Idiopathic pulmonary fibrosis (IPF) is an incurable fatal disease with increasing incidence and mortality. Despite recent coordinated attempts to rapidly translate findings which have been validated in vitro and in preclinical models into biologically promising interventions, there have been no major therapeutic breakthroughs. A final common pathway for the destructive remodeling which characterizes fibrosis is the apoptosis-resistant myofibroblast, which both creates and responds to an altered mediator and matrix microenvironment, thus perpetuating fibrogenesis. Halting the development of the myofibroblast phenotype, or reversing it once established, offers the best hope of successfully treating IPF. The cell surface glycoprotein Thy-1, a known tumor suppressor which acts as a context-dependent regulator of cell phenotype, is silenced in the myofibroblasts within fibroblastic foci, the signature histopathologic lesions in IPF, the presence of which portends a poor prognosis. Absence of Thy-1 in cultured lung fibroblasts promotes proliferation, cytokine and growth factor expression and responsiveness, migration, myofibroblastic differentiation, and resistance to apoptosis, all reversible upon re-expression of Thy-1. Thy-1 interacts with alpha v integrins and syndecan 4 at the cell surface, modulating cell-cell and cell-matrix interactions and mechanical coupling, to inhibit TGF-beta activation and myofibroblastic differentiation. The effects of Thy-1 on the myofibroblast phenotype are broad, including: decreased expression of a number of muscle-specific proteins and myogenic transcription factors, inhibition of contractility, and promotion of apoptosis. Furthermore, some of the functions of Thy-1 can be recapitulated by the administration of soluble Thy-1, and preliminary data demonstrate that soluble Thy-1 reverses established fibrosis in a mouse model, indicating that Thy-1 itself can be used or modified for therapeutic benefit. Taken together, this knowledge leads us to the overall hypothesis that Thy-1-mediated signaling broadly suppresses myofibroblastic transformation and promotes apoptosis, restoring homeostatic function in lung fibroblasts. Improved understanding of the molecular mechanisms involved will uncover novel strategies for reversing the pathogenic myofibroblast phenotype in IPF and other fibrotic disorders. In vitro and in vivo models based on heterogeneity and targeted disruption of Thy-1 offer excellent opportunities for defining myofibroblast-targeting strategies, as outlined in the following specific aims:
Aim 1 : To define the mechanisms by which Thy-1- inhibits myofibroblastic differentiation of lung fibroblasts;
Aim 2 : To define the mechanisms by which Thy-1 promotes myofibroblast apoptosis;
and Aim 3 : To harness Thy-1-modulated signaling to reverse the apoptosis-resistant myofibroblast phenotype in vivo. Significance: Because Thy-1 modulates multiple signaling pathways critical to fibrogenesis, capitalizing on its function is more attractive than targeting a single pathway, which has been shown to be an ineffective approach in IPF. The proposed studies will thus define mechanisms by which Thy-1 suppresses profibrotic differentiation of lung fibroblasts, and translate that knowledge into innovative therapeutic interventions for fibrotic lung disease.

Public Health Relevance

Targeting individual signaling pathways has failed time and again in treating idiopathic pulmonary fibrosis (IPF), a fatal and progressive lung disease which is increasing in incidence. The differentiation of a structural cell in the lung known as the fibroblast into the aggressive scar-forming myofibroblast, which resists programmed cell death (apoptosis) and gradually replaces the lung with scar tissue, is central to the pathogenesis of IPF. Restoring or replacing the function of Thy-1, a fibroblast molecule that globally suppresses the apoptosis-resistant myofibroblast phenotype, offers the best hope for treating this devastating disease.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
3R01HL111169-02S1
Application #
8677065
Study Section
Lung Injury, Repair, and Remodeling Study Section (LIRR)
Program Officer
Eu, Jerry Pc
Project Start
2012-08-01
Project End
2016-05-31
Budget Start
2013-06-01
Budget End
2014-05-31
Support Year
2
Fiscal Year
2013
Total Cost
$69,750
Indirect Cost
$24,750
Name
University of California San Diego
Department
Pediatrics
Type
Schools of Medicine
DUNS #
804355790
City
La Jolla
State
CA
Country
United States
Zip Code
92093
Fiore, Vincent F; Wong, Simon S; Tran, Coleen et al. (2018) ?v?3 Integrin drives fibroblast contraction and strain stiffening of soft provisional matrix during progressive fibrosis. JCI Insight 3:
Duong, Thu Elizabeth; Hagood, James S (2018) Epigenetic Regulation of Myofibroblast Phenotypes in Fibrosis. Curr Pathobiol Rep 6:79-96
Liu, Xiaoqiu; Wong, Simon S; Taype, Carmen A et al. (2017) Thy-1 interaction with Fas in lipid rafts regulates fibroblast apoptosis and lung injury resolution. Lab Invest 97:256-267
Glasser, Stephan W; Hagood, James S; Wong, Simon et al. (2016) Mechanisms of Lung Fibrosis Resolution. Am J Pathol 186:1066-77
Fiore, Vincent F; Strane, Patrick W; Bryksin, Anton V et al. (2015) Conformational coupling of integrin and Thy-1 regulates Fyn priming and fibroblast mechanotransduction. J Cell Biol 211:173-90
Hagood, James S (2014) Beyond the genome: epigenetic mechanisms in lung remodeling. Physiology (Bethesda) 29:177-85
Ramírez, Gustavo; Hagood, James S; Sanders, Yan et al. (2011) Absence of Thy-1 results in TGF-? induced MMP-9 expression and confers a profibrotic phenotype to human lung fibroblasts. Lab Invest 91:1206-18
Kis, Kornelia; Liu, Xiaoqiu; Hagood, James S (2011) Myofibroblast differentiation and survival in fibrotic disease. Expert Rev Mol Med 13:e27