α‐galactosylceramide generates lung regulatory T cells through the activated natural killer T cells in mice

  • Qianhui Chen
    Department of Respiratory Medicine Renmin Hospital of Wuhan University Wuhan China
  • Xuxue Guo
    Department of Respiratory Medicine Renmin Hospital of Wuhan University Wuhan China
  • Nishan Deng
    Department of Respiratory Medicine Renmin Hospital of Wuhan University Wuhan China
  • Linlin Liu
    Department of Respiratory Medicine Renmin Hospital of Wuhan University Wuhan China
  • Shuo Chen
    Department of Respiratory Medicine Renmin Hospital of Wuhan University Wuhan China
  • Ailing Wang
    Nursing Department Wuhan University School of Health Sciences Wuhan China
  • Ruiyun Li
    Department of Respiratory Medicine Renmin Hospital of Wuhan University Wuhan China
  • Yi Huang
    Department of Respiratory Medicine Renmin Hospital of Wuhan University Wuhan China
  • Xuhong Ding
    Department of Respiratory Medicine Renmin Hospital of Wuhan University Wuhan China
  • Hongying Yu
    Department of Respiratory Medicine Renmin Hospital of Wuhan University Wuhan China
  • Suping Hu
    Department of Respiratory Medicine Renmin Hospital of Wuhan University Wuhan China
  • Hanxiang Nie
    Department of Respiratory Medicine Renmin Hospital of Wuhan University Wuhan China

抄録

<jats:title>Abstract</jats:title><jats:p>Our previous study showed that intraperitoneal injection of α‐galactosylceramide (α‐GalCer) has the ability to activate lung <jats:styled-content style="fixed-case">iNKT</jats:styled-content> cells, but α‐GalCer‐activated <jats:styled-content style="fixed-case">iNKT</jats:styled-content> cells do not result in airway inflammation in wild‐type (<jats:styled-content style="fixed-case">WT</jats:styled-content>) mice. Many studies showed that <jats:styled-content style="fixed-case">iNKT</jats:styled-content> cells had the capacity to induce Treg cells, which gave rise to peripheral tolerance. Therefore, we examined the influence of intraperitoneal administration of α‐GalCer on the expansion and suppressive activity of lung Treg cells using <jats:styled-content style="fixed-case">iNKT</jats:styled-content> cell‐knockout mice and co‐culture experiments in vitro. We also compared airway inflammation and airway hyperresponsiveness (<jats:styled-content style="fixed-case">AHR</jats:styled-content>) after α‐GalCer administration in specific anti‐<jats:styled-content style="fixed-case">CD</jats:styled-content>25 <jats:styled-content style="fixed-case">mA</jats:styled-content>b‐treated mice. Our data showed that intraperitoneal injection of α‐GalCer could promote the expansion of lung Treg cells in <jats:styled-content style="fixed-case">WT</jats:styled-content> mice, but not in <jats:styled-content style="fixed-case">iNKT</jats:styled-content> cell‐knockout mice. However, α‐GalCer administration could not boost suppressive activity of Treg cells in <jats:styled-content style="fixed-case">WT</jats:styled-content> mice and <jats:styled-content style="fixed-case">iNKT</jats:styled-content> cell‐knockout mice. Interestingly, functional inactivation of Treg cells could induce airway inflammation and <jats:styled-content style="fixed-case">AHR</jats:styled-content> in <jats:styled-content style="fixed-case">WT</jats:styled-content> mice treated with α‐GalCer. Furthermore, α‐GalCer administration could enhance <jats:styled-content style="fixed-case">iNKT</jats:styled-content> cells to secrete <jats:styled-content style="fixed-case">IL</jats:styled-content>‐2, and neutralization of <jats:styled-content style="fixed-case">IL</jats:styled-content>‐2 reduced the expansion of Treg cells in vivo and in vitro. Thus, intraperitoneal administration of α‐GalCer can induce the generation of lung Treg cells in mice through the release of <jats:styled-content style="fixed-case">IL</jats:styled-content>‐2 by the activated <jats:styled-content style="fixed-case">iNKT</jats:styled-content> cells.</jats:p>

収録刊行物

被引用文献 (1)*注記

もっと見る

問題の指摘

ページトップへ