12 research outputs found

    The novel mineralocorticoid receptor antagonist finerenone attenuates neointima formation after vascular injury

    No full text
    <div><p>Background</p><p>The novel nonsteroidal mineralocorticoid receptor (MR) antagonist finerenone holds promise to be safe and efficient in the treatment of patients with heart failure and/or chronic kidney disease. However, its effects on vascular function remain elusive.</p><p>Purpose</p><p>The aim of this study was to determine the functional effect of selective MR antagonism by finerenone in vascular cells <i>in vitro</i> and the effect on vascular remodeling following acute vascular injury <i>in vivo</i>.</p><p>Methods and results</p><p><i>In vitro</i>, finerenone dose-dependently reduced aldosterone-induced smooth muscle cell (SMC) proliferation, as quantified by BrdU incorporation, and prevented aldosterone-induced endothelial cell (EC) apoptosis, as measured with a flow cytometry based caspase 3/7 activity assay.</p><p><i>In vivo</i>, oral application of finerenone resulted in an accelerated re-endothelialization 3 days following electric injury of the murine carotid artery. Furthermore, finerenone treatment inhibited intimal and medial cell proliferation following wire-induced injury of the murine femoral artery 10 days following injury and attenuated neointimal lesion formation 21 days following injury.</p><p>Conclusion</p><p>Finerenone significantly reduces apoptosis of ECs and simultaneously attenuates SMC proliferation, resulting in accelerated endothelial healing and reduced neointima formation of the injured vessels. Thus, finerenone appears to provide favorable vascular effects through restoring vascular integrity and preventing adverse vascular remodeling.</p></div

    Functional effects of finerenone <i>in vitro</i>.

    No full text
    <p>Human smooth muscle cells (SMC) and human endothelial cells (EC) were incubated either with aldosterone alone or with aldosterone and different concentrations of finerenone, each dissolved in dimethylsulfoxide (DMSO, final concentration 0.1%) for 24 hours. A-D, Cell proliferation was determined by BrdU incorporation assays (n = 10 for SMCs/n = 6 for ECs, *<i>P</i><0.05 to serum-free, #<i>P</i><0.05 to DMSO by ordinary 1way ANOVA followed by multiple comparisons using the Tukey method). E-F, Apoptosis was determined by flow-cytometry-based caspase 3/7 activity measurement (n = 3, **<i>P</i><0.01 to serum-free, #<i>P</i><0.05 and ##<i>P</i><0.01 to DMSO by ordinary 1way ANOVA followed by multiple comparisons using the Tukey method, aldosterone 10 nM was used for B and D-F).</p

    Finerenone reduces the intimal and medial leukocyte content.

    No full text
    <p>Wire-induced femoral artery dilation was performed in 10-week-old C57BL/6 mice. Finerenone or vehicle was daily delivered as oral gavage. A, Ten days after injury, leukocyte content was assessed by immunfluorescence staining for the pan-leukocyte marker CD45 (red). Co-immunostaining for CD31 (green) and staining of nuclei with DAPI (blue) was performed to assess the endothelial lining and the overall cell number for better morphological orientation and to allow quantification. B, The amount of leukocytes was determined as the total number of CD45<sup>+</sup> cells (n = 6, *<i>P</i><0.05, **<i>P</i><0.01 by ordinary 1way ANOVA followed by multiple comparisons using the Tukey method).</p

    Finerenone prevents medial and intimal cell proliferation.

    No full text
    <p>Wire-induced femoral artery dilation was performed in 10-week-old C57BL/6 mice. Finerenone or vehicle was daily delivered as oral gavage. A, Ten days after injury, cell proliferation was assessed by immunfluorescence staining for DAPI (blue), α-smooth muscle actin (α-SMA, red), and Ki-67 (green). B, The amount of proliferating cells was determined as Ki-67<sup>+</sup> cells/DAPI<sup>+</sup> cells (n = 6, **P<0.01, ***P<0.001 by ordinary 1way ANOVA followed by multiple comparisons using the Tukey method).</p

    Finerenone promotes early endothelial recovery.

    No full text
    <p>Electrical denudation of the carotid artery was performed in 10 weeks old C57BL/6J mice. Finerenone or vehicle was daily delivered as oral gavage. A, Three days following injury, endothelial regeneration was evaluated by injection of a 5% Evan’s blue solution and en face microscopy. B, The re-endothelialized distance was calculated by substraction of the deendothelialized distance from 4 mm (standardized denudated area, n = 9, **<i>P</i><0.01 by ordinary 1way ANOVA followed by multiple comparisons using the Tukey method).</p

    Role of the Phosphatase PTEN in Early Vascular Remodeling

    No full text
    <div><p>Background</p><p>The phosphatase PTEN represents an important physiological inhibitor of phosphatidylinositol-3 kinase (PI3-K)/protein kinase B (Akt) signalling, however, the functional role of PTEN in the initial phase of angioplasty-induced vascular injury remains elusive. In the present study we sought to determine PTEN's effect on vascular smooth muscle cell (VSMC) apoptosis following acute injury <i>in vivo</i> and <i>in vitro</i>.</p> <p>Methods and Results</p><p>Immunohistochemistry indicated a faint basal expression and equal distribution of PTEN in uninjured rat carotid arteries. 12 h following balloon-injury, PTEN expression was strongly increased in apoptotic (TUNEL+) VSMC. In vitro, stimulation with serum or different growth factors or subjecting VSMC to cyclic stretch had no effect on PTEN expression, whereas stimulation with H<sub>2</sub>O<sub>2</sub> robustly increased PTEN expression in a time- and dose-dependent manner. To evaluate the functional role of PTEN expression, human VSMC were transduced with WT-PTEN. Overexpression of PTEN increased the number of apoptotic VSMC (19.8%±4.4 vs. 5.6%±2.3; <i>P</i><0.001) as determined by TUNEL assay. In contrast, siRNA-mediated knock-down of PTEN attenuated the basal as well as H<sub>2</sub>O<sub>2</sub>-induced apoptosis of VSMC. Mechanistically, overexpression of PTEN prevented serum-induced Akt-phosphorylation, whereas siRNA-mediated knock down of PTEN augmented Akt-activation. Moreover, co-transfection of PTEN and a constitutive active Akt mutant prevented PTEN-dependent augmentation of VSMC apoptosis, indicating, that PTEN regulates VSMC apoptosis by inhibition of Akt phosphorylation/activation.</p> <p>Conclusion</p><p>By interfering with the PI3-K/Akt-dependent survival signalling, the oxidative stress-induced up regulation of PTEN in VSMC of injured arteries augments the sensitivity of VSMC to apoptotic stimuli in the early phase following vascular injury, augmenting the initial injury and cell loss of the injured vessel wall. Thus, these data add to our understanding of PTEN's role during vascular remodelling.</p> </div

    PTEN expression in human coronary VSMC <i>in vitro</i>.

    No full text
    <p><b>A</b>, PTEN-expression is not upregulated by mechanical stress in VSMC. Lysates of cells exposed to mechanical forces using a stretching device were analyzed by western blotting using specific antibodies. <b>B</b>, Lysates of SMC exposed to growth medium (FCS). PTEN expression was not upregulated after 24 h. Protein expression was determined using specific antibodies. Detection of Cdk4 served as loading control. <b>C</b>, Lysates of SMC exposed to oxidative stress using 500 µM H<sub>2</sub>O<sub>2</sub>. PTEN upregulation was triggered by oxidative stress within 24 h. Detection of p53 and Cdk4 served as apoptotic marker and loading control, respectively. <b>D</b>, The upregulation of PTEN protein levels was quantified by densitometric analysis of immunoblots (n = 3; *<i>P</i><0.05). <b>E</b>, The upregulation of PTEN activity is mediated by H<sub>2</sub>O<sub>2</sub>-induction. Shown is a phosphatase activity assay of immunoprecipitated protein from lysates of HcASMC with and without 24 h H<sub>2</sub>O<sub>2</sub>–treatment. Immunoprecipitations from lysates employing an IgG iso-antibody without H<sub>2</sub>O<sub>2</sub>-treatment with and without bpV supplementation, an anti-PTEN-antibody without H<sub>2</sub>O<sub>2</sub> and bpV treatment and an anti-PTEN-antibody with H<sub>2</sub>O<sub>2</sub>– and bpV-treatment served as controls. Results are expressed as mean OD650 ± SD using an ELISA plate reader (<sup>#</sup><i>P</i><0.001, *<i>P</i><0.001; n = 4).</p

    PTEN overexpression attenuates serum induced HcASMC proliferation (A). HcASMC transfected with a plasmid carrying WT-PTEN or a control vector carrying GFP alone were incubated either in basal medium or growth medium in the presence of BrdU.

    No full text
    <p>PTEN knock down enhances serum induced HcASMC proliferation (B). Cells were transfected with siRNA targeting PTEN or a scrambled control. Proliferation is expressed as mean OD450 ± SD as determined by anti-BrdU ELISA (<sup>#</sup><i>P</i><0.001, *<i>P</i><0.001; n = 4). HcASMC were transfected as following: mock transfected (without plasmid, but with transfection reagent); transfected with a plasmid coding for GFP (pGFP); transfected with a plasmid coding for PTEN (pPTEN); transfected with a non-targeting (scrambled) siRNA (Control siRNA); transfected with a targeting siRNA against PTEN (PTEN siRNA).</p

    PTEN overexpression augments SMC apoptosis under basal conditions as well as following exposure to oxidative stress.

    No full text
    <p>HcASMC transfected with a plasmid carrying WT-PTEN or a control (empty) vector and PTEN protein-expression levels were determined by immunoblotting (<b>A</b>). SMC were incubated in basal medium (<b>B</b>) and in basal medium supplemented with H<sub>2</sub>O<sub>2</sub> (<b>C</b>) and the relative number of apoptotic cells was evaluated following TUNEL-staining (expressed as % of total cells; *<i>P</i><0.001; n = 6). HcASMC were transfected as follows: non transfected (NT); transfected with an empty plasmid without PTEN-cDNA (pControl); transfected with a plasmid coding for PTEN (pPTEN).</p
    corecore