17 research outputs found

    A hematopoietic contribution to microhemorrhage formation during antiviral CD8 T cell-initiated blood-brain barrier disruption

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>The extent to which susceptibility to brain hemorrhage is derived from blood-derived factors or stromal tissue remains largely unknown. We have developed an inducible model of CD8 T cell-initiated blood-brain barrier (BBB) disruption using a variation of the Theiler's murine encephalomyelitis virus (TMEV) model of multiple sclerosis. This peptide-induced fatal syndrome (PIFS) model results in severe central nervous system (CNS) vascular permeability and death in the C57BL/6 mouse strain, but not in the 129 SvIm mouse strain, despite the two strains' having indistinguishable CD8 T-cell responses. Therefore, we hypothesize that hematopoietic factors contribute to susceptibility to brain hemorrhage, CNS vascular permeability and death following induction of PIFS.</p> <p>Methods</p> <p>PIFS was induced by intravenous injection of VP2<sub>121-130 </sub>peptide at 7 days post-TMEV infection. We then investigated brain inflammation, astrocyte activation, vascular permeability, functional deficit and microhemorrhage formation using T2*-weighted magnetic resonance imaging (MRI) in C57BL/6 and 129 SvIm mice. To investigate the contribution of hematopoietic cells in this model, hemorrhage-resistant 129 SvIm mice were reconstituted with C57BL/6 or autologous 129 SvIm bone marrow. Gadolinium-enhanced, T1-weighted MRI was used to visualize the extent of CNS vascular permeability after bone marrow transfer.</p> <p>Results</p> <p>C57BL/6 and 129 SvIm mice had similar inflammation in the CNS during acute infection. After administration of VP2<sub>121-130 </sub>peptide, however, C57BL/6 mice had increased astrocyte activation, CNS vascular permeability, microhemorrhage formation and functional deficits compared to 129 SvIm mice. The 129 SvIm mice reconstituted with C57BL/6 but not autologous bone marrow had increased microhemorrhage formation as measured by T2*-weighted MRI, exhibited a profound increase in CNS vascular permeability as measured by three-dimensional volumetric analysis of gadolinium-enhanced, T1-weighted MRI, and became moribund in this model system.</p> <p>Conclusion</p> <p>C57BL/6 mice are highly susceptible to microhemorrhage formation, severe CNS vascular permeability and morbidity compared to the 129 SvIm mouse. This susceptibility is transferable with the bone marrow compartment, demonstrating that hematopoietic factors are responsible for the onset of brain microhemorrhage and vascular permeability in immune-mediated fatal BBB disruption.</p

    Induction of Blood Brain Barrier Tight Junction Protein Alterations by CD8 T Cells

    Get PDF
    Disruption of the blood brain barrier (BBB) is a hallmark feature of immune-mediated neurological disorders as diverse as viral hemorrhagic fevers, cerebral malaria and acute hemorrhagic leukoencephalitis. Although current models hypothesize that immune cells promote vascular permeability in human disease, the role CD8 T cells play in BBB breakdown remains poorly defined. Our laboratory has developed a novel murine model of CD8 T cell mediated central nervous system (CNS) vascular permeability using a variation of the Theiler's virus model of multiple sclerosis. In previous studies, we observed that MHC class II−/− (CD4 T cell deficient), IFN-γR−/−, TNF-α−/−, TNFR1−/−, TNFR2−/−, and TNFR1/TNFR2 double knockout mice as well as those with inhibition of IL-1 and LTβ activity were susceptible to CNS vascular permeability. Therefore, the objective of this study was to determine the extent immune effector proteins utilized by CD8 T cells, perforin and FasL, contributed to CNS vascular permeability. Using techniques such as fluorescent activated cell sorting (FACS), T1 gadolinium-enhanced magnetic resonance imaging (MRI), FITC-albumin leakage assays, microvessel isolation, western blotting and immunofluorescent microscopy, we show that in vivo stimulation of CNS infiltrating antigen-specific CD8 T cells initiates astrocyte activation, alteration of BBB tight junction proteins and increased CNS vascular permeability in a non-apoptotic manner. Using the aforementioned techniques, we found that despite having similar expansion of CD8 T cells in the brain as wildtype and Fas Ligand deficient animals, perforin deficient mice were resistant to tight junction alterations and CNS vascular permeability. To our knowledge, this study is the first to demonstrate that CNS infiltrating antigen-specific CD8 T cells have the capacity to initiate BBB tight junction disruption through a non-apoptotic perforin dependent mechanism and our model is one of few that are useful for studies in this field. These novel findings are highly relevant to the development of therapies designed to control immune mediated CNS vascular permeability

    Astrocyte activation and occludin degradation occur prior to vascular permeability and motor deficits.

    No full text
    <p>At 0, 4, 12 and 24 hours post administration of VP2<sub>121–130</sub> peptide, C57BL/6 mice were analyzed for (A) FITC-Albumin leakage in whole brain, (B) motor performance using rotarod analysis, (C) GFAP expression in whole brain, (F) Occludin protein levels in isolated brain microvessels, (E) Claudin-5 protein levels in isolated brain microvessels and (D) active caspase-3 protein levels in isolated brain microvessels. Mice were analyzed with rotarod analysis (B) just prior to harvesting CNS tissue. Also shown are 24 hours post-administration of control E7 peptide or VP2<sub>121–130</sub> peptide in C57BL/6 Prf1<sup>−/−</sup> mouse tight junction protein levels of (G) claudin-5, (H) occludin.</p

    Vascular permeability following administration of VP2<sub>121–130</sub> peptide is dependent on perforin expression.

    No full text
    <p>Analysis of blood brain barrier disruption in a representative C57BL/6, C57BL/6 Prf1<sup>−/−</sup> and C57BL/6 FasL<sup>−/−</sup> mouse was analyzed with T1 weighted gadolinium-enhanced MRI. Shown is the extent of gadolinium leakage in C57BL/6 (A, D), C57BL/6 Prf1<sup>−/−</sup> (B, E), and C57BL/6 FasL<sup>−/−</sup> (C, F) mice 24 hours post administration of control E7 peptide or VP2<sub>121–130</sub> peptide. Upper panels (A–C) denote E7 peptide administration, lower panels (D–F) denote VP2<sub>121–130</sub> peptide administration.</p

    D<sup>b</sup>:VP2<sub>121–130</sub> epitope dominance among CNS infiltrating CD8 T cells isolated from the brain seven days post-TMEV infection.

    No full text
    <p>Shown are brain infiltrating CD8+ cells isolated from (A) C57BL/6 mice, (B) C57BL/6 Prf1<sup>−/−</sup> mice and (C) C57BL/6 FasL<sup>−/−</sup> mice were gated and analyzed for CD44 activation marker expression and D<sup>b</sup>:VP2<sub>121–130</sub> tetramer staining. In (D), C57BL/6 CD8+ cells were stained with negative control D<sup>b</sup>:E7 tetramer and anti-CD44. Upper gates denote percentage of CD44+ cells that stain with D<sup>b</sup>:VP2<sub>121–130</sub> or D<sup>b</sup>:E7 tetramer.</p

    Contrasting roles for CD4 vs. CD8 T-cells in a murine model of virally induced "T1 black hole" formation.

    Get PDF
    MRI is sensitive to tissue pathology in multiple sclerosis (MS); however, most lesional MRI findings have limited correlation with disability. Chronic T1 hypointense lesions or "T1 black holes" (T1BH), observed in a subset of MS patients and thought to represent axonal damage, show moderate to strong correlation with disability. The pathogenesis of T1BH remains unclear. We previously reported the first and as of yet only model of T1BH formation in the Theiler's murine encephalitis virus induced model of acute CNS neuroinflammation induced injury, where CD8 T-cells are critical mediators of axonal damage and related T1BH formation. The purpose of this study was to further analyze the role of CD8 and CD4 T-cells through adoptive transfer experiments and to determine if the relevant CD8 T-cells are classic epitope specific lymphocytes or different subsets. C57BL/6 mice were used as donors and RAG-1 deficient mice as hosts in our adoptive transfer experiments. In vivo 3-dimensional MRI images were acquired using a 7 Tesla small animal MRI system. For image analysis, we used semi-automated methods in Analyze 9.1; transfer efficiency was monitored using FACS of brain infiltrating lymphocytes. Using a peptide depletion method, we demonstrated that the majority of CD8 T-cells are classic epitope specific cytotoxic cells. CD8 T-cell transfer successfully restored the immune system's capability to mediate T1BH formation in animals that lack adaptive immune system, whereas CD4 T-cell transfer results in an attenuated phenotype with significantly less T1BH formation. These findings demonstrate contrasting roles for these cell types, with additional evidence for a direct pathogenic role of CD8 T-cells in our model of T1 black hole formation
    corecore