8 research outputs found

    The Effects of Adipose Stem Cell–Conditioned Media on Fibrogenesis of Dermal Fibroblasts Stimulated by Transforming Growth Factor-β1

    No full text
    Adipose-derived stem cells (ASCs) have been shown to enhance wound healing by human dermal fibroblasts; however, the interactions between ASCs and fibroblasts during injury remain unclear. Fibroblasts were treated with ASC-conditioned medium (ASC-CM) with and without transforming growth factor-β1 (TGF-β1) stimulation. Fibroblast proliferation, apoptosis, differentiation and expression of extracellular matrix genes and proteins, type I collagen, and type III collagen were measured. Also, wound-healing effect of ASC-CM was verified with in vivo animal study. ASC-CM inhibited proliferation and enhanced apoptosis of fibroblasts under TGF-β1 stimulation. Furthermore, 10% ASC-CM inhibited α-smooth muscle actin expression in fibroblasts, whereas 100% ASC-CM increased collagen, especially type III, expression in fibroblasts. ASC-CM was found to contain more basic fibroblast growth factor than hepatocyte growth factor, and 100% ASC-CM increased hepatocyte growth factor gene expression in fibroblasts. These results suggest ASCs affect fibrogenesis by dermal fibroblasts stimulated with TGF-β1 via paracrine signaling by adipocytokines present in ASC-CM. These results also suggest that higher concentrations of ASC-CM increase collagen production and inhibit fibroblast proliferation to avoid excessive fibrogenesis. We demonstrated that a lower ASC-CM concentration attenuated fibroblast differentiation. Additionally, 100% ASC-CM significantly reduced the wound size in an in vivo wound-healing model. In this study, we provided evidence that ASCs modulate fibrogenesis by fibroblasts via paracrine signaling, suggesting that application of ASCs during wound healing may improve the quality of wound repair

    GEF-H1-RhoA signaling pathway mediates LPS-induced NF-kB transactivation and IL-8 synthesis in endothelial cells

    Get PDF
    Secretion of proinflammatory cytokines by LPS activated endothelial cells contributes substantially to the pathogenesis of sepsis. However, the mechanism involved in this process is not well understood. In the present study, we determined the roles of GEF-H1 (Guanine-nucleotide exchange factor-H1)-RhoA signalling in LPS-induced interleukin-8 (IL-8, CXCL8) production in endothelial cells. First, we observed that GEF-H1 expression was upregulated in a dose- and time-dependent manner as consistent with TLR4 (Toll-like receptor 4) expression after LPS stimulation. Afterwards, Clostridium difficile toxin B-10463 (TcdB-10463), an inhibitor of Rho activities, reduced LPS-induced NF-κB phosphorylation. Inhibition of GEF-H1 and RhoA expression reduced LPS-induced NF-κB and p38 phosphorylation. TLR4 knockout blocked LPS-induced activity of RhoA, however, MyD88 knockout did not impair the LPS-induced activity of RhoA. Nevertheless, TLR4 and MyD88 knockout both significantly inhibited transactivation of NF-κB. GEF-H1-RhoA and MyD88 both induced significant changes in NF-κB transactivation and IL-8 synthesis. Co-inhibition of GEF-H1-RhoA and p38 expression produced similar inhibitory effects on LPS-induced NF-κB transactivation and IL-8 synthesis as inhibition of p38 expression alone, thus confirming that activation of p38 was essential for the GEF-H1-RhoA signalling pathway to induce NF-κB transactivation and IL-8 synthesis. Taken together, these results demonstrate that LPS-induced NF-κB activation and IL-8 synthesis in endothelial cells are regulated by the MyD88 pathway and GEF-H1-RhoA pathway

    GSK-3Beta-Dependent Activation of GEF-H1/ROCK Signaling Promotes LPS-Induced Lung Vascular Endothelial Barrier Dysfunction and Acute Lung Injury

    No full text
    The bacterial endotoxin or lipopolysaccharide (LPS) leads to the extensive vascular endothelial cells (EC) injury under septic conditions. Guanine nucleotide exchange factor-H1 (GEF-H1)/ROCK signaling not only involved in LPS-induced overexpression of pro-inflammatory mediator in ECs but also implicated in LPS-induced endothelial hyper-permeability. However, the mechanisms behind LPS-induced GEF-H1/ROCK signaling activation in the progress of EC injury remain incompletely understood. GEF-H1 localized on microtubules (MT) and is suppressed in its MT-bound state. MT disassembly promotes GEF-H1 release from MT and stimulates downstream ROCK-specific GEF activity. Since glycogen synthase kinase (GSK-3beta) participates in regulating MT dynamics under pathologic conditions, we examined the pivotal roles for GSK-3beta in modulating LPS-induced activation of GEF-H1/ROCK, increase of vascular endothelial permeability and severity of acute lung injury (ALI). In this study, we found that LPS induced human pulmonary endothelial cell (HPMEC) monolayers disruption accompanied by increase in GSK-3beta activity, activation of GEF-H1/ROCK signaling and decrease in beta-catenin and ZO-1 expression. Inhibition of GSK-3beta reduced HPMEC monolayers hyper-permeability and GEF-H1/ROCK activity in response to LPS. GSK-3beta/GEF-H1/ROCK signaling is implicated in regulating the expression of beta-catenin and ZO-1. In vivo, GSK-3beta inhibition attenuated LPS-induced activation of GEF-H1/ROCK pathway, lung edema and subsequent ALI. These findings present a new mechanism of GSK-3beta-dependent exacerbation of lung micro-vascular hyper-permeability and escalation of ALI via activation of GEF-H1/ROCK signaling and disruption of intracellular junctional proteins under septic condition

    Lipopolysaccharide induces human pulmonary micro-vascular endothelial apoptosis via the YAP signaling pathway

    Get PDF
    Gram-negative bacterial lipopolysaccharide (LPS) induces a pathologic increase in lung vascular leakage under septic conditions. LPS-induced human pulmonary micro-vascular endothelial cell (HPMEC) apoptosis launches and aggravates micro-vascular hyper-permeability and acute lung injury (ALI). Previous studies show that the activation of intrinsic apoptotic pathway is vital for LPS-induced EC apoptosis. Yes-associated protein (YAP) has been reported to positively regulate intrinsic apoptotic pathway in tumor cells apoptosis. However, the potential role of YAP protein in LPS-induced HPMEC apoptosis has not been determined. In this study, we found that LPS-induced activation and nuclear accumulation of YAP accelerated HPMECs apoptosis. LPS-induced YAP translocation from cytoplasm to nucleus by the increased phosphorylation on Y357 resulted in the interaction between YAP and transcription factor P73. Furthermore, inhibition of YAP by small interfering RNA (siRNA) not only suppressed the LPS-induced HPMEC apoptosis but also regulated P73-mediated up-regulation of BAX and down-regulation of BCL-2. Taken together, our results demonstrated that activation of the YAP/P73/(BAX and BCL-2)/caspase-3 signaling pathway played a critical role in LPS-induced HPMEC apoptosis. Inhibition of the YAP might be a potential therapeutic strategy for lung injury under sepsis

    Relation between dynamic changes of platelet counts and 30-day mortality in severely burned patients

    No full text
    Thrombocytopenia is a common event in severely burned patients and associated with adverse outcome. The underlying relationship between the dynamic changes of platelet counts and mortality has not been well defined. We performed a 6-year retrospective chart of adult patients with a burn index of 50 or greater admitted to two burn centers and collected data on patient demographics, laboratory results, and patient outcomes. The mean daily increase in the platelet count (∆PC/∆t) from day 3 to day 10 was calculated, and 30-day mortality was determined. For the study, 141 survivors and 65 nonsurvivors were enrolled. The sequential changes in PCs presented a biphasic pattern after admission, with a slump to the nadir during the first 3 days and a subsequent recovery. With respect to 30-day mortality, compared with the AUC of APACHE-Ⅱ score (0.841), no significant difference was noted between ΔPC/ΔT and APACHE-Ⅱ score (p = 0.0648). The ΔPC/ΔT associated with the best discrimination between survivors and nonsurvivors was 20.57 × 109/L due to the cutoff with optimal Youden index (0.453). By multiple logistic regression, ΔPC/ΔT < 20.57 × 109/L was one of the prognostic predictors of 30-day mortality. Furthermore, Kaplan–Meier estimates of hospital survival according to the size of ΔPC/ΔT revealed that a blunted increase with ΔPC/ΔT < 20.57 × 109/L was associated with increased 30-day mortality. A blunted daily increase in PCs, especially ΔPC/ΔT < 20.57 × 109/L, is associated with increased 30-day mortality, which provides prognostic information for mortality risk assessment in severely burned patients
    corecore