4.6 Article

Interleukin 6 trans-signaling is a critical driver of lung allograft fibrosis

期刊

AMERICAN JOURNAL OF TRANSPLANTATION
卷 21, 期 7, 页码 2360-2371

出版社

WILEY
DOI: 10.1111/ajt.16417

关键词

animal models; murine; basic (laboratory) research; science; bronchiolitis obliterans (BOS); cellular biology; cytokines; cytokine receptors; fibrosis; lung transplantation; pulmonology; translational research; science

资金

  1. National Heart, Lung, and Blood Institute [HL007749-26, HL094622, HL118017]
  2. Brian and Mary Campbell and Elizabeth Campbell-Carr research gift fund
  3. Cystic Fibrosis Foundation [LAMA16XX0]

向作者/读者索取更多资源

Histopathologic examination of lungs afflicted by chronic lung allograft dysfunction consistently shows both mononuclear cell inflammation and mesenchymal cell fibroproliferation, with interleukin 6 (IL-6) potentially playing a critical role in mediating the communication between these cells and contributing to the pathogenesis of CLAD.
Histopathologic examination of lungs afflicted by chronic lung allograft dysfunction (CLAD) consistently shows both mononuclear cell (MNC) inflammation and mesenchymal cell (MC) fibroproliferation. We hypothesize that interleukin 6 (IL-6) trans-signaling may be a critical mediator of MNC-MC crosstalk and necessary for the pathogenesis of CLAD. Bronchoalveolar lavage (BAL) fluid obtained after the diagnosis of CLAD has approximately twofold higher IL-6 and soluble IL-6 receptor (sIL-6R) levels compared to matched pre-CLAD samples. Human BAL-derived MCs do not respond to treatment with IL-6 alone but have rapid and prolonged JAK2-mediated STAT3 Tyr705 phosphorylation when exposed to the combination of IL-6 and sIL-6R. STAT3 phosphorylation within MCs upregulates numerous genes causing increased invasion and fibrotic differentiation. MNC, a key source of both IL-6 and sIL-6R, produce minimal amounts of these proteins at baseline but significantly upregulate production when cocultured with MCs. Finally, the use of an IL-6 deficient recipient in a murine orthotopic transplant model of CLAD reduces allograft fibrosis by over 50%. Taken together these results support a mechanism where infiltrating MNCs are stimulated by resident MCs to release large quantities of IL-6 and sIL-6R which then feedback onto the MCs to increase invasion and fibrotic differentiation.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.6
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据