28 research outputs found

    The number of MPCs is equal.

    No full text
    <p><b>A</b>: Schematic illustration of a zebrafish embryo at 24hpf (lateral view) and a detail of the region above the end of the yolk extension imaged for the analysis of the MPCs (lateral view). The four somites, which were considered for the quantification of MPCs are marked with red lines. <b>B</b>: Immunofluorescence staining with Pax7 at 24hpf in <i>grna</i><sup><i>−/−</i></sup>;<i>grnb</i><sup><i>−/−</i></sup> mutants and wildtype embryos. In each image two xanthophores are exemplarily marked with a red x. Images taken by spinning disk confocal microscopy. Anterior to the left. Lateral view. Orthogonal projections. Scale bar: 50μm. <b>C</b>: Quantification of Pax7-positive cells in the 4 somites (1–4) above the end of the yolk extension in <i>grna</i><sup><i>−/−</i></sup>;<i>grnb</i><sup><i>−/−</i></sup> mutants and wildtype embryos. Only Pax7-positive cells at the surface of the somites were counted. n = 15. S.E.M. Mann-Whitney test (two-tailed). All n.s. S1: p = 0.0854. S2: p = 0.7977. S3: p = 0.3489. S4: p = 0.2337.</p

    No SpMN axonopathy in Grna and Grnb single and double KOs.

    No full text
    <p><b>A</b>: Schematic illustration of a zebrafish embryo at 28hpf (lateral view) and a detail of the region above the end of the yolk extension imaged for the analysis of SpMN axons (lateral view). <b>B:</b> In Grna and Grnb single and double KOs the SpMN axons show no extended branching. Whole-mount immunofluorescence staining of 28hpf embryos with znp1 antibody. The 5 SpMN axons above the end of the yolk extension are shown. Images taken by spinning disk confocal microscopy. Anterior to the left. Lateral view. Orthogonal projections. Scale bar: 100μm. <b>C-E</b>: Quantification of the SpMN axon length in homozygous and heterozygous Grna and Grnb single and double KOs and wildtype siblings. The SpMN axon length of the 5 SpMN axons (1–5) above the end of the yolk extension is measured from the exit point of the spinal cord to the tip of the growth cone. <b>C</b>: Homozygous and heterozygous Grna KOs and wildtype siblings. n = 30. <b>D</b>: Homozygous and heterozygous Grnb KOs and wildtype siblings. n = 30. <b>E</b>: Homozygous and heterozygous Grna and Grnb KOs and wildtype siblings. n = 25. S.E.M. Two-way ANOVA. Bonferroni post-test. All non-significant (n.s.).</p

    No microgliosis and neurodegeneration in <i>grna</i><sup><i>−/−</i></sup>;<i>grnb</i><sup><i>−/−</i></sup> mutants.

    No full text
    <p><b>A</b>: Schematic illustration of a zebrafish larvae at 3dpf (lateral view) and a detail of the region (red line), dorsal view, imaged for the analysis of neutral red positive particles (B). The dashed red line marks the area that was imaged in the time lapse recordings of microglia (C). <b>B</b>: The number of neutral red positive particle in the region illustrated in A (Z-stack) is unchanged in wildtype and <i>grna</i><sup><i>−/−</i></sup>;<i>grnb</i><sup><i>−/−</i></sup> mutants. n = 15. S.E.M. Mann-Whitney test (two-tailed). p = 0.2884. <b>C-E</b>: Microglia in Tg(<i>apoeb</i>:lynEGFP) <i>grna</i><sup><i>−/−</i></sup>;<i>grnb</i><sup><i>−/−</i></sup> mutants and wildtype larvae at 3dpf are indistinguishable. <b>C:</b> Still images of the time lapse recordings in the optic tectum recorded by spinning disk confocal microscopy. Two microglia cells marked in each genotype by a white and yellow arrow. Dorsal view. Anterior to the left. n = 3. Scale bar: 50μm. Recording time: 60min. 1frame/min. <b>D:</b> The distance microglia move within one hour in <i>grna</i><sup><i>−/−</i></sup>;<i>grnb</i><sup><i>−/−</i></sup> mutants and wildtype larvae. Quantification of n = 3x5 randomly selected microglia from the time lapse recordings shown in C. S.E.M. Mann-Whitney test (two-tailed). p = 0.0671. <b>E:</b> Processes in the <i>grna</i><sup><i>−/−</i></sup>;<i>grnb</i><sup><i>−/−</i></sup> mutants and wildtype larvae persist for same durations. Quantification of n = 3x5 randomly selected processes from the time lapse recordings shown in C. S.E.M. Mann-Whitney test (two-tailed). p = 0.8296. <b>F:</b> Schematic illustration of a zebrafish larvae at 5dpf (lateral view) and a detail of the region, dorsal view, imaged for the analysis of acridine orange (AO) positive cells. <b>G</b>: The number of acridine orange positive cells in the region illustrated in C (Z-stack) is unchanged in wildtype and <i>grna</i><sup><i>−/−</i></sup>;<i>grnb</i><sup><i>−/−</i></sup> mutants. n = 15. S.E.M. Mann-Whitney test (two-tailed). p = 0.69.</p

    <i>grna</i><sup><i>−/−</i></sup>;<i>grnb</i><sup><i>−/−</i></sup> mutants swim like wildtype.

    No full text
    <p><b>A</b>: The swim path of wildtype, <i>grna</i><sup><i>−/−</i></sup>;<i>grnb</i><sup><i>−/−</i></sup> mutants, DMSO-treated, and PTZ-treated larvae is shown. PTZ treatment was used as a positive control. 5dpf. Movements < 2mm/s: black lines. Movements 2–6mm/s: green lines. Movements > 6mm/s: red lines. Recording time: 5min. <b>B</b>: The total distance moved within 5min in wildtype, <i>grna</i><sup><i>−/−</i></sup>;<i>grnb</i><sup><i>−/−</i></sup> mutants, DMSO-treated, and PTZ-treated larvae is shown. Wt-<i>grna</i><sup><i>−/−</i></sup>;<i>grnb</i><sup><i>−/−</i></sup>: p = 0.2386. Wt-DMSO: p = 0.0534. Wt-PTZ: ***p = 0.0002. DMSO-PTZ: **p = 0.004. <b>C</b>: A graph of the mean velocity of wildtype, <i>grna</i><sup><i>−/−</i></sup>;<i>grnb</i><sup><i>−/−</i></sup> mutants, DMSO-treated, and PTZ-treated larvae is shown. Time frame: 5min. Wt-<i>grna</i><sup><i>−/−</i></sup>;<i>grnb</i><sup><i>−/−</i></sup>: p = 0.5657. Wt-DMSO: p = 0.8081. Wt-PTZ: *p = 0.0137. DMSO-PTZ: **p = 0.0014. <b>D</b>: Percentage of time spent for movements with a velocity above 2mm/s in wildtype, <i>grna</i><sup><i>−/−</i></sup>;<i>grnb</i><sup><i>−/−</i></sup> mutants, DMSO-treated, and PTZ-treated larvae is plotted. Time frame: 5min. Wt-<i>grna</i><sup><i>−/−</i></sup>;<i>grnb</i><sup><i>−/−</i></sup>: p = 0.2585. Wt-DMSO: p = 0.0668. Wt-PTZ: **p = 0.0016. DMSO-PTZ: **p = 0.0037. <b>B-D:</b> n = 18. S.E.M. Mann-Whitney test (two-tailed).</p

    Generation of Grna and Grnb KOs using ZFNs.

    No full text
    <p><b>A</b>: Schematic illustration of human GRN and zebrafish Granulins. Human GRN has 7 ½ granulin domains, while 12 granulin domains are found in Grna, 9 in Grnb, and 1 ½ in Grn1 and Grn2. Grey: signal peptide. Black numbers: amino acids. Darker colour and white letters/numbers: granulin domains. <b>B-C:</b> Localisation of ZFN target sequences in <i>grna</i> and <i>grnb</i> and predicted protein sequence of selected alleles. The genomic structure of <i>grna</i> and <i>grnb</i> is depicted. ZFNs targeting <i>grna</i> and <i>grnb</i> are located in the first and fourth coding exon, respectively. ZFN-induced genomic lesions in <i>grna</i> can be detected with the restriction enzyme (RE) Eco91I and in <i>grnb</i> with the RE XcmI. Grey boxes: untranslated region (UTR). Coloured boxes: coding region. Light blue: ZFN binding sites in <i>grna</i>. Light red: ZFN binding sites in <i>grnb</i>. Green lines: binding sites of the RE. Dashed green line: cut site of the RE. Protein sequences of wildtype (wt) <i>grna</i> and 4 <i>grna</i> mutation alleles as well as wt <i>grnb</i> and 3 <i>grnb</i> mutation alleles are shown. *: Stop. <b>D-E</b>: Grna and Grnb protein is lost in all mutants. <b>D</b>: Grna signal is lost in all adult kidney samples from grna<sup>−/−</sup> mutants, whereas a signal is present in wt. A Calnexin blot serves as a loading control. <b>E</b>: The Grnb signal observed in wt is lost in all 1.5dpf samples from <i>grnb</i><sup>−/−</sup> mutants. Injection of <i>grnb</i> mRNA leads to an increase in signal. The loading control α-tubulin is present in all samples.</p

    Masculinity and the Trials of Modern Fiction

    No full text
    This monograph addresses a number of literary trials in nineteenth-century England and France. It examines the ways in which notions of gender were contested and constructed in the literary and legal discourses of the period. It also posits a new model for understanding the complex relationship between law and literature

    Additional file 6: Figure S6. of Glycine-alanine dipeptide repeat protein contributes to toxicity in a zebrafish model of C9orf72 associated neurodegeneration

    No full text
    Tardbp function is not impaired in repeat expressing fish. (A) GA80-GFPa zebrafish expressing GFP and (B) siblings not expressing GFP. Western blot analysis of 2 dpf old embryos with antibodies as indicated. Tardbp/Tardbpl_tv1 bands indicated by arrow heads. (PDF 3086 kb

    Image4_Loss of TDP-43 causes ectopic endothelial sprouting and migration defects through increased fibronectin, vcam 1 and integrin α4/β1.TIFF

    No full text
    Aggregation of the Tar DNA-binding protein of 43 kDa (TDP-43) is a pathological hallmark of amyotrophic lateral sclerosis and frontotemporal dementia and likely contributes to disease by loss of nuclear function. Analysis of TDP-43 function in knockout zebrafish identified an endothelial directional migration and hypersprouting phenotype during development prior lethality. In human umbilical vein cells (HUVEC) the loss of TDP-43 leads to hyperbranching. We identified elevated expression of FIBRONECTIN 1 (FN1), the VASCULAR CELL ADHESION MOLECULE 1 (VCAM1), as well as their receptor INTEGRIN α4β1 (ITGA4B1) in HUVEC cells. Importantly, reducing the levels of ITGA4, FN1, and VCAM1 homologues in the TDP-43 loss-of-function zebrafish rescues the angiogenic defects indicating the conservation of human and zebrafish TDP-43 function during angiogenesis. Our study identifies a novel pathway regulated by TDP-43 important for angiogenesis during development.</p

    Table1_Loss of TDP-43 causes ectopic endothelial sprouting and migration defects through increased fibronectin, vcam 1 and integrin α4/β1.XLSX

    No full text
    Aggregation of the Tar DNA-binding protein of 43 kDa (TDP-43) is a pathological hallmark of amyotrophic lateral sclerosis and frontotemporal dementia and likely contributes to disease by loss of nuclear function. Analysis of TDP-43 function in knockout zebrafish identified an endothelial directional migration and hypersprouting phenotype during development prior lethality. In human umbilical vein cells (HUVEC) the loss of TDP-43 leads to hyperbranching. We identified elevated expression of FIBRONECTIN 1 (FN1), the VASCULAR CELL ADHESION MOLECULE 1 (VCAM1), as well as their receptor INTEGRIN α4β1 (ITGA4B1) in HUVEC cells. Importantly, reducing the levels of ITGA4, FN1, and VCAM1 homologues in the TDP-43 loss-of-function zebrafish rescues the angiogenic defects indicating the conservation of human and zebrafish TDP-43 function during angiogenesis. Our study identifies a novel pathway regulated by TDP-43 important for angiogenesis during development.</p

    Image6_Loss of TDP-43 causes ectopic endothelial sprouting and migration defects through increased fibronectin, vcam 1 and integrin α4/β1.TIFF

    No full text
    Aggregation of the Tar DNA-binding protein of 43 kDa (TDP-43) is a pathological hallmark of amyotrophic lateral sclerosis and frontotemporal dementia and likely contributes to disease by loss of nuclear function. Analysis of TDP-43 function in knockout zebrafish identified an endothelial directional migration and hypersprouting phenotype during development prior lethality. In human umbilical vein cells (HUVEC) the loss of TDP-43 leads to hyperbranching. We identified elevated expression of FIBRONECTIN 1 (FN1), the VASCULAR CELL ADHESION MOLECULE 1 (VCAM1), as well as their receptor INTEGRIN α4β1 (ITGA4B1) in HUVEC cells. Importantly, reducing the levels of ITGA4, FN1, and VCAM1 homologues in the TDP-43 loss-of-function zebrafish rescues the angiogenic defects indicating the conservation of human and zebrafish TDP-43 function during angiogenesis. Our study identifies a novel pathway regulated by TDP-43 important for angiogenesis during development.</p
    corecore