Fibroblasts are the principal effector cells that mediate tissue remodeling in idiopathic pulmonary fibrosis (IPF) via their capacities for enhanced survival, proliferation, collagen deposition, and myofibroblast differentiation. Although research in the pathogenesis of pulmonary fibrosis has been dominated by studies investigating fibroblast activation signals, evidence indicates that this disorder is also characterized by a relative deficiency in counter-regulatory anti-fibrotic signals. Two such anti-fibrotic signals are the prostanoid prostaglandin E2 (PGE2) and plasminogen activator (PA) activity. Each of these has been shown to be deficient in patients with IPF, and deficiency of each has been established to be pathogenically important in animal models of pulmonary fibrosis. PGE2 is a lipid mediator derived from cyclooxygenase metabolism of the fatty acid arachidonic acid that acts via cell surface G protein-coupled E prostanoid receptors. The PA system is a proteolytic cascade that includes the protease urokinase-type PA (uPA) and its associated inhibitor (plasminogen activator inhibitor-1). Although PGE2 inhibits the activation of all relevant pro-fibrotic cellular phenotypes in lung fibroblasts via intracellular cyclic AMP (cAMP) signaling, the downstream mechanisms by which it does so are incompletely understood. The PA system is recognized to orchestrate fibrinolysis and to modulate cellular adhesion and cellular signaling, but little is known about its direct effects on fibroblasts or their relevant phenotypes. Finally, there is no information about cross-talk between PGE2 and the PA system in lung cells of any kind, including fibroblasts. This project seeks to understand the mechanisms by which both mediators modulate fibroblast function, to characterize the cross-talk between them, and to determine how fibrotic lung injury influences the responses of fibroblasts to each of them. The general hypothesis is that the PGE2 and PA systems up-regulate each other and interact to influence pulmonary fibroblast phenotypes in a manner which favors lung repair over fibrosis. This hypothesis will be tested in fibroblast cell lines and in primary cells isolated from normal and fibrotic murine and human lungs.
Aim 1 will examine the roles of cAMP effectors protein kinase A and guanylate exchange protein activated by cAMP as well as the phosphatase PTEN in mediating PGE2 effects on fibroblast phenotypes.
Aim 2 will determine the mechanisms by which PGE2 and PA activity influence the expression of each other, while the role of each in mediating the actions of the other will be explored in Aim 3.
Aim 4 will compare the effects of PGE2 and PA activity on phenotypes of fibroblasts derived from normal vs. injured mouse lungs and from histologically normal vs. IPF human lungs. The proposed studies will provide novel insights into the regulation of fibroblast activation by these two mediators, and will inform future efforts to target these molecules therapeutically.

Public Health Relevance

The development of a serious condition known as lung scarring (pulmonary fibrosis) is opposed by two substances produced by the body, prostaglandin E2 and urokinase plasminogen activator. This proposal will examine how these two substances act and interact to suppress scarring responses of the key lung cell type known as the fibroblast. These studies will enhance our understanding of how scarring responses are regulated, and may provide insight as to whether these substances could be administered to patients to treat this devastating condition.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
5R01HL094311-04
Application #
8294649
Study Section
Lung Injury, Repair, and Remodeling Study Section (LIRR)
Program Officer
Link, Rebecca P
Project Start
2009-08-11
Project End
2013-05-31
Budget Start
2012-07-01
Budget End
2013-05-31
Support Year
4
Fiscal Year
2012
Total Cost
$375,913
Indirect Cost
$128,413
Name
University of Michigan Ann Arbor
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
073133571
City
Ann Arbor
State
MI
Country
United States
Zip Code
48109
Penke, Loka R; Speth, Jennifer M; Dommeti, Vijaya L et al. (2018) FOXM1 is a critical driver of lung fibroblast activation and fibrogenesis. J Clin Invest 128:2389-2405
Lin, Y-C; Sung, Y K; Jiang, X et al. (2017) Simultaneously Targeting Myofibroblast Contractility and Extracellular Matrix Cross-Linking as a Therapeutic Concept in Airway Fibrosis. Am J Transplant 17:1229-1241
Wettlaufer, Scott H; Penke, L Raghu; Okunishi, Katsuhide et al. (2017) Distinct PKA regulatory subunits mediate PGE2 inhibition of TGF?-1-stimulated collagen I translation and myofibroblast differentiation. Am J Physiol Lung Cell Mol Physiol 313:L722-L731
Schneider, Daniel J; Speth, Jennifer M; Penke, Loka R et al. (2017) Mechanisms and modulation of microvesicle uptake in a model of alveolar cell communication. J Biol Chem 292:20897-20910
Gu, Hongmei; Fisher, Amanda J; Mickler, Elizabeth A et al. (2016) Contribution of the anaphylatoxin receptors, C3aR and C5aR, to the pathogenesis of pulmonary fibrosis. FASEB J 30:2336-50
Wettlaufer, Scott H; Scott, Jacob P; McEachin, Richard C et al. (2016) Reversal of the Transcriptome by Prostaglandin E2 during Myofibroblast Dedifferentiation. Am J Respir Cell Mol Biol 54:114-27
Zaslona, Zbigniew; Peters-Golden, Marc (2015) Prostanoids in Asthma and COPD: Actions, Dysregulation, and Therapeutic Opportunities. Chest 148:1300-1306
Huang, Steven K; Scruggs, Anne M; McEachin, Richard C et al. (2014) Lung fibroblasts from patients with idiopathic pulmonary fibrosis exhibit genome-wide differences in DNA methylation compared to fibroblasts from nonfibrotic lung. PLoS One 9:e107055
Okunishi, Katsuhide; DeGraaf, Angela J; Zas?ona, Zbigniew et al. (2014) Inhibition of protein translation as a novel mechanism for prostaglandin E2 regulation of cell functions. FASEB J 28:56-66
Degraaf, Angela Juliette; Zas?ona, Zbigniew; Bourdonnay, Emilie et al. (2014) Prostaglandin E2 reduces Toll-like receptor 4 expression in alveolar macrophages by inhibition of translation. Am J Respir Cell Mol Biol 51:242-50

Showing the most recent 10 out of 24 publications