11 research outputs found

    IDH3 mediates apoptosis of alveolar epithelial cells type 2 due to mitochondrial Ca2+ uptake during hypocapnia

    Get PDF
    In adult respiratory distress syndrome (ARDS) pulmonary perfusion failure increases physiologic dead-space (VD/VT) correlating with mortality. High VD/VT results in alveolar hypocapnia, which has been demonstrated to cause edema formation, atelectasis, and surfactant depletion, evoked, at least in part, by apoptosis of alveolar epithelial cells (AEC). However, the mechanism underlying the hypocapnia-induced AEC apoptosis is unknown. Here, using fluorescent live-cell imaging of cultured AEC type 2 we could show that in terms of CO2 sensing the tricarboxylic acid cycle enzyme isocitrate dehydrogenase (IDH) 3 seems to be an important player because hypocapnia resulted independently from pH in an elevation of IDH3 activity and subsequently in an increase of NADH, the substrate of the respiratory chain. As a consequence, the mitochondrial transmembrane potential (ΔΨ) rose causing a Ca2+ shift from cytosol into mitochondria, whereas the IDH3 knockdown inhibited these responses. Furthermore, the hypocapnia-induced mitochondrial Ca2+ uptake resulted in reactive oxygen species (ROS) production, and both the mitochondrial Ca2+ uptake and ROS production induced apoptosis. Accordingly, we provide evidence that in AEC type 2 hypocapnia induces elevation of IDH3 activity leading to apoptosis. This finding might give new insight into the pathogenesis of ARDS and may help to develop novel strategies to reduce tissue injury in ARDS

    Tracking CNS and systemic sources of oxidative stress during the course of chronic neuroinflammation

    Get PDF
    The functional dynamics and cellular sources of oxidative stress are central to understanding MS pathogenesis but remain elusive, due to the lack of appropriate detection methods. Here we employ NAD(P)H fluorescence lifetime imaging to detect functional NADPH oxidases (NOX enzymes) in vivo to identify inflammatory monocytes, activated microglia, and astrocytes expressing NOX1 as major cellular sources of oxidative stress in the central nervous system of mice affected by experimental autoimmune encephalomyelitis (EAE). This directly affects neuronal function in vivo, indicated by sustained elevated neuronal calcium. The systemic involvement of oxidative stress is mirrored by overactivation of NOX enzymes in peripheral CD11b(+) cells in later phases of both MS and EAE. This effect is antagonized by systemic intake of the NOX inhibitor and anti-oxidant epigallocatechin-3-gallate. Together, this persistent hyper-activation of oxidative enzymes suggests an "oxidative stress memory" both in the periphery and CNS compartments, in chronic neuroinflammation

    Functional Impairment of Microglia Coincides with Beta-Amyloid Deposition in Mice with Alzheimer-Like Pathology

    Get PDF
    <div><p>Microglial cells closely interact with senile plaques in Alzheimer’s disease and acquire the morphological appearance of an activated phenotype. The significance of this microglial phenotype and the impact of microglia for disease progression have remained controversial. To uncover and characterize putative changes in the functionality of microglia during Alzheimer’s disease, we directly assessed microglial behavior in two mouse models of Alzheimer’s disease. Using <i>in vivo</i> two-photon microscopy and acute brain slice preparations, we found that important microglial functions - directed process motility and phagocytic activity - were strongly impaired in mice with Alzheimer’s disease-like pathology compared to age-matched non-transgenic animals. Notably, impairment of microglial function temporally and spatially correlated with Aβ plaque deposition, and phagocytic capacity of microglia could be restored by interventionally decreasing amyloid burden by Aβ vaccination. These data suggest that major microglial functions progressively decline in Alzheimer’s disease with the appearance of Aβ plaques, and that this functional impairment is reversible by lowering Aβ burden, e.g. by means of Aβ vaccination.</p></div

    Phagocytic capacity of cortical microglia is impaired in two mouse models of cerebral amyloidosis.

    No full text
    <p>(<b>A</b>) Representative images (left) and microglial phagocytic index (in arbitrary units, a.u., right) of 9 month old <i>APPPS1</i> mice and wildtype littermate controls (3 mice per genotype; p<0.001). Images show microglia (Iba-1, red), Aβ (4G8, blue) and fluorescent microspheres (green). Orthogonal views of z-stack images are shown in the bottom panel. (<b>B</b>) Representative images (left) and microglial phagocytic index of 20 month old <i>APP23</i> and age-matched control mice (3 mice per genotype, p<0.001, right) are shown. Data are mean ± s.e.m, ***p<0.001. Scale bars: 10 µm.</p

    Passive anti-Aβ vaccination reduces plaque burden and restores hippocampal microglial phagocytic activity.

    No full text
    <p>5 month old <i>APPPS1</i> mice (n = 3 mice per group) and wildtype littermates (n = 2 mice per group) were biweekly injected intraperitoneally with IgG (black bar) or anti-Aβ antibody (Ab9, grey bar) for 6 weeks. The area covered by Thiazine Red-positive Aβ plaques in cortex (<b>A</b>) and hippocampus (<b>B</b>) of 6.5 month old <i>APPPS1</i> mice or age-matched controls treated with IgG or Ab9 is shown in the left panel. Absolute values of microglial phagocytic indices in the cortex (<b>A</b>) and hippocampus (<b>B</b>) of the same mice are depicted on the right panel. All data are mean ± s.e.m, *p<0.05, **p<0.01. a.u. = arbitrary units.</p

    Impairment of microglial phagocytosis in <i>APPPS1</i> mice correlates with Aβ plaque deposition.

    No full text
    <p>(<b>A</b>) Aβ plaque load (brain area covered by Thiazine red-positive plaques) and relative microglial phagocytic activity normalized to corresponding wildtype littermate in the cortex of 7–9 week, 4 and 9 month old <i>APPPS1</i> mice. 7–9 week old mice were sub-classified according to apparent 4G8 positive plaque deposition as with (+) or without (−) detectable plaque load. (<b>B</b>) Correlation between extent of plaque load and relative microglial phagocytic activity in the cortex of <i>APPPS1</i> mice. (<b>C, D</b>) Thiazine red-covered area and relative phagocytic activity of microglia in the hippocampus of 7–9 week and 4 month old mice (<b>C</b>) and in the cerebellum of 4 month old <i>APPPS1</i> mice (<b>D</b>). Absolute values of microglial phagocytic indices from <i>APPPS1</i> mice were normalized to wildtype littermate controls (3–4 mice per age group and genotype, ***p<0.001). (<b>E</b>) Phagocytic index (3 independent experiments, p = 0.181) and representative images of primary microglial cultures from wildtype and <i>APPPS1</i> mice. Microglia (Iba-1, red), nuclei (DRAQ5, blue) and microspheres (green). All data are mean ± s.e.m, *p<0.05, **p<0.01. a.u. = arbitrary units. Scale bars: 10 µm.</p

    Lesion-directed microglial process movement is impaired in a mouse model of cerebral amyloidosis.

    No full text
    <p>(<b>A</b>) Representative intravital two-photon microscopy images and (<b>B</b>) time course of microglial process movement towards a laser-induced micro-lesion (dashed circle) in 8 month old live anaesthetized <i>APPPS1</i>-<i>Cx3cr1</i><sup>+/gfp</sup> (n = 6) and <i>Cx3cr1</i><sup>+/gfp</sup> mice (n = 8). Aβ plaques are stained with Methoxy-XO4 (blue, *). (<b>C</b>) Representative images and (<b>D</b>) relative microglial response to laser lesions in acute cortical cerebral slices of 10 month old <i>APPPS1</i>-<i>Cx3cr1</i><sup>+/gfp</sup> (n = 8) and <i>Cx3cr1</i><sup>+/gfp</sup> (n = 7) mice. Aβ plaques are stained with Thiazine Red (red, *). Data are mean ± s.e.m, *p<0.05. Scale bars: 10 µm. a.u. = arbitrary units.</p
    corecore