首页> 美国卫生研究院文献>PLoS Pathogens >Helicobacter pylori Perturbs Iron Trafficking in the Epithelium to Grow on the Cell Surface
【2h】

Helicobacter pylori Perturbs Iron Trafficking in the Epithelium to Grow on the Cell Surface

机译:幽门螺杆菌干扰铁在上皮中的运输使其在细胞表面生长

代理获取
本网站仅为用户提供外文OA文献查询和代理获取服务,本网站没有原文。下单后我们将采用程序或人工为您竭诚获取高质量的原文,但由于OA文献来源多样且变更频繁,仍可能出现获取不到、文献不完整或与标题不符等情况,如果获取不到我们将提供退款服务。请知悉。

摘要

Helicobacter pylori (Hp) injects the CagA effector protein into host epithelial cells and induces growth factor-like signaling, perturbs cell-cell junctions, and alters host cell polarity. This enables Hp to grow as microcolonies adhered to the host cell surface even in conditions that do not support growth of free-swimming bacteria. We hypothesized that CagA alters host cell physiology to allow Hp to obtain specific nutrients from or across the epithelial barrier. Using a polarized epithelium model system, we find that isogenic ΔcagA mutants are defective in cell surface microcolony formation, but exogenous addition of iron to the apical medium partially rescues this defect, suggesting that one of CagA's effects on host cells is to facilitate iron acquisition from the host. Hp adhered to the apical epithelial surface increase basolateral uptake of transferrin and induce its transcytosis in a CagA-dependent manner. Both CagA and VacA contribute to the perturbation of transferrin recycling, since VacA is involved in apical mislocalization of the transferrin receptor to sites of bacterial attachment. To determine if the transferrin recycling pathway is involved in Hp colonization of the cell surface, we silenced transferrin receptor expression during infection. This resulted in a reduced ability of Hp to colonize the polarized epithelium. To test whether CagA is important in promoting iron acquisition in vivo, we compared colonization of Hp in iron-replete vs. iron-deficient Mongolian gerbils. While wild type Hp and ΔcagA mutants colonized iron-replete gerbils at similar levels, ΔcagA mutants are markedly impaired in colonizing iron-deficient gerbils. Our study indicates that CagA and VacA act in concert to usurp the polarized process of host cell iron uptake, allowing Hp to use the cell surface as a replicative niche.
机译:幽门螺杆菌(Hp)将CagA效应蛋白注射到宿主上皮细胞中,并诱导生长因子样信号传导,扰乱细胞间连接,并改变宿主细胞极性。即使在不支持自由游动细菌生长的条件下,这也可以使Hp在附着于宿主细胞表面的微菌落中生长。我们假设CagA改变宿主细胞的生理,以使Hp从上皮屏障或跨上皮屏障获得特定的营养。使用极化的上皮模型系统,我们发现等基因的ΔcagA突变体在细胞表面微菌落形成中存在缺陷,但是在根尖培养基中外源添加铁可部分挽救该缺陷,这表明CagA对宿主细胞的作用之一是促进从中获取铁主人。粘附在顶端上皮表面上的Hp以CagA依赖性方式增加转铁蛋白的基底外侧摄取并诱导其转胞作用。 CagA和VacA都对转铁蛋白再循环产生干扰,因为VacA参与了转铁蛋白受体在细菌附着位点的顶端错误定位。为了确定转铁蛋白再循环途径是否参与细胞表面的Hp定殖,我们在感染过程中沉默了转铁蛋白受体的表达。这导致Hp定居极化上皮的能力降低。为了测试CagA在促进体内铁吸收中是否重要,我们比较了铁缺乏和铁缺乏的蒙古沙鼠中Hp的定植。虽然野生型Hp和ΔcagA突变体以相似的水平在富铁沙鼠中定植,但ΔcagA突变体在缺乏铁的沙鼠中显着受损。我们的研究表明,CagA和VacA协同作用,阻止了宿主细胞铁摄取的极化过程,从而使Hp可以利用细胞表面作为复制位。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
代理获取

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