion, or electrical stunning. Other methods can be used in unconscious animals. Each of these methods has clear limitations for research and for animal welfare. The zebrafish's small size makes efficient …
- Published
- 2016
- Full Text
- View/download PDF
23. Barentsz is essential for the posterior localization of oskar mRNA and colocalizes with it to the posterior pole
- Author
-
Fredericus J.M. van Eeden, Matthew J.D. Weston, Mark Petronczki, Daniel St Johnston, and Isabel M. Palacios
- Subjects
Male ,Molecular Sequence Data ,RNA-binding protein ,Biology ,oskar ,Microtubules ,Article ,03 medical and health sciences ,Oogenesis ,0302 clinical medicine ,Microtubule ,Animals ,Drosophila Proteins ,RNA, Messenger ,Cloning, Molecular ,Pole plasm assembly ,030304 developmental biology ,Recombination, Genetic ,0303 health sciences ,Sequence Homology, Amino Acid ,urogenital system ,Cell Polarity ,RNA-Binding Proteins ,Drosophila embryogenesis ,Drosophila ,axis formation ,pole plasm ,microtubules ,RNA transport ,Cell Biology ,Molecular biology ,Cell biology ,Phenotype ,Mutation ,Oocytes ,Pole plasm ,Insect Proteins ,Exon junction complex ,Kinesin ,Female ,Polymorphism, Restriction Fragment Length ,030217 neurology & neurosurgery - 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
24. Role of sonic hedgehog in branchiomotor neuron induction in zebrafish
- Author
-
Pascal Haffter, Kana Takahashi, Fredericus J.M. van Eeden, Anand Chandrasekhar, James T. Warren, John Y. Kuwada, and Heike E. Schauerte
- Subjects
Embryology ,Embryo, Nonmammalian ,animal structures ,Rhombomere ,Hindbrain ,Nervous System ,Transforming Growth Factor beta ,medicine ,Animals ,Hedgehog Proteins ,Sonic hedgehog ,Hedgehog ,Zebrafish ,Cell Nucleus ,Embryonic Induction ,Motor Neurons ,Neurons ,Genetics ,biology ,Intracellular Signaling Peptides and Proteins ,Gene Expression Regulation, Developmental ,Proteins ,Zebrafish Proteins ,Motor neuron ,biology.organism_classification ,Spinal cord ,Cell biology ,Rhombencephalon ,Branchial Region ,medicine.anatomical_structure ,Spinal Cord ,nervous system ,Mutation ,embryonic structures ,Trans-Activators ,biology.protein ,Neuron ,Developmental Biology - 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.
- Published
- 1998
- Full Text
- View/download PDF
25. Regulation ofnetrin-1aExpression by Hedgehog Proteins
- Author
-
Heike E. Schauerte, Fredericus J.M. van Eeden, Sara K. Pasquali, Reza Fazel, James D. Lauderdale, John Y. Kuwada, and Pascal Haffter
- Subjects
Central Nervous System ,Tail ,Blastomeres ,Embryo, Nonmammalian ,Recombinant Fusion Proteins ,Notochord ,Embryonic Development ,Biology ,Cellular and Molecular Neuroscience ,Netrin ,Morphogenesis ,medicine ,Animals ,Hedgehog Proteins ,Nerve Growth Factors ,Molecular Biology ,Hedgehog ,In Situ Hybridization ,Zebrafish ,Floor plate ,Tumor Suppressor Proteins ,Neural tube ,Gene Expression Regulation, Developmental ,Proteins ,Cell Biology ,Anatomy ,Netrin-1 ,Zebrafish Proteins ,Hedgehog signaling pathway ,Cell biology ,medicine.anatomical_structure ,Neurulation ,Somites ,Trans-Activators ,Head ,Neural plate ,Signal Transduction - 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.
- Published
- 1998
- Full Text
- View/download PDF
26. Zebrafish segmentation and pair-rule patterning
- Author
-
Pascal Haffter, Fredericus J.M. van Eeden, Scott A. Holley, and Christiane Nüsslein-Volhard
- Subjects
Genetics ,animal structures ,biology ,Embryogenesis ,Mutant ,Cell Biology ,biology.organism_classification ,Somite ,medicine.anatomical_structure ,Segment polarity gene ,Somitogenesis ,embryonic structures ,Paraxial mesoderm ,biology.protein ,medicine ,Sonic hedgehog ,Zebrafish ,Developmental Biology - 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. Dev. Genet. 23:65–76, 1998. © 1998 Wiley-Liss, Inc.
- Published
- 1998
- Full Text
- View/download PDF
27. The Zebrafish as a Model of Vascular Development and Disease
- Author
-
Fredericus J.M. van Eeden and Robert N. Wilkinson
- Subjects
Pathology ,medicine.medical_specialty ,animal structures ,Angiogenesis ,Vascular disease ,Vascular biology ,Disease ,Biology ,medicine.disease ,biology.organism_classification ,medicine.anatomical_structure ,Vasculogenesis ,Animal model ,embryonic structures ,medicine ,Neuroscience ,Zebrafish ,Blood vessel - 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.
- Published
- 2014
- Full Text
- View/download PDF
28. Positive and Negative Regulation of Gli Activity by Kif7 in the Zebrafish Embryo
- Author
-
Fredericus J.M. van Eeden, Ashish K. Maurya, Weixin Niah, Audrey Iyu, Stone Elworthy, Raymond Teck Ho Lee, Jin Ben, Philip W. Ingham, Ashley Shu Mei Ng, Zhonghua Zhao, and Weimiao Yu
- Subjects
Cancer Research ,Neural Tube ,animal structures ,Embryo, Nonmammalian ,lcsh:QH426-470 ,Morpholino ,Neural tube patterning ,Kinesins ,Biology ,Zinc Finger Protein Gli2 ,Zinc Finger Protein GLI1 ,Retina ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cerebellar Diseases ,Cerebellum ,Genetics ,Animals ,Humans ,Abnormalities, Multiple ,Cilia ,Eye Abnormalities ,Molecular Biology ,Zebrafish ,Genetics (clinical) ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology ,Zinc finger ,Oncogene Proteins ,0303 health sciences ,Cilium ,Gene Expression Regulation, Developmental ,Extremities ,Kidney Diseases, Cystic ,Zebrafish Proteins ,biology.organism_classification ,Molecular biology ,Hedgehog signaling pathway ,Repressor Proteins ,lcsh:Genetics ,Trans-Activators ,Ectopic expression ,Drosophila melanogaster ,030217 neurology & neurosurgery ,Research Article ,Transcription Factors - 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., Author Summary Hedgehog (Hh) proteins activate one of a handful of signaling pathways that orchestrate the development of animal embryos, controlling cell type specification, proliferation and survival in a variety of contexts. In Drosophila, the Cos2 protein plays a key role in modulating the response of cells to Hh signaling, while mutant forms of its human counterpart KIF7 are associated with a class of developmental defects known as ciliopathies. Studies in mouse have implied that Kif7 functions principally in the primary cilium, an organelle required for Hh signaling in vertebrates but absent from most Drosophila cells, suggesting a divergence in the mechanisms of action between phyla. Here we describe the generation of kif7 mutations in the zebrafish as well as the first analysis of endogenous Kif7 protein distribution in a vertebrate embryo. We find that Kif7 acts principally to restrain Gli1 activity and suggest that it functions to sequester the Gli transcription factors, similar to its Drosophila counterpart Cos2, which we also show can partially substitute for Kif7 function in the zebrafish embryo. Consistent with this model we show that Kif7 protein accumulates both in the primary cilia and in cytoplasmic puncta from which it disperses in response to Hh pathway activation.
- Published
- 2013
29. Blood flow suppresses vascular Notch signalling via dll4 and is required for angiogenesis in response to hypoxic signalling
- Author
-
Martin Gering, Kathryn McMahon, Peter Novodvorsky, Caroline Gray, Oliver Watson, Timothy J. A. Chico, David C. Crossman, Alexander M.K. Rothman, Fredericus J.M. van Eeden, Allan Lawrie, Andrea Haase, University of St Andrews. School of Medicine, and University of St Andrews. Office of the Principal
- Subjects
medicine.medical_specialty ,animal structures ,Notch ,Endothelium ,Physiology ,Angiogenesis ,Notch signaling pathway ,Neovascularization, Physiologic ,Diacetyl ,R Medicine ,03 medical and health sciences ,0302 clinical medicine ,Vascular Biology ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,Hypoxia ,Zebrafish ,030304 developmental biology ,0303 health sciences ,Gene knockdown ,biology ,Receptors, Notch ,Tumor Suppressor Proteins ,Intracellular Signaling Peptides and Proteins ,Membrane Proteins ,Kinase insert domain receptor ,Blood flow ,Original Articles ,Hypoxia (medical) ,Zebrafish Proteins ,biology.organism_classification ,Vascular Endothelial Growth Factor Receptor-2 ,Cell biology ,Angio-/arteriogenesis ,Endocrinology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Blood Circulation ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,Signal Transduction - Abstract
This work was supported by BHF project grants (09/287/28051 and 12/12/ 29433) awarded to T.J.A.C. F.J.M.E. was supported by EC-FP7 HEALTHF4-2010-242048. MRC Centre Grant (G0700091) awarded to Professor PW Ingham provided a clinical training fellowship to OJW. Funding for Open Access was provided by he research was funded by the British Heart Foundation. 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. 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. 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. Publisher PDF
- Published
- 2013
30. Hedgehog signaling via a calcitonin receptor-like receptor can induce arterial differentiation independently of VEGF signaling in zebrafish
- Author
-
Stefan Schulte-Merker, Philip W. Ingham, Roger Patient, Robert N. Wilkinson, Fredericus J.M. van Eeden, Marco J. Koudijs, and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
Vascular Endothelial Growth Factor A ,Biochemistry ,hematopoietic stem-cells ,Animals, Genetically Modified ,chemistry.chemical_compound ,0302 clinical medicine ,endothelial-growth-factor ,Sonic hedgehog ,Experimental Zoology ,Zebrafish ,0303 health sciences ,Receptors, Notch ,Calcitonin Receptor-Like Protein ,Gene Expression Regulation, Developmental ,Hematology ,Arteries ,Hedgehog signaling pathway ,Cell biology ,Vascular endothelial growth factor ,Patched-1 Receptor ,Vascular endothelial growth factor A ,Gene Knockdown Techniques ,Core Binding Factor Alpha 2 Subunit ,Signal transduction ,notch ,Signal Transduction ,Patched Receptors ,medicine.medical_specialty ,kinase ,Immunology ,Notch signaling pathway ,Receptors, Cell Surface ,Biology ,Article ,aortic endothelium ,03 medical and health sciences ,blood ,Internal medicine ,medicine ,Animals ,Hedgehog Proteins ,gene ,Hedgehog ,030304 developmental biology ,pathway ,sonic-hedgehog ,Membrane Proteins ,Cell Biology ,CALCRL ,Zebrafish Proteins ,Hematopoietic Stem Cells ,Endocrinology ,chemistry ,Experimentele Zoologie ,Mutation ,biology.protein ,WIAS ,embryonic vascular development ,030217 neurology & neurosurgery - 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
31. A zebrafish model to study and therapeutically manipulate hypoxia signaling in tumorigenesis
- Author
-
Fredericus J.M. van Eeden, Stone Elworthy, Philip M. Elks, Stephen A. Renshaw, Ellen van Rooijen, Sarah S Walmsley, Emma C Judson, Sarah McKee, Simon S. Cross, and Kirankumar Santhakumar
- Subjects
Cell physiology ,Cancer Research ,medicine.medical_specialty ,endocrine system diseases ,Transgene ,9,10-Dimethyl-1,2-benzanthracene ,Biology ,urologic and male genital diseases ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Liver Neoplasms, Experimental ,Internal medicine ,Intestinal Neoplasms ,medicine ,Animals ,neoplasms ,Transcription factor ,Zebrafish ,030304 developmental biology ,0303 health sciences ,Tumor Suppressor Proteins ,Zebrafish Proteins ,biology.organism_classification ,female genital diseases and pregnancy complications ,Cell Hypoxia ,Disease Models, Animal ,Endocrinology ,HIF1A ,Cell Transformation, Neoplastic ,Oncology ,Hypoxia-inducible factors ,030220 oncology & carcinogenesis ,Cancer research ,Hypoxia-Inducible Factor 1 ,Signal transduction ,Carcinogenesis ,Signal Transduction - 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. Cancer Res; 72(16); 4017–27. ©2012 AACR.
- Published
- 2012
32. Activation of hypoxia-inducible factor-1α (Hif-1α) delays inflammation resolution by reducing neutrophil apoptosis and reverse migration in a zebrafish inflammation model
- Author
-
Constantino Carlos Reyes-Aldasoro, Xingang Wang, Philip W. Ingham, Stephen A. Renshaw, Fredericus J.M. van Eeden, Giles Dixon, Moira K. B. Whyte, Philip M. Elks, and Sarah R. Walmsley
- Subjects
Male ,Programmed cell death ,Hypoxia-Inducible Factor 1 ,animal structures ,Embryo, Nonmammalian ,Neutrophils ,TK ,Immunology ,Molecular Sequence Data ,Danio ,Inflammation ,Apoptosis ,Biochemistry ,Animals, Genetically Modified ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Cell Movement ,medicine ,Animals ,Amino Acid Sequence ,Transcription factor ,Zebrafish ,030304 developmental biology ,0303 health sciences ,biology ,Aryl Hydrocarbon Receptor Nuclear Translocator ,Genetic Variation ,Cell Biology ,Hematology ,Zebrafish Proteins ,biology.organism_classification ,Hypoxia-Inducible Factor 1, alpha Subunit ,Cell biology ,Disease Models, Animal ,Hypoxia-inducible factors ,030220 oncology & carcinogenesis ,Mutagenesis, Site-Directed ,Female ,medicine.symptom ,RC - 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
33. A Zebrafish Model for VHL and Hypoxia Signaling
- Author
-
Kirankumar Santhakumar, Fredericus J.M. van Eeden, Emile E. Voest, Stefan Schulte-Merker, Rachel H. Giles, Ive Logister, and Ellen van Rooijen
- Subjects
Genetics ,Vascular Tumors ,biology ,Renal cell carcinoma ,Cancer research ,medicine ,Disease ,Von hippel lindau ,biology.organism_classification ,medicine.disease ,Zebrafish ,Zinc finger nuclease ,Caenorhabditis elegans - Published
- 2011
- Full Text
- View/download PDF
34. von Hippel-Lindau tumor suppressor mutants faithfully model pathological hypoxia-driven angiogenesis and vascular retinopathies in zebrafish
- Author
-
Jeroen Korving, Stefan Schulte-Merker, Fredericus J.M. van Eeden, Ive Logister, Rachel H. Giles, Jeroen Bussmann, Emile E. Voest, Ellen van Rooijen, and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
Tumor suppressor gene ,endocrine system diseases ,Angiogenesis ,Neuroscience (miscellaneous) ,Medicine (miscellaneous) ,Biology ,Retinal Neovascularization ,urologic and male genital diseases ,General Biochemistry, Genetics and Molecular Biology ,Neovascularization ,chemistry.chemical_compound ,Immunology and Microbiology (miscellaneous) ,Von Hippel–Lindau tumor suppressor ,medicine ,Animals ,Edema ,Humans ,Macula Lutea ,Hypoxia ,Molecular Biology ,Zebrafish ,neoplasms ,Tumor Suppressor Proteins ,Retinal Detachment ,Anatomy ,Zebrafish Proteins ,biology.organism_classification ,female genital diseases and pregnancy complications ,Vascular endothelial growth factor ,Vascular endothelial growth factor B ,Vascular endothelial growth factor A ,Disease Models, Animal ,Receptors, Vascular Endothelial Growth Factor ,chemistry ,Immunology ,Mutation ,Cancer research ,biology.protein ,Mutant Proteins ,medicine.symptom ,Developmental Biology ,Signal Transduction - Abstract
SUMMARYBiallelic 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
35. 09-P016 Repression of Hedgehog signalling is required for the acquisition of dorsolateral cell fates in the zebrafish otic vesicle
- Author
-
Fredericus J.M. van Eeden, Katherine L. Hammond, and Tanya T. Whitfield
- Subjects
Embryology ,Cell ,Dorsolateral ,Anatomy ,Biology ,biology.organism_classification ,Cell biology ,Hedgehog signalling ,medicine.anatomical_structure ,medicine ,Otic vesicle ,Psychological repression ,Zebrafish ,Developmental Biology - Published
- 2009
- Full Text
- View/download PDF
36. Zebrafish mutants in the von Hippel-Lindau tumor suppressor display a hypoxic response and recapitulate key aspects of Chuvash polycythemia
- Author
-
Fredericus J.M. van Eeden, Jeroen Korving, Rachel H. Giles, Stefan Schulte-Merker, Thorsten Schwerte, Ellen van Rooijen, Ive Logister, Emile E. Voest, University of Groningen, and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
endocrine system diseases ,PROTEIN ,urologic and male genital diseases ,Biochemistry ,Gene Knockout Techniques ,Von Hippel–Lindau tumor suppressor ,Basic Helix-Loop-Helix Transcription Factors ,Hypoxia ,Zebrafish ,Conserved Sequence ,IN-VIVO ,Genetics ,biology ,DANIO-RERIO ,Hematology ,Phenotype ,female genital diseases and pregnancy complications ,Cell biology ,Von Hippel-Lindau Tumor Suppressor Protein ,Stem cell ,EXPRESSION ,Tumor suppressor gene ,Recombinant Fusion Proteins ,Immunology ,Molecular Sequence Data ,Polycythemia ,HEMATOPOIESIS ,Synteny ,Germline mutation ,VHL ,Animals ,Humans ,Point Mutation ,Cell Lineage ,Amino Acid Sequence ,RNA, Messenger ,neoplasms ,Germ-Line Mutation ,EXCESSIVE ERYTHROCYTOSIS ,Sequence Homology, Amino Acid ,Tumor Suppressor Proteins ,Hematopoietic Tissue ,MICE OVEREXPRESSING ERYTHROPOIETIN ,Cell Biology ,Zebrafish Proteins ,biology.organism_classification ,GENE ,Erythropoietin receptor ,Disease Models, Animal ,ENDOTHELIAL GROWTH-FACTOR ,biology.protein ,Sequence Alignment - 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
37. Genetic analysis of the two zebrafish patched homologues identifies novel roles for the hedgehog signaling pathway
- Author
-
Fredericus J.M. van Eeden, Marjo J. den Broeder, Evelyn Groot, Marco J. Koudijs, and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
Patched Receptors ,Patched ,Embryo, Nonmammalian ,DNA Mutational Analysis ,Molecular Sequence Data ,Receptors, Cell Surface ,Tretinoin ,Biology ,03 medical and health sciences ,0302 clinical medicine ,GLI1 ,GLI3 ,Animals ,Hedgehog Proteins ,Eye Abnormalities ,Hedgehog ,Zebrafish ,lcsh:QH301-705.5 ,Body Patterning ,030304 developmental biology ,Genetics ,0303 health sciences ,Base Sequence ,Sequence Homology, Amino Acid ,Veratrum Alkaloids ,Animal Structures ,Gene Expression Regulation, Developmental ,Membrane Proteins ,Zebrafish Proteins ,biology.organism_classification ,Hedgehog signaling pathway ,Patched-1 Receptor ,Phenotype ,Somites ,lcsh:Biology (General) ,Mutation ,biology.protein ,Mutant Proteins ,RNA Splice Sites ,030217 neurology & neurosurgery ,Research Article ,Signal Transduction ,Genetic screen ,Developmental Biology - Abstract
BackgroundAberrant 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.ResultsHere we describe a splice-donor mutation in Ptc1, calledptc1hu1602, which in a homozygous state results in a subtle eye and somite phenotype. Since we recently positionally cloned aptc2mutant, aptc1;ptc2double mutant was generated, showing severely increased levels ofptc1,gli1andnkx2.2a, confirming an aberrant activation of Hh signaling. As a consequence, a number of phenotypes were observed that have not been reported previously usingShhmRNA overexpression. Somites ofptc1;ptc2double 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.ConclusionThe generation and characterization of theptc1;ptc2double 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
38. Hedgehog signaling plays a cell-autonomous role in maximizing cardiac developmental potential
- Author
-
Deborah Yelon, Fredericus J.M. van Eeden, Marco Koudijs, Alexandra L. Joyner, Natalie A. Thomas, and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
Cellular differentiation ,Article ,Mice ,Fate mapping ,Cell autonomous ,Animals ,Hedgehog Proteins ,Progenitor cell ,Molecular Biology ,Hedgehog ,Zebrafish ,Cell Proliferation ,biology ,Myocardium ,Stem Cells ,Endothelial Cells ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Heart ,Cell Biology ,biology.organism_classification ,Hedgehog signaling pathway ,Cell biology ,Transplantation ,Immunology ,Signal transduction ,Signal Transduction ,Developmental Biology - 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
39. The zebrafish mutants dre, uki, and lep encode negative regulators of the hedgehog signaling pathway
- Author
-
Ellen van Rooijen, Erno Wienholds, Astrid Keijser, Robert Geisler, Fredericus J.M. van Eeden, Saskia Houwing, Marjo J. den Broeder, and Marco J. Koudijs
- Subjects
Cancer Research ,Embryo, Nonmammalian ,lcsh:QH426-470 ,Cyclopamine ,Eukaryotes ,Teleost Fishes ,Mutant ,Dwarfism ,Biology ,Development ,medicine.disease_cause ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Proliferating Cell Nuclear Antigen ,Genetics ,medicine ,Animals ,Body Size ,Hedgehog Proteins ,Molecular Biology ,Hedgehog ,Zebrafish ,Genetics (clinical) ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology ,Cell Proliferation ,Cancer Biology ,Danio (zebrafish) ,0303 health sciences ,Mutation ,biology.organism_classification ,Hedgehog signaling pathway ,Cell biology ,Genetics/Disease Models ,lcsh:Genetics ,chemistry ,Genetics/Gene Function ,Vertebrates ,Trans-Activators ,Hedgehog interacting protein ,030217 neurology & neurosurgery ,Genetic screen ,Signal Transduction ,Research Article - 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., Synopsis In a screen aimed at finding genes that control proliferation in the zebrafish embryo, three mutants were identified. Mutants showed an increase in size of several structures including the brain, the retina, and the fins. Surprisingly, although size was increased in the embryos, adults remained small. Cloning of these genes revealed that increased Hedgehog signaling was at the basis of the phenotype, because all three genes encoded known repressors of the Hedgehog signaling pathway: Ptc2, Su(Fu), and Hip. Hedgehog is known to play a role in proliferation. For instance, ectopic Hedgehog signaling can lead to several tumors including basal cell carcinoma and medulloblastoma. However, the phenotypes were still a surprise, because earlier experiments in zebrafish embryos suggested that activation should lead to patterning rather than proliferation defects. Current models of the pathway predict that these genes act independently to inhibit the signal but curiously, redundancy amongst these genes was not found, because triple mutants looked like the single mutants. The conclusion is that weak activation of Hedgehog signaling can already lead to stimulation of growth in the absence of patterning defects, and that the Hedgehog signal is probably kept in check by the last inhibitor: Ptc1. A mutant for the ptc1 gene has recently been created and will put the model to the test.
- Published
- 2005
40. The Wnt/beta-catenin pathway regulates cardiac valve formation
- Author
-
Erno Wienholds, Adam Hurlstone, Harry Begthel, Hans Clevers, Edwin Cuppen, Jeroen Korving, Ronald H.A. Plasterk, Danica Zivkovic, Fredericus J.M. van Eeden, Anna-Pavlina G. Haramis, and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
medicine.medical_specialty ,Beta-catenin ,Genes, APC ,Genotype ,Adenomatous polyposis coli ,Adenomatous Polyposis Coli Protein ,Endocardial cushion formation ,Internal medicine ,Proto-Oncogene Proteins ,medicine ,Animals ,Humans ,Cell Lineage ,Zebrafish ,beta Catenin ,Multidisciplinary ,biology ,Wnt signaling pathway ,Gene Expression Regulation, Developmental ,Proteins ,Zebrafish Proteins ,biology.organism_classification ,Heart Valves ,Hedgehog signaling pathway ,Cell biology ,Wnt Proteins ,Cytoskeletal Proteins ,Endocrinology ,DKK1 ,Catenin ,embryonic structures ,Mutation ,biology.protein ,Trans-Activators ,Intercellular Signaling Peptides and Proteins ,Cell Division ,Signal Transduction - Abstract
Truncation of the tumour suppressor adenomatous polyposis coli (Apc) constitutively activates the Wnt/beta-catenin signalling pathway. Apc has a role in development: for example, embryos of mice with truncated Apc do not complete gastrulation. To understand this role more fully, we examined the effect of truncated Apc on zebrafish development. Here we show that, in contrast to mice, zebrafish do complete gastrulation. However, mutant hearts fail to loop and form excessive endocardial cushions. Conversely, overexpression of Apc or Dickkopf 1 (Dkk1), a secreted Wnt inhibitor, blocks cushion formation. In wild-type hearts, nuclear beta-catenin, the hallmark of activated canonical Wnt signalling, accumulates only in valve-forming cells, where it can activate a Tcf reporter. In mutant hearts, all cells display nuclear beta-catenin and Tcf reporter activity, while valve markers are markedly upregulated. Concomitantly, proliferation and epithelial-mesenchymal transition, normally restricted to endocardial cushions, occur throughout the endocardium. Our findings identify a novel role for Wnt/beta-catenin signalling in determining endocardial cell fate.
- Published
- 2003
41. Two distinct cell populations in the floor plate of the zebrafish are induced by different pathways
- Author
-
Philip W. Ingham, Pascal Haffter, Christiane Nüsslein-Volhard, Jörg Odenthal, and Fredericus J.M. van Eeden
- Subjects
animal structures ,Mutant ,floor plate ,Notochord ,sonic hedgehog ,Mice ,Species Specificity ,medicine ,Paraxial mesoderm ,Animals ,Hedgehog Proteins ,Sonic hedgehog ,Molecular Biology ,Zebrafish ,In Situ Hybridization ,fkd4 ,Floor plate ,Body Patterning ,Genetics ,Motor Neurons ,biology ,Neural tube ,Gene Expression Regulation, Developmental ,Proteins ,Cell Biology ,biology.organism_classification ,Cell biology ,Somite ,medicine.anatomical_structure ,Spinal Cord ,embryonic structures ,Mutation ,biology.protein ,Trans-Activators ,Developmental Biology - 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.
- Published
- 2000
42. The polarisation of the anterior-posterior and dorsal-ventral axes during Drosophila oogenesis
- Author
-
Daniel St Johnston and Fredericus J.M. van Eeden
- Subjects
Polarity (international relations) ,fungi ,Drosophila embryogenesis ,Cell Polarity ,Anatomy ,Biology ,oskar ,Oocyte ,Embryonic stem cell ,Oogenesis ,Hedgehog signaling pathway ,Cell biology ,medicine.anatomical_structure ,Cell polarity ,Mutation ,Genetics ,medicine ,Oocytes ,Animals ,Drosophila ,Developmental Biology - Abstract
Recent work on Drosophila oogenesis has begun to reveal how the first asymmetries in development arise and how these relate to the later events that localise the positional cues which define the embryonic axes. The Cadherin-dependent positioning of the oocyte creates an anterior-posterior polarity that is transmitted to the embryo through the localisation and localised translation of bicoid, oskar, and nanos mRNA. In contrast, dorsal-ventral polarity arises from the random migration of the nucleus to the anterior of the oocyte, where it determines where gurken mRNA is translated and localised. Gurken signalling then defines the embryonic dorsal-ventral axis by restricting pipe expression to the ventral follicle cells, where Pipe regulates the production of an unidentified cue that activates the Toll signalling pathway.
- Published
- 1999
43. Mutations affecting pigmentation and shape of the adult zebrafish
- Author
-
Robert N. Kelsh, Michael Brand, M. Hammerschmidt, Michael Granato, Fredericus J.M. van Eeden, J. Odenthal, Shuo Lin, Mary C. Mullins, Carl-Philipp Heisenberg, Makoto Furutani-Seiki, D. A. Kane, Y.-J. Jiang, Christiane Nüsslein-Volhard, Elisabeth Vogelsang, P. Haffter, Fabian B. Haas, Nancy Hopkins, and Michael Farrell
- Subjects
Genetics ,biology ,Genetic marker ,Genetic model ,Drosophila melanogaster ,biology.organism_classification ,Developmental biology ,Phenotype ,Gene ,Zebrafish ,Caenorhabditis elegans ,Developmental Biology - Abstract
Mutations causing a visible phenotype in the adult serve as valuable visible genetic markers in multicellular genetic model organisms such as Drosophila melanogaster, Caenorhabditis elegans and Arabidopsis thaliana. In a large scale screen for mutations affecting early development of the zebrafish, we identified a number of mutations that are homozygous viable or semiviable. Here we describe viable mutations which produce visible phenotypes in the adult fish. These predominantly affect the fins and pigmentation, but also the eyes and body length of the adult. A number of dominant mutations caused visible phenotypes in the adult fish. Mutations in three genes, long fin, another long fin and wanda affected fin formation in the adult. Four mutations were found to cause a dominant reduction of the overall body length in the adult. The adult pigment pattern was found to be changed by dominant mutations in wanda, asterix, obelix, leopard, salz and pfeffer. Among the recessive mutations producing visible phenotypes in the homozygous adult, a group of mutations that failed to produce melanin was assayed for tyrosinase activity. Mutations in sandy produced embryos that failed to express tyrosinase activity. These are potentially useful for using tyrosinase as a marker for the generation of transgenic lines of zebrafish.
44. Genome-wide mapping of Hif-1α binding sites in zebrafish
- Author
-
Fredericus J.M. van Eeden, Sinnakaruppan Mathavan, Bernd Pelster, David Greenald, Justin Jeyakani, Ian M Sealy, and Lee Kong Chian School of Medicine (LKCMedicine)
- Subjects
Chromatin Immunoprecipitation ,Mutant ,Microarray ,Real-Time Polymerase Chain Reaction ,Response Elements ,Transcriptome ,Von Hippel-Lindau ,Genetics ,Animals ,Humans ,Nucleotide Motifs ,Hypoxia ,Zebrafish ,Transcription factor ,Gene ,Regulation of gene expression ,Binding Sites ,Genome ,biology ,Hypoxia Response Element ,Tumor Suppressor Proteins ,Computational Biology ,High-Throughput Nucleotide Sequencing ,Reproducibility of Results ,Zebrafish Proteins ,biology.organism_classification ,Hypoxia-Inducible Factor 1, alpha Subunit ,ChIP-seq ,Hypoxia-inducible factors ,Gene Expression Regulation ,Mutation ,Hif-1α ,von Hippel-Lindau ,Chromatin immunoprecipitation ,Research Article ,Genome-Wide Association Study ,Protein Binding ,Biotechnology - 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. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-2169-x) contains supplementary material, which is available to authorized users.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.