Progressive fibrosis is a hallmark of interstitial lung diseases such as idiopathic pulmonary fibrosis. Fibrosis stiffens the lung parenchyma. Preliminary data provided in this proposal demonstrate that normal lung parenchymal tissue is more compliant than previously predicted, and that highly localized increases in the stiffness of fibrotic lesions are much greater than previously recognized. Fibroblasts grown on substrates as stiff as fibrotic lesions engage in rapid proliferation and abundant matrix synthesis;in marked contrast these behaviors are largely suppressed on substrates as compliant as normal lung tissue. The central hypothesis that we pose based on these data is that the mechanical environment present in lung fibrosis triggers a """"""""fibrogenesis program"""""""" in resident fibroblasts that promotes feedback amplification of the disease. We propose four specific aims: (1) quantify the stiffness of the parenchyma in normal lung tissue and developing and established fibrotic lesions;(2) test whether the transition in matrix stiffness from normal to fibrotic levels is a necessary precondition for lung fibroblast proliferation and fibrogenic activation;(3) test the role played by cytoskeletal dynamics and serum response factor activation in driving stiffness-dependent fibroblast biology;and (4) identify key transcription factors coordinating fibroblast transitions between quiescent and fibrogenic states when transferred between compliant and stiff matrices.
These aims will be carried out in novel 2D and 3D models that allow clear delineation of the effects of lung matrix stiffness on key fibrogenic behaviors of lung fibroblasts. Throughout the experimental plan we will examine the interplay between matrix stiffness and the soluble environment present in fibrosis as it impacts on fibroblast biology. This research will generate novel insights into the molecular mechanisms of stiffness-dependent fibroblast activation in fibrotic lungs, and identify critical regulators of fibrogenesis suitable for therapeutic targeting.

Public Health Relevance

Lung fibrosis stiffens affected tissue. The experiments proposed here will test whether pathophysiological changes in stiffness promote fibrosis by stimulating lung fibroblasts. Understanding the role mechanical factors play in fibroblast activation could lead to new strategies to treat fibrosis.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
5R01HL092961-02
Application #
7907679
Study Section
Lung Injury, Repair, and Remodeling Study Section (LIRR)
Program Officer
Reynolds, Herbert Y
Project Start
2009-08-06
Project End
2013-07-31
Budget Start
2010-08-01
Budget End
2011-07-31
Support Year
2
Fiscal Year
2010
Total Cost
$413,123
Indirect Cost
Name
Harvard University
Department
Public Health & Prev Medicine
Type
Schools of Public Health
DUNS #
149617367
City
Boston
State
MA
Country
United States
Zip Code
02115
Dodi, Amos E; Ajayi, Iyabode O; Chang, Christine et al. (2018) Regulation of fibroblast Fas expression by soluble and mechanical pro-fibrotic stimuli. Respir Res 19:91
Tschumperlin, Daniel J; Ligresti, Giovanni; Hilscher, Moira B et al. (2018) Mechanosensing and fibrosis. J Clin Invest 128:74-84
Oh, Raymond S; Haak, Andrew J; Smith, Karry M J et al. (2018) RNAi screening identifies a mechanosensitive ROCK-JAK2-STAT3 network central to myofibroblast activation. J Cell Sci 131:
Haak, Andrew J; Tan, Qi; Tschumperlin, Daniel J (2018) Matrix biomechanics and dynamics in pulmonary fibrosis. Matrix Biol 73:64-76
Jorgenson, Amy J; Choi, Kyoung Moo; Sicard, Delphine et al. (2017) TAZ activation drives fibroblast spheroid growth, expression of profibrotic paracrine signals, and context-dependent ECM gene expression. Am J Physiol Cell Physiol 312:C277-C285
Sicard, Delphine; Fredenburgh, Laura E; Tschumperlin, Daniel J (2017) Measured pulmonary arterial tissue stiffness is highly sensitive to AFM indenter dimensions. J Mech Behav Biomed Mater 74:118-127
Tjin, Gavin; White, Eric S; Faiz, Alen et al. (2017) Lysyl oxidases regulate fibrillar collagen remodelling in idiopathic pulmonary fibrosis. Dis Model Mech 10:1301-1312
Dieffenbach, Paul B; Haeger, Christina Mallarino; Coronata, Anna Maria F et al. (2017) Arterial stiffness induces remodeling phenotypes in pulmonary artery smooth muscle cells via YAP/TAZ-mediated repression of cyclooxygenase-2. Am J Physiol Lung Cell Mol Physiol 313:L628-L647
Tan, Qi; Choi, Kyoung Moo; Sicard, Delphine et al. (2017) Human airway organoid engineering as a step toward lung regeneration and disease modeling. Biomaterials 113:118-132
Haak, Andrew J; Girtman, Megan A; Ali, Mohamed F et al. (2017) Phenylpyrrolidine structural mimics of pirfenidone lacking antifibrotic activity: A new tool for mechanism of action studies. Eur J Pharmacol 811:87-92

Showing the most recent 10 out of 26 publications