15 research outputs found

    Genome engineering of isogenic human ES cells to model autism disorders.

    Get PDF
    Isogenic pluripotent stem cells are critical tools for studying human neurological diseases by allowing one to study the effects of a mutation in a fixed genetic background. Of particular interest are the spectrum of autism disorders, some of which are monogenic such as Timothy syndrome (TS); others are multigenic such as the microdeletion and microduplication syndromes of the 16p11.2 chromosomal locus. Here, we report engineered human embryonic stem cell (hESC) lines for modeling these two disorders using locus-specific endonucleases to increase the efficiency of homology-directed repair (HDR). We developed a system to: (1) computationally identify unique transcription activator-like effector nuclease (TALEN) binding sites in the genome using a new software program, TALENSeek, (2) assemble the TALEN genes by combining golden gate cloning with modified constructs from the FLASH protocol, and (3) test the TALEN pairs in an amplification-based HDR assay that is more sensitive than the typical non-homologous end joining assay. We applied these methods to identify, construct, and test TALENs that were used with HDR donors in hESCs to generate an isogenic TS cell line in a scarless manner and to model the 16p11.2 copy number disorder without modifying genomic loci with high sequence similarity

    A novel highly potent therapeutic antibody neutralizes multiple human chemokines and mimics viral immune modulation.

    Get PDF
    Chemokines play a key role in leukocyte recruitment during inflammation and are implicated in the pathogenesis of a number of autoimmune diseases. As such, inhibiting chemokine signaling has been of keen interest for the development of therapeutic agents. This endeavor, however, has been hampered due to complexities in the chemokine system. Many chemokines have been shown to signal through multiple receptors and, conversely, most chemokine receptors bind to more than one chemokine. One approach to overcoming this complexity is to develop a single therapeutic agent that binds and inactivates multiple chemokines, similar to an immune evasion strategy utilized by a number of viruses. Here, we describe the development and characterization of a novel therapeutic antibody that targets a subset of human CC chemokines, specifically CCL3, CCL4, and CCL5, involved in chronic inflammatory diseases. Using a sequential immunization approach, followed by humanization and phage display affinity maturation, a therapeutic antibody was developed that displays high binding affinity towards the three targeted chemokines. In vitro, this antibody potently inhibits chemotaxis and chemokine-mediated signaling through CCR1 and CCR5, primary chemokine receptors for the targeted chemokines. Furthermore, we have demonstrated in vivo efficacy of the antibody in a SCID-hu mouse model of skin leukocyte migration, thus confirming its potential as a novel therapeutic chemokine antagonist. We anticipate that this antibody will have broad therapeutic utility in the treatment of a number of autoimmune diseases due to its ability to simultaneously neutralize multiple chemokines implicated in disease pathogenesis

    Inhibition of chemokine signaling on native leukocytes by MAb d5d7.

    No full text
    <p>(<b>A</b>) Inhibition by MAb d5d7 antibody, vCCI-Fc, and IgG controls of chemokine-induced phosphorylation of CCR5<sup>Ser349</sup> in CD8+ T cells. Phosphorylation was induced with a pool of 50 ng/mL each CCL3, CCL4, and CCL5 (each of which produced ∼80% maximal response when tested individually). Data are expressed as percent of the maximum number of CCR5<sup>pSer349</sup>-positive CD8+ T cells after chemokine induction without inhibitor present and are representative of results using PBMC from six different donors. (<b>B</b>) Inhibition of CD11b up-regulation on monocytes induced with a pool of CCL3 and CCL5. Chosen chemokine concentrations were those that produced ∼80% maximal response when tested individually (16 ng/mL CCL3 and 80 ng/mL CCL5). Data are expressed as percent of the maximal increase in mean fluorescence intensity determined after chemokine induction without inhibitor present and are representative of results using blood samples from eight different donors.</p

    Affinity and potency measurements of 18V4F Fab variants.

    No full text
    *<p>Underlined positions indicate mutations that were found to be enriched throughout the selection process. Affinity measurements were obtained as described in the Methods section. Standard deviations are reported and calculated from at least three independent experiments.</p

    Binding and <i>in vitro</i> activity of murine 18V4F hybridoma antibody.

    No full text
    <p>(<b>A</b>) ELISA binding of original 18V4F hybridoma antibody to a panel of chemokines (determined in triplicate, shown as mean +/βˆ’ standard deviation). Directly coated chemokines were used here for direct comparisons, however in other experiments CCL3 showed a significantly enhanced signal when biotinylated and coated on streptavidin plates. (<b>B</b>) Chemotaxis inhibition by 18V4F hybridoma antibody of CCR5-transfected Ba/F3 cells to 5 ng/mL of CCL3, CCL4, and CCL5. Data are representative of at least three similar experiments. All chemotaxis data are represented as a percent of maximum migration in the absence of inhibitors and is fit using a standard four parameter dose-response model (GraphPad).</p

    Chemotaxis inhibition by affinity matured 18V4F variants.

    No full text
    <p>Chemotaxis inhibition by humanized 18V4F Fab, d5 variant, d7 variant, d5d7, and a negative control Fab of CCR5-transfected Ba/F3 cells to 5 ng/mL of (<b>A</b>) CCL3, (<b>B</b>) CCL4, and (<b>C</b>) CCL5. Data are representative of at least two similar experiments. A loss in potency of humanized 18V4F Fab was observed compared with the 18V4F hybridoma shown in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0043332#pone-0043332-g001" target="_blank">Figure 1b</a> and is likely a result of both the humanization process and loss in avidity caused by switching from full IgG to Fab fragment.</p

    SCID-hu mouse model of leukocyte migration.

    No full text
    <p>(<b>A</b>) NSG (NOD/SCID/IL2r-Ξ³null) mice were injected i.v. with human PBMC and allowed to engraft for 10 d. MAb d5d7 was administered i.v. just before chemokines were injected s.c. in Matrigel. After 7 d the skin sites were harvested and single cell suspensions were generated. Human leukocytes were tagged with specific antibodies and analyzed by flow cytometry. (<b>B</b>) Inhibition by MAb d5d7 of skin leukocyte migration into chemokine-embedded Matrigel plugs in NSG mice engrafted with human PBMC. The negative control group consisted of animals treated with s.c. injection of Matrigel + PBS and i.v. administration of control IgG. All other groups had s.c. injections of Matrigel containing CCL3, CCL4, and CCL5 (400 ng each) with i.v. administration of PBS, control IgG, or MAb d5d7 antibody. Data were analyzed using a student t test.</p

    Comparison of vCCI and d5d7 binding epitopes.

    No full text
    <p>Competitive binding ELISA examining molecules that can disrupt the d5d7-CCL3 binding interaction using d5d7 as a homologous competitor and vCCI-Fc, commercial anti-CCL3 antibody, and control IgG as heterologous competitors. Data are representative of at least two similar experiments. Competition experiments were also completed to analyze the d5d7-CCL4 and d5d7-CCL5 binding interactions and similar binding competition was observed between d5d7 and vCCI-Fc (data not shown).</p
    corecore