13 research outputs found

    Retinal Expression of Wnt-Pathway Mediated Genes in Low-Density Lipoprotein Receptor-Related Protein 5 (Lrp5) Knockout Mice

    Get PDF
    Mutations in low-density lipoprotein receptor-related protein 5 (Lrp5) impair retinal angiogenesis in patients with familial exudative vitreoretinopathy (FEVR), a rare type of blinding vascular eye disease. The defective retinal vasculature phenotype in human FEVR patients is recapitulated in Lrp5 knockout (Lrp5βˆ’/βˆ’)(Lrp5^{-/-}) mouse with delayed and incomplete development of retinal vessels. In this study we examined gene expression changes in the developing Lrp5βˆ’/βˆ’Lrp5^{βˆ’/βˆ’} mouse retina to gain insight into the molecular mechanisms that underlie the pathology of FEVR in humans. Gene expression levels were assessed with an Illumina microarray on total RNA from Lrp5βˆ’/βˆ’Lrp5^{βˆ’/βˆ’} and WT retinas isolated on postnatal day (P) 8. Regulated genes were confirmed using RT-qPCR analysis. Consistent with a role in vascular development, we identified expression changes in genes involved in cell-cell adhesion, blood vessel morphogenesis and membrane transport in Lrp5βˆ’/βˆ’Lrp5^{βˆ’/βˆ’} retina compared to WT retina. In particular, tight junction protein claudin5 and amino acid transporter slc38a5 are both highly down-regulated in Lrp5βˆ’/βˆ’Lrp5^{βˆ’/βˆ’} retina. Similarly, several Wnt ligands including Wnt7b show decreased expression levels. Plasmalemma vesicle associated protein (plvap), an endothelial permeability marker, in contrast, is up-regulated consistent with increased permeability in Lrp5βˆ’/βˆ’Lrp5^{βˆ’/βˆ’} retinas. Together these data suggest that Lrp5 regulates multiple groups of genes that influence retinal angiogenesis and may contribute to the pathogenesis of FEVR

    Immunology of AAV-mediated gene transfer in the eye

    Get PDF
    The eye has been at the forefront of translational gene therapy largely owing to suitable disease targets, anatomic accessibility and well-studied immunologic privilege. These advantages have fostered research culminating in several clinical trials and AAV has emerged as the vector of choice for many ocular therapies. Pre-clinical and clinical investigations have assessed the humoral and cellular immune responses to a variety of naturally occurring and engineered AAV serotypes as well as their delivered transgenes and these data have been compared to potential clinical sequelae. Encouragingly, AAV appears safe and effective with clinical follow up surpassing 5 years in some studies. As disease targets continue to expand for AAV in the eye, thorough and deliberate assessment of immunologic safety is critical. With careful study, the development of these technologies should concurrently inform the biology of the immune response in the eye

    Neuroprotection mediated by ST266 requires full complement of proteins secreted by amnion-derived multipotent progenitor cells.

    No full text
    ST266 is the biological secretome of cultured Amnion-derived Multipotent Progenitor cells containing multiple growth factors and cytokines. While intranasally-administered ST266 improves the phenotype in experimental optic neuritis, specific ST266 components mediating these effects are not known. We compared the effects of ST266 with and without removal of large molecular weight proteins both in vitro and in the multiple sclerosis model experimental autoimmune encephalomyelitis (EAE) in C57BL/6J mice. Mice were treated daily with intranasal vehicle, ST266 or lower molecular weight fraction of ST266. Retinal ganglion cells were counted in isolated retinas, and optic nerves were assessed for inflammation and demyelination. ST266 treatment significantly improved retinal ganglion cell survival and reduced optic nerve demyelination in EAE mice. The lower molecular weight ST266 fraction significantly improved optic nerve demyelination, but only showed a trend towards improved retinal ganglion cell survival. ST266 fractions below 50kDa increased Schwann cell proliferation in vitro, but were less effective than non-fractionated ST266. Demyelination attenuation was partially associated with the lower molecular weight ST266 fraction, but removal of higher molecular weight biomolecules from ST266 diminishes its neuroprotective effects, suggesting at least some high molecular weight proteins play a role in ST266-mediated neuroprotection

    Common genes regulated in <i>Lrp5<sup>βˆ’/βˆ’</sup></i> and <i>Norrin<sup>βˆ’/βˆ’</sup></i> retinas.

    No full text
    <p>Note: Retinas were isolated from P8 <i>Lrp5<sup>βˆ’/βˆ’</sup></i> mice and age matched WT control mice. RNA was isolated and assessed with Illumina gene expression microarray. (βˆ’) indicates decreased expression in <i>Lrp5<sup>βˆ’/βˆ’</sup></i> retina compared to WT retina, and (+) indicates increased expression in <i>Lrp5<sup>βˆ’/βˆ’</sup></i> retina.</p><p>*: Genes regulated in P7 <i>Norrin<sup>βˆ’/βˆ’</sup></i> retina were adapted from Schafer et. al. IOVS, 2009 <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0030203#pone.0030203-Schafer1" target="_blank">[33]</a>.</p

    Delayed development of the superficial retinal vasculature and persistent hyaloid vessels in <i>Lrp5</i> null mice.

    No full text
    <p>(A) Left: retinal whole mounts stained with isolectin B<sub>4</sub>-594 from WT and <i>Lrp5</i> null mice at post-natal day (P) 8. Right: quantification of vascularized retinal area in WT and <i>Lrp5</i> null mice at P8. nβ€Š=β€Š5–10 per group, ***<i>p</i><0.001. (B) Retinal cross sections of WT mice and <i>Lrp5</i> null mice at P8 stained for endothelial cells with isolectin B<sub>4</sub>-594 (red) and cell nuclei (DAPI, blue). Arrows indicate persistent hyaloid vessels in <i>Lrp5</i> null retina. Scale bars: 500 Β΅m.</p

    Abnormal vascular development in the inner and outer retina and forebrain of <i>Lrp5</i> null mice.

    No full text
    <p>(A) Retinal whole mounts of WT and <i>Lrp5</i> null mice stained for endothelial cells with isolectin B<sub>4</sub>-594 at P12 and 17. Enlarged images highlight the abnormal vessel growth in the <i>Lrp5</i> null retina. (B) Retinal cross sections from P30 WT and <i>Lrp5</i> null mice stained for endothelial cells with isolectin B<sub>4</sub>-594 (red) and cell nuclei with DAPI (blue). Arrows indicate deep layers of retinal vasculature which is present in WT retina but absent in <i>Lrp5</i> null retina. (C) Cross sections of the forebrain from P8 WT and <i>Lrp5</i> null mice with endothelial cells stained with isolectin B<sub>4</sub>-594. Scale bar: 100 Β΅m.</p

    Expression levels of <i>Lrp5</i>, <i>Norrin</i>, <i>Frizzled4</i> and <i>Dvl</i> mRNA during retinal development in wild type mice.

    No full text
    <p>Quantification of mRNA (A) <i>Lrp5</i>, (B) <i>Norrin</i>, (C) <i>Frizzled4</i>, (D) <i>Dvl1</i>, (E) <i>Dvl2</i>, and (F) <i>Dlv3</i> was performed with RT-qPCR during normal retinal development from P1-P17. Expression levels were normalized against house keeping gene <i>Cyclophilin A</i>. Trend lines were fitted with polynomial, linear or power regression.</p

    Confirmation of gene expression differentially regulated in Lrp5 null retina with RT-qPCR.

    No full text
    <p>Quantification of mRNA (A) <i>Cln5</i> and <i>Slc38a5</i>, (B) <i>Gja1</i>, <i>Msfd2</i>, <i>Sox18</i>, and <i>vWF</i>, and (C) <i>Plvap</i> and <i>EMP1</i> in WT and <i>Lrp5</i> null retina with RT-qPCR at P8. Expression levels were normalized against housekeeping gene <i>Cyclophilin A</i>. *<i>p</i><0.05, **<i>p</i><0.01.</p

    Wnt ligands and receptor regulated in P8 <i>Lrp5</i> null retina.

    No full text
    <p>Wnt ligand <i>Norrin</i>, Wnt5a, Wnt7b, and Wnt10b mRNA expression in WT and <i>Lrp5<sup>βˆ’/βˆ’</sup></i> whole retinas were quantified with RT-qPCR and normalized to <i>Cyclophilin A</i> expression. *<i>p</i><0.05, **<i>p</i><0.01.</p
    corecore