36 research outputs found

    A Role for VEGFR2 Activation in Endothelial Responses Caused by Barrier Disruptive OxPAPC Concentrations

    Get PDF
    Introduction: Oxidation products of 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphatidylcholine (OxPAPC) differentially modulate endothelial cell (EC) barrier function in a dose-dependent fashion. Vascular endothelial growth factor receptor-2 (VEGFR2) is involved in the OxPAPC-induced EC inflammatory activation. This study examined a role of VEGFR2 in barrier dysfunction caused by high concentrations of OxPAPC and evaluated downstream signaling mechanisms resulting from the effect of OxPAPC in EC from pulmonary and systemic circulation

    Stiffness-activated GEF-H1 expression exacerbates LPS-induced lung inflammation.

    No full text
    Acute lung injury (ALI) is accompanied by decreased lung compliance. However, a role of tissue mechanics in modulation of inflammation remains unclear. We hypothesized that bacterial lipopolysacharide (LPS) stimulates extracellular matrix (ECM) production and vascular stiffening leading to stiffness-dependent exacerbation of endothelial cell (EC) inflammatory activation and lung barrier dysfunction. Expression of GEF-H1, ICAM-1, VCAM-1, ECM proteins fibronectin and collagen, lysyl oxidase (LOX) activity, interleukin-8 and activation of Rho signaling were analyzed in lung samples and pulmonary EC grown on soft (1.5 or 2.8 kPa) and stiff (40 kPa) substrates. LPS induced EC inflammatory activation accompanied by expression of ECM proteins, increase in LOX activity, and activation of Rho signaling. These effects were augmented in EC grown on stiff substrate. Stiffness-dependent enhancement of inflammation was associated with increased expression of Rho activator, GEF-H1. Inhibition of ECM crosslinking and stiffening by LOX suppression reduced EC inflammatory activation and GEF-H1 expression in response to LPS. In vivo, LOX inhibition attenuated LPS-induced expression of GEF-H1 and lung dysfunction. These findings present a novel mechanism of stiffness-dependent exacerbation of vascular inflammation and escalation of ALI via stimulation of GEF-H1-Rho pathway. This pathway represents a fundamental mechanism of positive feedback regulation of inflammation

    Effect of substrate stiffness and LOX inhibition on LPS-induced EC activation.

    No full text
    <p><b>A</b> - Pulmonary EC were grown on polyacrylamide gels of different stiffness (1.5 kPa, and 40 kPa) and treated with TNFα (2 ng/ml) or LPS (200 ng/ml) for 6 hrs. <b>Left panel</b>: Expression of ICAM-1 and VCAM-1 was determined by western blot analysis. Equal protein loading was confirmed by membrane re-probing with β-actin antibody. Bar graphs depict the quantitative analysis of western blot densitometry data; *P<0.05 vs. 1.5 kPa; n = 4. <b>Right panel</b>: Analysis of fibronectin mRNA levels after 12-hr treatment with LPS or TNFα was performed by RT-PCR; *p<0.05. <b>B</b> – Pulmonary EC were cultured on 2.8 kPa substrates for 3 days in the presence of LPS (200 ng/ml) with or without BAPN (300 µM). After cell detachment, fresh EC were plated on deposited extracellular matrix and stimulated with TNFα (2 ng/ml, 6 hrs). ICAM-1 expression and MYPT phosphorylation was analyzed by western blot. Equal protein loading was confirmed by membrane re-probing with β-actin antibody. Bar graphs depict the quantitative analysis of western blot densitometry data; *P<0.05 vs. LPS+BAPN treatment; n = 4. <b>C</b> - IL-8 production in response to TNFα (2 ng/ml, 6 hrs) was evaluated by ELISA assay. *P<0.05.</p
    corecore