The early stages of respiratory system development including branching morphogenesis and cell lineage commitment have been studied extensively and this has led to a dramatic increase in our understanding of how the trachea and lungs are specified and how endodermal lineages commit to different epithelial cell fates. However, less is known about the late stages of development and maturation, which are required to form the air-vascular interface important for gas exchange in the postnatal lung. These later steps of lung development involve dramatic changes in cellular differentiation and morphology, resulting in the dilation and expansion of the distal saccules and eventual septation of these saccules into alveoli to increase surface area for efficient gas exchange. The importance of these steps is highlighted by defects in these morphological processes resulting in poorly understood syndromes including bronchopulmonary dysplasia, which can lead to severe neonatal distress or death. Despite the importance of lung sacculation and alveologenesis, the molecular pathways involved and the cellular remodeling that is required for these steps remains poorly understood. We have previously shown that the class I histone deacetylases 1 and 2 (HDAC1 and -2) are essential for development of the proximal Sox2+ lung endoderm progenitors, resulting in the loss of proximal airway epithelial differentiation. We now show that the class I HDAC, HDAC3, regulates late lung epithelial maturation and cellular remodeling essential for sacculation and early alveologenesis. Our preliminary data suggest that HDAC3 regulates sacculation and early alveologenesis through repression of a miRNA program including the miR17-92 cluster. De- repression of miR17-92 in HDAC3 deficient lung epithelium leads to inhibition of Tgf-? signaling as miR17-92 targets multiple components of the Tgf-? pathway including Tgf-?2, Tgf-?R1, Tgf-?R2. The resultant decrease in Tgf-? signaling activity leads to a failure of AT1 cells to spread and remodel, causing a lack of saccule/alveolar airspace expansion and neonatal death. Interestingly, these defects are not associated with an observed change in AT1 cell specification or commitment. Preliminary data also shows that HDAC3 regulates the alveolar epithelial response to injury in the adult lung. These findings will have a broad impact on our understanding of the epigenetic mechanisms and cellular remodeling required for proper late lung development as well as postnatal repair and regeneration.

Public Health Relevance

This proposal will examine the pathways that regulate late lung development. The information gained will have a direct impact on our understanding of the cellular changes that occur just prior to birth and the importance of these processes in generating a functional lung. While these late stages of lung development and maturation are critical for neonatal health, little is understood about the molecular pathways that are important for their successful execution. Our studies will provide crucial information to better understand diseases of the neonate including bronchopulmonary dysplasia.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
5R01HL132999-04
Application #
9751372
Study Section
Lung Injury, Repair, and Remodeling Study Section (LIRR)
Program Officer
Lin, Sara
Project Start
2016-09-01
Project End
2020-06-30
Budget Start
2019-07-01
Budget End
2020-06-30
Support Year
4
Fiscal Year
2019
Total Cost
Indirect Cost
Name
University of Pennsylvania
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
042250712
City
Philadelphia
State
PA
Country
United States
Zip Code
19104
Zacharias, William J; Frank, David B; Zepp, Jarod A et al. (2018) Regeneration of the lung alveolus by an evolutionarily conserved epithelial progenitor. Nature 555:251-255
Herriges, Michael J; Tischfield, David J; Cui, Zheng et al. (2017) The NANCI-Nkx2.1 gene duplex buffers Nkx2.1 expression to maintain lung development and homeostasis. Genes Dev 31:889-903
Liu, Ying; Conlon, Donna M; Bi, Xin et al. (2017) Lack of MTTP Activity in Pluripotent Stem Cell-Derived Hepatocytes and Cardiomyocytes Abolishes apoB Secretion and Increases Cell Stress. Cell Rep 19:1456-1466
Zepp, Jarod A; Zacharias, William J; Frank, David B et al. (2017) Distinct Mesenchymal Lineages and Niches Promote Epithelial Self-Renewal and Myofibrogenesis in the Lung. Cell 170:1134-1148.e10
Stabler, Collin T; Morrisey, Edward E (2017) Developmental pathways in lung regeneration. Cell Tissue Res 367:677-685
Frank, David B; Peng, Tien; Zepp, Jarod A et al. (2016) Emergence of a Wave of Wnt Signaling that Regulates Lung Alveologenesis by Controlling Epithelial Self-Renewal and Differentiation. Cell Rep 17:2312-2325