14 research outputs found

    Increased Oxidative and Nitrative Stress Accelerates Aging of the Retinal Vasculature in the Diabetic Retina.

    No full text
    Hyperglycemia-induced retinal oxidative and nitrative stress can accelerate vascular cell aging, which may lead to vascular dysfunction as seen in diabetes. There is no information on whether this may contribute to the progression of diabetic retinopathy (DR). In this study, we have assessed the occurrence of senescence-associated markers in retinas of streptozotocin-induced diabetic rats at 8 and 12 weeks of hyperglycemia as compared to normoglycemic aging (12 and 14 months) and adult (4.5 months) rat retinas. We have found that in the diabetic retinas there was an up-regulation of senescence-associated markers SA-β-Gal, p16INK4a and miR34a, which correlated with decreased expression of SIRT1, a target of miR34a. Expression of senescence-associated factors primarily found in retinal microvasculature of diabetic rats exceeded levels measured in adult and aging rat retinas. In aging rats, retinal expression of senescence associated-factors was mainly localized at the level of the retinal pigmented epithelium and only minimally in the retinal microvasculature. The expression of oxidative/nitrative stress markers such as 4-hydroxynonenal and nitrotyrosine was more pronounced in the retinal vasculature of diabetic rats as compared to normoglycemic aging and adult rat retinas. Treatments of STZ-rats with the anti-nitrating drug FeTPPS (10mg/Kg/day) significantly reduced the appearance of senescence markers in the retinal microvasculature. Our results demonstrate that hyperglycemia accelerates retinal microvascular cell aging whereas physiological aging affects primarily cells of the retinal pigmented epithelium. In conclusion, hyperglycemia-induced retinal vessel dysfunction and DR progression involve vascular cell senescence due to increased oxidative/nitrative stress

    SA-β-gal in RPE flat mounts.

    No full text
    <p>Bright-field images show SA-<b>β</b>-gal positive areas found in RPE flat mounts from age-matched (4.5 months old) normoglycemic control rats (<b>A</b>),STZ-rats, at 8 (<b>B</b>) and 12 weeks (<b>C</b>) of hyperglycemia, or in normoglycemic aging rats at12 (<b>D</b>) and 14 (<b>E</b>) months of age. Scale bar equal to <i>20</i><b>μ</b><i>m</i>.</p

    Measurements of nitrative modifications.

    No full text
    <p>Representative images of immunohistochemical analysis using anti-nitrotyrosine in retinal flat mounts to demonstrate changes in retinal NY formation in the different treatment groups. Double labeling with isolectin B4 was used to specifically assess NY immunoreactivity in the retinal microvasculature. Areas of merging labeling (yellow) are indicated by the white arrows. Non-vascular areas immunoreactive to NY are indicated by white arrowheads. Scale bar equal to <i>20</i><b>μ</b><i>m</i>.</p

    Measurements of p16<sup>INK4a</sup> levels in rat retinas.

    No full text
    <p><b>A)</b> Expression of p16<sup>INK4a</sup> at mRNA level was measured using qPCR in retinal extracts from control, STZ, and aging rat retinas (as indicated above). Levels of p16<sup>INK4A</sup> specific mRNA are expressed as a ratio to <b>β</b>-actin and normalized to baseline controls. x ± S.D, *p<0.009 vs control 4.5 month rat retina; #p<0.01 <i>vs</i> D<sub>8wks</sub>, n = 6. <b>B)</b> Western blotting analysis measuring p16<sup>INK4a</sup> protein levels; bar histogram depicts p16<sup>INK4a</sup> protein levels normalized to <b>β</b>-actin in retinal extracts. x ± S.D,*p<0.01 <i>vs</i> C; <sup>#</sup>p<0.04 <i>vs</i> D<sub>8wks</sub> diabetic, n = 6. Control retinas = white bar; aging retinas = gray bar; diabetic retinas = black bar. <b>C-G)</b> Frozen retinal sections were probed with anti-p16<sup>INK4a</sup> (<i>green</i>) antibodies and isolectin B4 (<i>red</i>) to detect anti-p16<sup>INK4a</sup> -specific immunoreactivity in retinal vessels of control (<b>C</b>), diabetic (<b>D-E</b>), and aging (<b>F-G</b>) rats. Areas of merging labeling (yellow) are indicated by the white arrows. White asterisks show p16<sup>INK4a</sup> positivity at the RPE/choroid level. Hoescht staining was used to detect cellular nuclei (<i>blue</i>). Scale bar equal to 50 <b>μ</b>m.</p

    FeTPPS effects on lipid peroxidation and nitrotyrosine formation.

    No full text
    <p>Representative images of anti-nitrotyrosine (<b>A,B</b>) and anti-4-HNE (<b>C,D</b>) immunoreactivity (<i>green</i>) in retinal flat mount preparations of STZ-diabetic rats in comparison to retinas of diabetic rats that were treated with FeTPPS. Double labeling for isolectin B4 (<i>red</i>) was performed to visualize merging areas (yellow) corresponding to vascular structures. Scale bar equal to <i>20</i><b>μ</b><i>m</i>.</p

    Measurements of lipid oxidative modifications.

    No full text
    <p>Bar histogram is representative of measurements of chloroform/methanol extracted hydroperoxides (<b>A</b>) in retinal rat tissues in the different treatment groups. x ± S.D, *p<0.0001 <i>vs</i> C, <sup>#</sup>p<0.0001 <i>vs</i> D<sub>8wks</sub>, °p<0.0001 vs D<sub>8wks</sub>, n = 6. Immunohistochemical analysis using anti-4-HNE specific antibodies in retinal flat mounts is shown in panels B and C. Areas of merging labeling (yellow) are indicated by the white arrows. White arrowheads indicate extravascular areas immunoreactive to 4-HNE. Scale bar equal to <i>20</i><b>μ</b><i>m</i>.</p

    Assessment of miR34a expression.

    No full text
    <p>MiR-34a detection was evidenced by <i>in situ</i> hybridization in retinal sections of control (4.5 months old), 8 wks diabetic, and 14 months old aging rats. Representative images of control (<b>A</b>), diabetic (<b>B</b>), and aging (<b>C</b>) rat retinas probed for MiR-34a-DIG are depicted in each panel. <b>D)</b> Control retina probed with scrambled miRNA. Scale bar <i>50</i><b>μ</b><i>m</i>. In <b>E</b>) bar histogram representing relative fluorescence units of miR34a amplicons measured by qPCR.x ± S.D, *p<0.01 <i>vs</i> C 4.5 month rat retina, n = 6.</p
    corecore