Maternal smoking during pregnancy causes a broad range of effects on the developing lung of the fetus that greatly impairs its long-term function and capacity. The molecular mechanisms underlying these effects are not known. Since both normal lung development and injury/repair utilize common alveolar epithelial-mesenchymal signaling pathways to maintain homeostasis, we propose that in utero nicotine exposure disrupts the critical homeostatic epithelial-mesenchymal paracrine signaling pathways, resulting in the transdifferentiation of alveolar intersitial fibroblasts (AIFs) to myofibroblasts (MYFs). This AIF-to-MYF transdifferentiation is characterized by down regulation of the Peroxisome Proliferator Activated Receptor gamma (PPAR() signaling pathway and up regulation of the Wingless/Int (Wnt) signaling pathway. Although the effects of in utero nicotine exposure on PPAR( signaling have recently been described, there is no information on its effect on the Wnt signaling in the developing lung. We hypothesize that in utero nicotine exposure up-regulates AIF Wnt signaling and that by specific molecular targeting to down regulate the Wnt signaling and/or up regulate the PPAR( signaling, in utero nicotine-induced lung injury can be prevented or even reversed. Using well established in vitro and in vivo models of in utero nicotine-induced lung injury, through real-time RT- PCR, Western analysis, immunohistochemistry, laser capture microdissection, metabolomics, antisense and over expression studies, and in vivo pulmonary function tests, we will determine (1) how in utero nicotine exposure alters lung development, resulting in altered structure and function, and (2) if PPAR( agonists and/or Wnt antagonists, either alone or in combination, can prevent and/or reverse perinatal nicotine exposure-induced alterations in PPAR( and Wnt signaling in lung mesenchyme, and hence lung structure and function. Since the paradigm and models used by us are based on a universal developmental model for the formation, establishment, and homeostasis of the alveolar acinus, the information generated will not be limited only to in utero nicotine-induced injury but is likely to have a much larger applicability to lung development and injury/repair in general. Therefore, by utilizing functional genomics, the studies proposed herein are likely to translate into novel and innovative molecular preventive and therapeutic approaches for all chronic lung diseases in general. PROJECT NARRATIVE: Maternal smoking during pregnancy poses significant risks not only during gestation and immediate newborn period, but may also results in life long medical complications including devastating respiratory problems. The mechanisms underlying the harmful effects of maternal smoking on the lung are complex and incompletely understood. Using novel and innovative concepts, and the state-of-the-art technology, the proposed studies will unravel mechanisms that underlie not only the in utero smoke exposure-induced lung injury, but also almost all other forms of chronic lung diseases.

Agency
National Institute of Health (NIH)
Institute
Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD)
Type
Research Project (R01)
Project #
5R01HD051857-03
Application #
7897882
Study Section
Lung Injury, Repair, and Remodeling Study Section (LIRR)
Program Officer
Ilekis, John V
Project Start
2008-09-30
Project End
2013-07-31
Budget Start
2010-08-01
Budget End
2011-07-31
Support Year
3
Fiscal Year
2010
Total Cost
$280,932
Indirect Cost
Name
La Biomed Research Institute/ Harbor UCLA Medical Center
Department
Type
DUNS #
069926962
City
Torrance
State
CA
Country
United States
Zip Code
90502
Hwang, Jung S; Rehan, Virender K (2018) Recent Advances in Bronchopulmonary Dysplasia: Pathophysiology, Prevention, and Treatment. Lung 196:129-138
Sakurai, Reiko; Lee, Cindy; Shen, Humphrey et al. (2018) A Combination of the Aerosolized PPAR-? Agonist Pioglitazone and a Synthetic Surfactant Protein B Peptide Mimic Prevents Hyperoxia-Induced Neonatal Lung Injury in Rats. Neonatology 113:296-304
Sakurai, Reiko; Liu, Jie; Gong, Ming et al. (2016) Perinatal nicotine exposure induces myogenic differentiation, but not epithelial-mesenchymal transition in rat offspring lung. Pediatr Pulmonol 51:1142-1150
Taylor, Sneha K; Sakurai, Reiko; Sakurai, Tokusho et al. (2016) Inhaled Vitamin D: A Novel Strategy to Enhance Neonatal Lung Maturation. Lung 194:931-943
Cannon, Daniel T; Liu, Jie; Sakurai, Reiko et al. (2016) Impaired Lung Mitochondrial Respiration Following Perinatal Nicotine Exposure in Rats. Lung 194:325-8
Torday, John S; Rehan, Virender K (2016) On the evolution of the pulmonary alveolar lipofibroblast. Exp Cell Res 340:215-9
Ji, Bo; Zhao, Guo-Zhen; Sakurai, Reiko et al. (2016) Effect of Maternal Electroacupuncture on Perinatal Nicotine Exposure-Induced Lung Phenotype in Offspring. Lung 194:535-46
Al Alam, Denise; El Agha, Elie; Sakurai, Reiko et al. (2015) Evidence for the involvement of fibroblast growth factor 10 in lipofibroblast formation during embryonic lung development. Development 142:4139-50
Paek, David S; Sakurai, Reiko; Saraswat, Aditi et al. (2015) Metyrapone alleviates deleterious effects of maternal food restriction on lung development and growth of rat offspring. Reprod Sci 22:207-22
Liu, Jie; Sakurai, Reiko; Rehan, Virender K (2015) PPAR-? agonist rosiglitazone reverses perinatal nicotine exposure-induced asthma in rat offspring. Am J Physiol Lung Cell Mol Physiol 308:L788-96

Showing the most recent 10 out of 41 publications