9 research outputs found

    Antibody Targeting of Cathepsin S Inhibits Angiogenesis and Synergistically Enhances Anti-VEGF

    Get PDF
    Angiogenesis is a key hallmark of tumourigenesis and its inhibition is a proven strategy for the development of novel anti-cancer therapeutics. An important aspect of early angiogenesis is the co-ordinated migration and invasion of endothelial cells through the hypoxic tumour tissue. Cathepsin S has been shown to play an important role in angiogenesis as has vascular endothelial growth factor (VEGF). We sought to assess the anti-angiogenic effect of Fsn0503, a novel cathepsin S inhibitory antibody, when combined with anti-VEGF on vascular development. where it significantly retarded the development of vasculature in human xenograft models. Furthermore, when Fsn0503 was combined with an anti-VEGF antibody, a synergistic inhibition of microvascular development was observed.Taken together, this data demonstrates that the antibody-mediated targeting of cathepsin S represents a novel method of inhibiting angiogenesis. Furthermore, when used in combination with anti-VEGF therapies, Fsn0503 has the potential to significantly enhance current treatments of tumour neovascularisation and may also be of use in the treatment of other conditions associated with inappropriate angiogenesis

    Slc25a12 disruption alters myelination and neurofilaments: a model for a hypomyelination syndrome and childhood neurodevelopmental disorders

    No full text
    BACKGROUND: SLC25A12, a susceptibility gene for autism spectrum disorders that is mutated in a neurodevelopmental syndrome, encodes a mitochondrial aspartate-glutamate carrier (aspartate-glutamate carrier isoform 1 [AGC1]). AGC1 is an important component of the malate/aspartate shuttle, a crucial system supporting oxidative phosphorylation and adenosine triphosphate production. METHODS: We characterized mice with a disruption of the Slc25a12 gene, followed by confirmatory in vitro studies. RESULTS: Slc25a12-knockout mice, which showed no AGC1 by immunoblotting, were born normally but displayed delayed development and died around 3 weeks after birth. In postnatal day 13 to 14 knockout brains, the brains were smaller with no obvious alteration in gross structure. However, we found a reduction in myelin basic protein (MBP)-positive fibers, consistent with a previous report. Furthermore, the neocortex of knockout mice contained abnormal neurofilamentous accumulations in neurons, suggesting defective axonal transport and/or neurodegeneration. Slice cultures prepared from knockout mice also showed a myelination defect, and reduction of Slc25a12 in rat primary oligodendrocytes led to a cell-autonomous reduction in MBP expression. Myelin deficits in slice cultures from knockout mice could be reversed by administration of pyruvate, indicating that reduction in AGC1 activity leads to reduced production of aspartate/N-acetylaspartate and/or alterations in the dihydronicotinamide adenine dinucleotide/nicotinamide adenine dinucleotide(+) ratio, resulting in myelin defects. CONCLUSIONS: Our data implicate AGC1 activity in myelination and in neuronal structure and indicate that while loss of AGC1 leads to hypomyelination and neuronal changes, subtle alterations in AGC1 expression could affect brain development, contributing to increased autism susceptibility

    Fsn0503 inhibits angiogenesis <i>in vivo</i>; CD34 Immunohistochemical analysis of Fsn0503 treated tumors.

    No full text
    <p>(<b>A</b>) Pattern of small vessel (white arrows) and large vessel (black arrows) distribution in isotype control and Fsn0503 treated tumors (10 mg/kg 5 times a week for 4 weeks) (20×). (<b>B</b>) Analysis of total vessel number as characterized by CD34 staining shows that Fsn0503 caused an increase in small vessel number and a significant decrease in the number of large vessels (p<0.001). (<b>C</b>) A significant reduction is observed in the mean vessel area of tumours treated with Fsn0503 (p<0.001).</p

    Fsn0503 demonstrates anti-angiogenic effects <i>in vitro</i>.

    No full text
    <p>(<b>A</b>) Fsn0503 attenuates HUVEC invasion using a modified Boyden chamber assay at 500 nM. (<b>B</b>) Fsn0503 (500 nM) inhibits HUVEC degradation of quenched fluorescent substrate DQ gelatin. Cells treated with Fsn0503 or controls were analysed for the presence of fluorescent degradation products by confocal microscopy (representative images shown). (<b>C</b>) A reduction of 70% of DQ gelatin degradation was observed when quantified by mean energy per cell (p = 0.03) (5 fields per assay). (<b>D</b>) Fsn0503 (400 nM) inhibits HUVEC tube formation compared to isotype control (representative images shown). (<b>E</b>) Tube formation was assessed by counting nodes with 1, 2, 3, or more branches and the average calculated per field of view. The number of nodes with 3 or more branches, which are indicative of normal tube formation, were significantly reduced in Fsn0503 treated samples (p<0.01).</p

    Cathepsin S expression and activity is up-regulated by pro-angiogenic stimuli.

    No full text
    <p>(<b>A</b>) Total RNA was isolated from endothelial cell lines (HUVEC and HMEC-1) and used to assess Cathepsin S mRNA by RT-PCR. GAPDH serves as a control. (<b>B,C</b>) Human recombinant VEGF (10 ng/ml) up-regulates cathepsin S mRNA and protein in HUVECs when assessed by RT-PCR and western blot using GAPDH and tubulin as respective controls (<b>D</b>) Human recombinant VEGF (10 ng/ml) stimulates Cathepsin S-like activity, as assessed by the cleavage of fluorigenic substrate, Cbz-Val-Val-Arg-AMC, by 37% in HUVEC cell lysates. (<b>E,F</b>) To demonstrate the effect of hypoxia on Cathepsin S up-regulation total RNA and protein was isolated from HUVEC endothelial cells (grown in normal or hypoxic conditions) and examined by RT-PCR and western blot. GAPDH and tubulin serves as a control.</p
    corecore