In utero nicotine exposure affects lung growth and differentiation by altering specific physiologic molecular signaling pathways that are necessary for fetal lung development, resulting in the offspring's predisposition to childhood asthma. We now have preliminary evidence that these alterations in the structure and function of the lung caused by nicotine exposure during pregnancy can be passed from one generation to the next, i.e., from generation 1 (G1) to G2 and G3, etc. We have previously shown that nicotine alters the normal differentiation of the mesenchymal cells in the developing fetal lung by stimulating the Wnt pathway, causing the myogenic phenotype, consistent with asthma in the offspring. Moreover, we have found that peroxisome proliferator- activated receptor gamma (PPAR?) agonists can inhibit or reverse this effect of nicotine. Armed with this knowledge of nicotine's effect on asthma in G1 offspring, we will now determine its transgenerational effect and whether this effect is determined by nicotine-induced epigenetic changes in the gonads.
In Specific Aim 1 A, we will determine the transgenerational development of asthma in G2 and G3 offspring of G1 rat offspring exposed to nicotine in utero in a gender-specific manner. We will determine if the transgenerational increase in the risk of asthma following in utero nicotine exposure is greater in G2 males than in females.
In Specific Aim 1 B, we will determine whether exposure of G2 offspring to nicotine in utero further exacerbates the transgenerational asthma risk in G3.
In Specific Aim 2, we will elucidate the effects of nicotine on epigenetic mechanisms in the lung and gonads as the putative basis for the transgenerational effect of nicotine on asthma.
In Specific Aim 2 A, we will determine the epigenetic effects of nicotine on the methylation and acetylation of DNA in the G1 offspring lungs and gonads.
In Specific Aim 2 B, we will determine if the PPAR? agonist rosiglitazone will inhibit 1) the epigenetic changes in the lung and gonads, and thus 2) prevent the transgenerational effect of nicotine on asthma. The concept put forward in this proposal is totally novel and innovative, and it addresses the fundamental mechanism (s) explaining the detrimental effects of maternal smoking not only on the exposed offspring, but also on the many generations that follow. Using this comprehensive cell-molecular-epigenetic approach to understand the transgenerational effects of smoking on the prevalence of asthma will lead to effective and targeted interventions and prevention of this disease, which at present is a major public health challenge.

Public Health Relevance

Childhood asthma is a major public health problem worldwide with maternal smoking during pregnancy as a significant contributor to this ever growing epidemic. In addition, there is emerging evidence for transgenerational transmission of asthma risk following exposure to maternal smoke during pregnancy. The studies proposed in this application aim to examine the molecular basis underlying the increased transgenerational asthma risk following in utero exposure to maternal smoke and are likely to provide pivotal molecular data that can have significant impact on understanding the pathogenesis of, and devising new treatments for, childhood asthma.

Agency
National Institute of Health (NIH)
Institute
Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD)
Type
Exploratory/Developmental Grants (R21)
Project #
5R21HD071731-02
Application #
8502717
Study Section
Special Emphasis Panel (ZRG1-CVRS-G (03))
Program Officer
Raju, Tonse N
Project Start
2012-07-05
Project End
2014-06-30
Budget Start
2013-07-01
Budget End
2014-06-30
Support Year
2
Fiscal Year
2013
Total Cost
$159,299
Indirect Cost
$40,674
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
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
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
Gong, Ming; Antony, Sahaya; Sakurai, Reiko et al. (2016) Bone marrow mesenchymal stem cells of the intrauterine growth-restricted rat offspring exhibit enhanced adipogenic phenotype. Int J Obes (Lond) 40:1768-1775
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
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

Showing the most recent 10 out of 19 publications