首页> 外文OA文献 >Mechanisms for Arsenic-Stimulated Sinusoidal Endothelial Cell Capillarization
【2h】

Mechanisms for Arsenic-Stimulated Sinusoidal Endothelial Cell Capillarization

机译:砷刺激的鼻窦内皮细胞毛细血管化的机制

摘要

The vascular effects of arsenic in drinking water are a global public health concern that contribute to disease in millions of people worldwide. However, the cellular and molecular mechanisms for these pathogenic effects of arsenic are not well defined. This thesis examined the hypothesis that arsenic stimulates pathogenic signals through surface receptors on liver sinusoidal endothelial cells (LSECs) to stimulate NADPH oxidase (NOX) activity that is required for arsenic-stimulated LSEC capillarization. In mice and isolated LSECs, we demonstrated that exposure to arsenic promoted capillarization and increased expression of platelet endothelial cell adhesion molecule (PECAM-1) through a time and dose dependent mechanism. Superoxide generating NOX enzyme complexes participate in vascular remodeling and angiogenesis and are central to arsenic stimulated cell signaling. LSEC arsenic exposure increased NOX dependent superoxide generation that was inhibited using gp91ds-tat protein, NSC23766, a Rac1-GTPase inhibitor, or quenched by the intracellular superoxide scavenger, Tempol. These inhibitors also blocked arsenic-stimulated LSEC PECAM-1 expression and defenestration. In vivo arsenic exposures failed to promote LSEC capillarization in p47phox knockout mice. These data demonstrated that arsenic stimulates capillarization through a NOX dependent mechanism. Given that arsenic rapidly activates NOX in vascular cells, we hypothesized that signaling for these responses was receptor mediated. Since arsenic-stimulated LSEC defenestration and capillarization is Rac1 and NOX dependent, we examined whether a g-protein coupled receptor (GPCR) upstream of Rac1 initiated these effects. Pre-treatment LSECs with Pertussis toxin (PTX), an inhibitor of Gi/o, prevented arsenic-stimulated defenestration. Since capillarization is a gain in barrier function, LSEC expression of the sphingosine-1-phosphate type 1 (S1P1) receptor, a major Gi/o linked regulator of endothelial barrier function, and its role in arsenic-stimulated defenestration were investigated. S1P1 was highly expressed in LSECs relative to large vessels. In ex vivo studies, inhibiting LSEC S1P1 with a selective antagonist, VPC23109, blocked arsenic-stimulated superoxide generation, defenestration, and PECAM-1 expression. These data demonstrated that arsenic targets a specific LSEC GPCR to promote vascular remodeling, and the first demonstrating that S1P1 regulates oxidant-dependent LSEC capillarization. Taken together, these data demonstrate that S1P1 activated NOX stimulates LSEC capillarization, which aids in our understanding of mechanisms underlying arsenic-induced liver disease.
机译:饮用水中砷对血管的影响是全球性的公共卫生问题,它引起了全球数百万人的疾病。但是,对砷的这些致病作用的细胞和分子机制尚不明确。本论文检验了如下假说:砷通过肝窦形内皮细胞(LSEC)上的表面受体刺激病原体信号,从而刺激NADPH氧化酶(NOX)活性,这是砷刺激LSEC毛细血管化所必需的。在小鼠和孤立的LSEC中,我们证明了砷暴露通过时间和剂量依赖性机制促进了毛细血管化和血小板内皮细胞粘附分子(PECAM-1)的表达增加。产生超氧化物的NOX酶复合物参与血管重塑和血管生成,并且对砷刺激的细胞信号传导至关重要。 LSEC砷暴露增加了NOx依赖的超氧化物的生成,使用gp91ds-tat蛋白NSC23766(一种Rac1-GTPase抑制剂)可抑制NOX依赖的超氧化物的生成,或通过细胞内超氧化物清除剂Tempol淬灭。这些抑制剂还阻断了砷刺激的LSEC PECAM-1的表达和防御。体内砷暴露未能促进p47phox基因敲除小鼠的LSEC毛细血管化。这些数据表明砷通过NOX依赖性机制刺激毛细血管化。鉴于砷迅速激活了血管细胞中的NOX,我们假设这些反应的信号传导是受体介导的。由于砷刺激的LSEC脱孔和毛细血管化依赖于Rac1和NOX,因此我们检查了Rac1上游的g蛋白偶联受体(GPCR)是否引发了这些作用。用Gi / o抑制剂百日咳毒素(PTX)预处理LSEC可以防止砷刺激的去窗孔。由于毛细血管化是屏障功能的获得,因此研究了鞘氨醇-1-磷酸1型(S1P1)受体(血管内皮屏障功能的主要Gi / o关联调节剂)的LSEC表达及其在砷刺激的脱窗作用中的作用。相对于大型血管,S1P1在LSEC中高表达。在离体研究中,用选择性拮抗剂VPC23109抑制LSEC S1P1可以阻断砷刺激的超氧化物的产生,防御和PECAM-1的表达。这些数据表明,砷靶向特定的LSEC GPCR以促进血管重塑,并且第一个证明S1P1调节依赖于氧化剂的LSEC毛细血管化。综上所述,这些数据表明S1P1激活的NOX刺激LSEC毛细血管化,这有助于我们了解砷诱发的肝病的潜在机制。

著录项

  • 作者

    Straub Adam C.;

  • 作者单位
  • 年度 2009
  • 总页数
  • 原文格式 PDF
  • 正文语种 en
  • 中图分类

相似文献

  • 外文文献
  • 中文文献
  • 专利

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号