The broad long-term objective of our research is to reduce the incidence of ventilator-induced lung injury (VILI), characterized by pneumothorax, progressive impairment in pulmonary mechanics, alveolar cell dysfunction, and profound changes in lung fluid balance and blood-gas barrier permeability. In the previous funding periods we identified cyclic and tonic deformation magnitudes above which we measured acute alterations in epithelial cell viability and barrier properties. Moreover, the injurious deformation magnitudes we applied in vitro correlated well with tidal volumes and positive end-expiratory pressures used clinically that have been associated with clinical morbidity in ventilator induced lung injury (VILI). These findings underscore the potential relevance of using experimental models with carefully controlled conditions to identify the mechanisms responsible for stretch-induced disruption of the alveolar epithelial barrier, and to identify opportunities for injury intervention. The in vitro model for VILI that we established during the first 6 years of funding is limited to healthy rat alveolar epithelial cell monolayers exposed to 1 hr periods of deformation. In this competitive renewal our major objective is to determine specific stretch-induced mechanical and molecular signals that modulate alveolar epithelial permeability during clinically relevant conditions - including chronic continuous cycling and ARDS.
In Aim 1 we hypothesize that the environmental stressors of sepsis, high cycling rates and long stretch durations each compromise epithelial monolayer barrier properties in both primary cell monolayers and intact lungs.
In Aim 2 we hypothesize that stretch induces barrier alterations by activation of mitogen- activated protein kinases, which initiate nuclear factor kappa B dependent cytokine expression and release to ultimately increase monolayer permeability.
In Aim 3 we will test the hypotheses that acute (<1 hr) stretch reorganizes the cytoskeleton, which increases monolayer permeability directly by dissociating tight junctional (TJ) proteins from actin, and indirectly by initiating kinase-mediated TJ phosphorylation and TJ complex disassembly. Thus, we build on the foundation established during previous funding periods - both to deepen our understanding of basic mechanisms and to enhance the clinical translation of our findings - and progress towards our goal of reducing acute VILI due to epithelial over-distension. ? ? ?

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
5R01HL057204-09
Application #
7409603
Study Section
Respiratory Integrative Biology and Translational Research Study Section (RIBT)
Program Officer
Harabin, Andrea L
Project Start
1997-07-07
Project End
2011-03-31
Budget Start
2008-04-01
Budget End
2009-03-31
Support Year
9
Fiscal Year
2008
Total Cost
$417,426
Indirect Cost
Name
University of Pennsylvania
Department
Biomedical Engineering
Type
Schools of Engineering
DUNS #
042250712
City
Philadelphia
State
PA
Country
United States
Zip Code
19104
Song, Min Jae; Davidovich, Nurit; Lawrence, Gladys G et al. (2016) Superoxide mediates tight junction complex dissociation in cyclically stretched lung slices. J Biomech 49:1330-1335
Song, M J; Davis, C I; Lawrence, G G et al. (2016) Local influence of cell viability on stretch-induced permeability of alveolar epithelial cell monolayers. Cell Mol Bioeng 9:65-72
Yehya, Nadir; Xin, Yi; Oquendo, Yousi et al. (2015) Cecal ligation and puncture accelerates development of ventilator-induced lung injury. Am J Physiol Lung Cell Mol Physiol 308:L443-51
Davidovich, N; Huang, J; Margulies, S S (2013) Reproducible uniform equibiaxial stretch of precision-cut lung slices. Am J Physiol Lung Cell Mol Physiol 304:L210-20
Dipaolo, Brian C; Davidovich, Nurit; Kazanietz, Marcelo G et al. (2013) Rac1 pathway mediates stretch response in pulmonary alveolar epithelial cells. Am J Physiol Lung Cell Mol Physiol 305:L141-53
Davidovich, Nurit; DiPaolo, Brian C; Lawrence, Gladys G et al. (2013) Cyclic stretch-induced oxidative stress increases pulmonary alveolar epithelial permeability. Am J Respir Cell Mol Biol 49:156-64
Davidovich, N; Chhour, P; Margulies, S S (2013) Uses of Remnant Human Lung Tissue for Mechanical Stretch Studies. Cell Mol Bioeng 6:175-182
Yehya, Nadir; Yerrapureddy, Adi; Tobias, John et al. (2012) MicroRNA modulate alveolar epithelial response to cyclic stretch. BMC Genomics 13:154
Cohen, Taylor S; DiPaolo, Brian C; Lawrence, Gladys Gray et al. (2012) Sepsis enhances epithelial permeability with stretch in an actin dependent manner. PLoS One 7:e38748
DiPaolo, Brian C; Margulies, Susan S (2012) Rho kinase signaling pathways during stretch in primary alveolar epithelia. Am J Physiol Lung Cell Mol Physiol 302:L992-1002

Showing the most recent 10 out of 29 publications