8 research outputs found

    Ugt1a1 activity toward bilirubin in Gunn rats that received conformal HIR before hepatocyte transplantation.

    No full text
    <p>Ugt1a1 activities in liver homogenates of different recipient groups as in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0046775#pone-0046775-g002" target="_blank">Figure 2</a> are shown (means±SD) as percent of the activity in the liver of Wistar-RHA rats (2.6 µmol/g liver per hour).</p

    Regiospecific liver repopulation in DPPIV<sup>−</sup> recipients by donor DPPIV<sup>+</sup> hepatocytes following conformal HIR of one half of the median lobe.

    No full text
    <p>All liver lobes, except one half of the median lobe were shielded from HIR. Three months after hepatocyte transplantation, liver sections were stained histochemically for DPPIV activity. A region at the junction of the irradiated and non-irradiated regions of the median lobe is shown. Panel a. Low power view showing clusters of DPPIV-positive donor hepatocytes (red, white arrow) in the upper part of the section that had received preparative HIR, and the absence of such clusters in the lower part of the section that had been shielded from HIR. Panel b. High power view showing a donor hepatocyte cluster. The black bars indicate 100 µm.</p

    Immunofluorescent staining for ugt1a1 in different liver lobes.

    No full text
    <p>Three months after hepatocyte transplantation following HIR to median lobe (ML), 20% of the irradiated median lobe was replaced by donor hepatocytes, whereas the unirradiated left (LL), right (RL) and caudate (CL) lobes contained only small ugt1a1+ hepatocyte clusters.</p

    Evaluation of Therapeutic Oligonucleotides for Familial Amyloid Polyneuropathy in Patient-Derived Hepatocyte-Like Cells

    No full text
    <div><p>Familial amyloid polyneuropathy (FAP) is caused by mutations of the transthyretin (<i>TTR</i>) gene, predominantly expressed in the liver. Two compounds that knockdown TTR, comprising a small interfering RNA (siRNA; ALN-TTR-02) and an antisense oligonucleotide (ASO; IONIS-TTR<sub>Rx</sub>), are currently being evaluated in clinical trials. Since primary hepatocytes from FAP patients are rarely available for molecular analysis and commercial tissue culture cells or animal models lack the patient-specific genetic background, this study uses primary cells derived from urine of FAP patients. Urine-derived cells were reprogrammed to induced pluripotent stem cells (iPSCs) with high efficiency. Hepatocyte-like cells (HLCs) showing typical hepatic marker expression were obtained from iPSCs of the FAP patients. <i>TTR</i> mRNA expression of FAP HLCs almost reached levels measured in human hepatocytes. To assess TTR knockdown, siTTR1 and TTR-ASO were introduced to HLCs. A significant downregulation (>80%) of <i>TTR</i> mRNA was induced in the HLCs by both oligonucleotides. TTR protein present in the cell culture supernatant of HLCs was similarly downregulated. Gene expression of other hepatic markers was not affected by the therapeutic oligonucleotides. Our data indicate that urine cells (UCs) after reprogramming and hepatic differentiation represent excellent primary human target cells to assess the efficacy and specificity of novel compounds.</p></div

    Characterization of urine cells.

    No full text
    <p>(A) Brightfield image of UCs at p2 after isolation. FAP3 derived cells are shown as one typical example. Scale bar, 100 μM. (B) Flow cytometry analysis of UCs at p2 (n = 4). Total expression of markers used for characterization of mesenchymal stem cells is given. (C) Relative mRNA expression of epithelial, fibroblast and renal marker genes in UCs (n = 6). Data were normalized to housekeeping gene.</p

    TTR knockdown in FAP patient derived hepatocyte-like cells.

    No full text
    <p>(A) qRT-PCR analysis of <i>TTR</i> mRNA expression in FAP HLCs treated with compounds (n = 5). Mean±SE of FAP1 to FAP5 are shown. (B) Immunocytochemistry stainings of TTR in FAP HLCs 24 hours after treatment with compounds. FAP4 derived cells are shown as one typical example. Exposure time was consistently adjusted to 1/6 second. Scale bars, 50 μM. (C) TTR Western blot analysis of cell culture supernatants derived from HLCs. FAP4 derived cells are shown as one typical example. (D) Quantification of TTR expression as obtained by Western blots. Mean±SE of FAP1, FAP4 and FAP5 are shown. (E) qPCR analysis of different hepatic and housekeeping marker genes in FAP HLCs after treatment with siTTR1 (triangle) and TTR-ASO (squares). Mean of FAP1 to FAP5 are shown. Data were normalized and set relative to untreated control cells. Absolute ΔΔCt-values are given. Dotted line indicates significance level. ** p< 0.01; ns, not significant.</p

    Reprogramming of urine cells obtained from FAP patients.

    No full text
    <p>(A) Typical brightfield images of UCs from a FAP patient at different times after reprogramming. FAP2 derived cells are shown. Scale bars, 100 μM. (B) Immunofluorescence stainings of cells after reprogramming. DAPI was used for nuclear counterstaining. FAP5 derived cells are shown as one typical example. Scale bars, 50 μM. (C) Gene expression analysis of reprogrammed cells. Relative expression to housekeeping gene is shown. Mean±SE of FAP1 to FAP5 are shown. (D) EB expression profile of pluripotency- and lineage-specific markers derived from FAP iPSCs. Dots indicate sample scores of cells derived from patients FAP1 (black), FAP2 (red) and FAP4 (green). Human embryonic stem (hES) and iPS cell lines served as control (box plot). (E) DNA sequencing chromatograms of the <i>TTR</i> gene derived from FAP iPSCs. Heterozygotic variant nucleotides are shown by arrow.</p
    corecore