Login / Signup

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

David S WheelerKeizo MisumiNatalie M WalkerRagini VittalMichael P CombsYoshiro AokiRussell R BraeuerVibha N Lama
Published in: American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons (2021)
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.
Keyphrases
  • oxidative stress
  • endothelial cells
  • cell proliferation
  • bone marrow
  • mesenchymal stem cells
  • cell therapy
  • patient safety
  • peripheral blood
  • kidney transplantation
  • idiopathic pulmonary fibrosis
  • protein kinase