Acute heart transplant (HTx) rejection is typically averted by available immunosuppressants, which control recipient CD4+ and CD8+ T cell responses to alloantigens presented by the HTx. Unfortunately, these drugs are unable to prevent chronic rejection, -an immune-driven process of pathogenic fibrotic remodeling of the myocardium and vasculature. Chronic rejection is a significant clinical PROBLEM and leads to the dysfunction and loss the majority of HTx in a little over ten years post transplantation. Approaches regulating chronic rejection-promoting immune responses AND shaping HTx repair are needed to solve this problem. We have discovered a subset of Treg that express ST2, the receptor for interleukin(IL)-33. Our preliminary data suggest that ST2+Treg respond to IL-33 with mechanisms controlling local inflammation and supporting tissue repair. IL-33 is expressed by cells of the HTx and is released in a functional form during tissue damage. In early studies we revealed that IL-33 administration post MHC-mismatched heart transplantation expanded Treg that tripled graft survival in the absence of immunosuppression. Our recent comparisons of ST2+ and ST2- Treg found that, while both are able to control T cell mediated acute HTx rejection, IL-33 stimulation of ST2+Treg is critical for prevention of chronic rejection-associated immune infiltration and myocardial fibrosis. These data support our CENTRAL HYPOTHESIS that ST2+Treg are important for preventing chronic HTx rejection due, not only to their capacity to suppress AlloAg-reactive T cells, but also their ability to facilitate tissue repair and regulat myeloid cells in response to IL-33. The OBJECTIVE of this application is to identify exploitable mechanisms by which ST2+Treg and IL-33 control inflammation and mediate cardiac tissue repair after transplantation. To that end, we will perform HTx and cardiac injury studies utilizing transgenic mice that allow specific gene targeting in Treg and donor and recipient mice lacking IL-33. Our data also suggest that chronic HTx rejection may arises as a result of IL-12 cytokines that suppress ST2+Treg and favor IL-33 stimulation of deleterious CD8+T cell responses. Thus, we will also take advantage of HTx models where ST2 is absent on CD8+ T cells or Treg and IL-33 and IL-12 cytokines targeted in host cells. The requirement of Treg suppression of CD4+ and CD8+ T cell responses for maintenance of peripheral tolerance and induction of Tx tolerance is well established. The concept that local or systemic IL-33 stimulates the reparative capacity of ST2+Treg during tissue injury is novel and untested. The proposed studies are INNOVATIVE because they offer a new way of thinking about the role Treg play in coordinating the cytokine networks controlling HTx outcomes. These studies are also SIGNIFICANT, as they will identify targetable mechanisms controlling the reparative capacity of ST2+Treg in transplantation.

Public Health Relevance

Chronic heart transplant rejection has no present effective treatment and leads to gradual allograft failure due to immune-mediated fibrotic and vascular remodeling. Interleukin-33 (IL-33), a molecule released during heart damage and rejection, supports regulatory and potential reparative functions in immune cells that, if properly harnessed, could counter chronic rejection. This project will provide precise understanding of the mechanisms controlling IL-33-related immune functions after heart transplantation and may lead to new therapies able to prevent or resolve chronic rejection-associated heart transplant damage.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
5R01HL122489-03
Application #
9266808
Study Section
Transplantation, Tolerance, and Tumor Immunology Study Section (TTT)
Program Officer
Desvigne-Nickens, Patrice
Project Start
2015-07-01
Project End
2020-04-30
Budget Start
2017-05-01
Budget End
2018-04-30
Support Year
3
Fiscal Year
2017
Total Cost
Indirect Cost
Name
University of Pittsburgh
Department
Surgery
Type
Schools of Medicine
DUNS #
004514360
City
Pittsburgh
State
PA
Country
United States
Zip Code
15213
Chang, Jing; Xia, Yuefeng; Wasserloos, Karla et al. (2017) Cyclic stretch induced IL-33 production through HMGB1/TLR-4 signaling pathway in murine respiratory epithelial cells. PLoS One 12:e0184770
Yazdani, Hamza O; Chen, Hui-Wei; Tohme, Samer et al. (2017) IL-33 exacerbates liver sterile inflammation by amplifying neutrophil extracellular trap formation. J Hepatol :
Xu, Hui; Turnquist, Heth R; Hoffman, Rosemary et al. (2017) Role of the IL-33-ST2 axis in sepsis. Mil Med Res 4:3
Matta, B M; Reichenbach, D K; Blazar, B R et al. (2017) Alarmins and Their Receptors as Modulators and Indicators of Alloimmune Responses. Am J Transplant 17:320-327
Xu, Jing; Guardado, Jesse; Hoffman, Rosemary et al. (2017) IL33-mediated ILC2 activation and neutrophil IL5 production in the lung response after severe trauma: A reverse translation study from a human cohort to a mouse trauma model. PLoS Med 14:e1002365
Tjota, Melissa Y; Camacho, Daniel F; Turnquist, Heth R et al. (2017) IL-33 Drives Monocyte Recruitment to Lung Interstitium through Chemokine Upregulation. Immunohorizons 1:101-108
Liew, Foo Yew; Girard, Jean-Philippe; Turnquist, Heth Roderick (2016) Interleukin-33 in health and disease. Nat Rev Immunol 16:676-689
Liu, Quan; Turnquist, Heth R (2016) Controlling the burn and fueling the fire: defining the role for the alarmin interleukin-33 in alloimmunity. Curr Opin Organ Transplant 21:45-52
Matta, Benjamin M; Reichenbach, Dawn K; Zhang, Xiaoli et al. (2016) Peri-alloHCT IL-33 administration expands recipient T-regulatory cells that protect mice against acute GVHD. Blood 128:427-39