12 research outputs found

    Glycogen synthase kinase 3α and 3β have distinct functions during cardiogenesis of zebrafish embryo-4

    No full text
    <p><b>Copyright information:</b></p><p>Taken from "Glycogen synthase kinase 3α and 3β have distinct functions during cardiogenesis of zebrafish embryo"</p><p>http://www.biomedcentral.com/1471-213X/7/93</p><p>BMC Developmental Biology 2007;7():93-93.</p><p>Published online 3 Aug 2007</p><p>PMCID:PMC1988812.</p><p></p>ybridization with staining revealed that randomized looping was observed in morphants (B-D). The expression of (F) and (H) appeared normal in morphants

    Glycogen synthase kinase 3α and 3β have distinct functions during cardiogenesis of zebrafish embryo-1

    No full text
    <p><b>Copyright information:</b></p><p>Taken from "Glycogen synthase kinase 3α and 3β have distinct functions during cardiogenesis of zebrafish embryo"</p><p>http://www.biomedcentral.com/1471-213X/7/93</p><p>BMC Developmental Biology 2007;7():93-93.</p><p>Published online 3 Aug 2007</p><p>PMCID:PMC1988812.</p><p></p> to severe (B) defects in axis formation. At 72 hpf, both and morphants displayed pericardial edema (F, G, I, J) and an unlooped, stretched heart (I, J)

    Glycogen synthase kinase 3α and 3β have distinct functions during cardiogenesis of zebrafish embryo-8

    No full text
    <p><b>Copyright information:</b></p><p>Taken from "Glycogen synthase kinase 3α and 3β have distinct functions during cardiogenesis of zebrafish embryo"</p><p>http://www.biomedcentral.com/1471-213X/7/93</p><p>BMC Developmental Biology 2007;7():93-93.</p><p>Published online 3 Aug 2007</p><p>PMCID:PMC1988812.</p><p></p> to severe (B) defects in axis formation. At 72 hpf, both and morphants displayed pericardial edema (F, G, I, J) and an unlooped, stretched heart (I, J)

    Glycogen synthase kinase 3α and 3β have distinct functions during cardiogenesis of zebrafish embryo-2

    No full text
    <p><b>Copyright information:</b></p><p>Taken from "Glycogen synthase kinase 3α and 3β have distinct functions during cardiogenesis of zebrafish embryo"</p><p>http://www.biomedcentral.com/1471-213X/7/93</p><p>BMC Developmental Biology 2007;7():93-93.</p><p>Published online 3 Aug 2007</p><p>PMCID:PMC1988812.</p><p></p> of either -(-MO) or -mRNA (-MO), was injected into one-celled stage embryos and the heart morphology was observed at the stage as indicated. The elongation of heart tube was normally developed at 24 hpf in the wild-type (A) and in the morphants (C); whereas the heart of morphant did not elongate to from a heart-tube (B). The wild-type (D) and morphant's heart (F) developed normally at 30 hpf, but the heart of morphant was still retardant development at 30 hpf (E), and even ceased at heart-cone stage at 36 hpf (F). Compared to the wild-type (G), however, the heart positioning was abnormally in the morphant at 36 hpf (I, J). Eventually, both and morphants displayed an unlooped and stretched heart (L, M). The heart morphology of embryos injected with the control MO was also observed at 72 hpf (N). a: atrium; v: ventricle

    Glycogen synthase kinase 3α and 3β have distinct functions during cardiogenesis of zebrafish embryo-5

    No full text
    <p><b>Copyright information:</b></p><p>Taken from "Glycogen synthase kinase 3α and 3β have distinct functions during cardiogenesis of zebrafish embryo"</p><p>http://www.biomedcentral.com/1471-213X/7/93</p><p>BMC Developmental Biology 2007;7():93-93.</p><p>Published online 3 Aug 2007</p><p>PMCID:PMC1988812.</p><p></p>d the left-predominant asymmetry persists through the stages of jogging (G). However, in morphants, the expression of becomes symmetrical at 20 hpf (B, D). In morphants, in which the heart fails to jog, is more evenly distributed in the heart region (H, I). The left-sided domain was greatly reduced in morphant hearts at 16 hpf (F). All are dorsal views. B, E are higher magnifications of A, D, respectively. Lines mark the midline. L, embryo left

    Glycogen synthase kinase 3α and 3β have distinct functions during cardiogenesis of zebrafish embryo-6

    No full text
    <p><b>Copyright information:</b></p><p>Taken from "Glycogen synthase kinase 3α and 3β have distinct functions during cardiogenesis of zebrafish embryo"</p><p>http://www.biomedcentral.com/1471-213X/7/93</p><p>BMC Developmental Biology 2007;7():93-93.</p><p>Published online 3 Aug 2007</p><p>PMCID:PMC1988812.</p><p></p>phants. At 60–72 hpf,(A, B) and (C, D) expression was greatly up-regulated in morphants. Tg() embryos were injected with -MO and observed by two-photon fluorescence imaging of a live transgenic zebrafish heart at 100 hpf. The endocardial cells and blood are labeled yellow; the Hc-GFP-positive myocardial cells are labeled red. Valves are clearly observed in wild-type embryos (E; white arrows), but not in morphants (F). b, blood cells; V, ventricle; A, atrium

    Glycogen synthase kinase 3α and 3β have distinct functions during cardiogenesis of zebrafish embryo-7

    No full text
    <p><b>Copyright information:</b></p><p>Taken from "Glycogen synthase kinase 3α and 3β have distinct functions during cardiogenesis of zebrafish embryo"</p><p>http://www.biomedcentral.com/1471-213X/7/93</p><p>BMC Developmental Biology 2007;7():93-93.</p><p>Published online 3 Aug 2007</p><p>PMCID:PMC1988812.</p><p></p>analyzed by western blot. The antibody used is indicated in the left of each blot. Anti-GSK3 antibody enables to recognize both GSK3α and GSK3β proteins; anti-α-tubulin antibody was used as a loading control. The protein levels of GSK3α and GSK3β were reduced greatly in the protein lysates extracted from the – and -MO-injected embryos, respectively

    One crisis, diverse impacts—Tissue-specificity of folate deficiency-induced circulation defects in zebrafish larvae

    No full text
    <div><p>Folate (vitamin B9) is an essential nutrient required for cell survival, proliferation, differentiation and therefore embryogenesis. Folate deficiency has been associated with many diseases, including congenital heart diseases and megaloblastic anemia, yet the mechanisms underlying these remains elusive. Here, we examine the impact of folate deficiency on the development of the circulation system using a zebrafish transgenic line which displays inducible folate deficiency. Impaired hematopoiesis includes decreased hemoglobin levels, decreased erythrocyte number, increased erythrocyte size and aberrant <i>c-myb</i> expression pattern were observed in folate deficient embryos. Cardiac defects, including smaller chamber size, aberrant cardiac function and <i>cmlc2</i> expression pattern, were also apparent in folate deficient embryos. Characterization of intracellular folate content in folate deficiency revealed a differential fluctuation among the different folate derivatives that carry a single carbon group at different oxidation levels. Rescue attempts by folic acid and nucleotides resulted in differential responses among affected tissues, suggesting that different pathomechanisms are involved in folate deficiency-induced anomalies in a tissue-specific manner. The results of the current study provide an explanation for the inconsistent outcome observed clinically in patients suffering from folate deficiency and/or receiving folate supplementation. This study also supports the use of this model for further research on the defective cardiogenesis and hematopoiesis caused by folate deficiency.</p></div

    Zebrafish larval hematopoiesis and response to rescuing agents.

    No full text
    <p>(A, B) Hemoglobin of larvae in control and FD groups, with/without folate supplementation, were stained with o-dianisidine at 3 dpf. Hemoglobin signals were distributed most abundantly in the heart (dashed rectangles) and common cardinal veins (arrowheads) of control larvae (normal). Ectopic accumulation of hemoglobin in caudal veins (arrows) was often observed in FD larvae (mild and severe). The severity of anomalies was categorized and quantified based on the level and distribution of hemoglobin signals in larval heart and common cardinal veins. The images shown were the lateral (the upper panel) and ventral (the lower panel) views of larvae. Average of at least six independent experiments with the total sample number of 51–139 for each group are reported. (C, D) The relative number and size of embryonic erythrocytes were analyzed with flow cytometry for both control and FD embryos of 2-dpf generated by crossing Tg (hsp:EGFP-γGH) and Tg (gata1:dsRed). The numbers of erythrocytes were presented as the percentage of red fluorescent cells to total cell number. The size of erythrocytes was normalized with those of control larvae. Presented are data collected from at least three independent experiments with a total embryo number of approximately 30–40 for each group. (E) Hematopoiesis in both control and FD embryos was characterized by whole mount <i>in situ</i> hybridization with a riboprobe specific to <i>c-myb</i>, a hematopoietic stem cells marker. Reduced signals (arrowheads) with spatially and temporally altered distribution (arrows) were observed in embryos with severe folate deficiency. The larval responses to rescuing agents were quantified based on the distribution patterns of the <i>c-myb</i> signal at 32 hpf larvae (F) as shown in (E), and on the hemoglobin level (G) as shown in (A). There were approximately 10 to 40 larvae included for each group. (H) The 1-dpf wild-type larvae exposed to folic acid or 5-CHO-THF for 1 hour were examined for oxidative stress with H2DCFDA staining. C or CTL, heat-shocked non-fluorescent transgenic control; M or MFD, mild folate deficiency; S or SFD, severe folate deficiency; 5-CHO, 5-formyltetrahydrofolate; NAC, N-acetyl-L-cysteine; FA, folic acid. *, p<0.05; **, p<0.01; ***, p<0.001.</p

    Prospective causal links involved in FD-induced developmental defects of the zebrafish circulation system.

    No full text
    <p>FD increases embryonic oxidative stress, leading to activated Erk (I) and impeded cell migration (II), which contributed to impeded hematopoiesis and cardiogenesis, respectively. FD also disturbed one-carbon metabolism, which interferes with nucleotide synthesis (III) and cell proliferation/hematopoiesis. The impeded one-carbon metabolism may also disturb intracellular methylation potential, which hampers primordial cell migration (II), leading to cardiogenic defects. This scheme is depicted based on the results reported in the current study (solid lines) and those in the literature (dashed line).</p
    corecore