Unrepaired DNA damage is a prominent feature of vascular cells in diseases that include pulmonary arterial hypertension (PAH). During the current grant cycle, we identified a central role for PPAR?1 in DNA damage sensing and repair that was perturbed in PAH patient pulmonary arterial endothelial cells (PAEC). We observed that PPAR? forms a complex with the DNA damage sensor Mre11, RAD51 and NBS1 (MRN) and with the ubiquitin ligase UBR5. We went on to show that this interaction is necessary for the degradation of ATM interacting protein (ATMIN), thereby permitting phosphorylation of ATM and initiation of the DNA repair process. In PAH PAEC, high interleukin (IL)6 levels are directly related to the phosphorylation of PPAR? at serine 245 and its impaired interaction with MRN and UBR5, causing elevated ATMIN and impaired DNA damage sensing via phosphoATM.
In Specific Aim 1, we extend these observations by determining whether high endogenous IL6 levels phosphorylate PPAR? by activating CDK5 and whether phosphorylation of PPAR? at serine 245 disrupts its interaction with UBR5 and MRN. We further determine whether this is a function of loss of BMPR2, the gene mutant in familial PAH and reduced in idiopathic (I) PAH. We also investigate whether sites of unrepaired DNA damage are associated with specific changes in chromatin accessibility and gene regulation that impact cell phenotype. We also determine whether reversal of DNA damage in PAH PAEC, mediated by Nutlin-3-induced p53 and p53-PPAR? dependent genes, restores chromatin accessibility and gene regulation at sites of DNA damage and thus improves PAEC function. To investigate the relationship between cultured PAEC and cells in the intact pulmonary arteries (PA) from PAH and control lungs, we dissociate the cells and incorporate single cell RNA Seq and proximity ligation in situ hybridization (PLISH) on fixed tissue sections to investigate DNA damage in all cells of the vessel wall as this relates to aberrant gene expression.
In Specific Aim 2, we use two murine models to study the role of DNA damage in the pathogenesis of pulmonary hypertension (PH). Having shown in mice with EC loss of Bmpr2 that reoxygenation after hypoxia causes DNA damage and persistent PH that is reversed with Nutlin-3, we will use single cell RNA Seq to identify gene expression changes that are linked to damaged and repaired DNA. We will also investigate whether the mouse with DNA damage resulting from loss of ATM in EC has PH, and the PA gene expression changes involved.
In Specific Aim 3, we pursue the DNA damage associated with phosphoPPAR? in PAH monocytes and determine whether this is a feature of loss of BMPR2 that is associated with a highly pro-inflammatory pattern of gene expression that can be reversed by roscovitine, the CDK5 inhibitor. Our studies should provide a major inroad into understanding the molecular and functional sequelae of unrepaired DNA in vascular disease and their potential for reversibility of disease.

Public Health Relevance

We seek to understand how abnormalities in a cell surface receptor BMPR2 and transcription factors PPAR gamma and p53 make vascular and inflammatory cells vulnerable to DNA damage that leads to the development of pulmonary arterial hypertension. We investigate how the landscape of chromatin accessibility and gene regulation is altered and assess ways in which we can reverse the DNA damage and pulmonary hypertension by restoring normal function of the BMPR2-PPAR gamma axis.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
2R01HL087118-09
Application #
9737710
Study Section
Respiratory Integrative Biology and Translational Research Study Section (RIBT)
Program Officer
Xiao, Lei
Project Start
2008-04-01
Project End
2023-04-30
Budget Start
2019-05-01
Budget End
2020-04-30
Support Year
9
Fiscal Year
2019
Total Cost
Indirect Cost
Name
Stanford University
Department
Pediatrics
Type
Schools of Medicine
DUNS #
009214214
City
Stanford
State
CA
Country
United States
Zip Code
94305
Zamanian, Roham T; Hedlin, Haley; Greuenwald, Paul et al. (2018) Features and Outcomes of Methamphetamine-associated Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 197:788-800
Miyagawa, Kazuya; Shi, Minyi; Chen, Pin-I et al. (2018) Smooth Muscle Contact Drives Endothelial Regeneration by BMPR2-Notch1 Mediated Metabolic and Epigenetic Changes. Circ Res :
Bonnet, Sébastien; Provencher, Steeve; Guignabert, Christophe et al. (2017) Translating Research into Improved Patient Care in Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 195:583-595
Newman, John H; Rich, Stuart; Abman, Steven H et al. (2017) Enhancing Insights into Pulmonary Vascular Disease through a Precision Medicine Approach. A Joint NHLBI-Cardiovascular Medical Research and Education Fund Workshop Report. Am J Respir Crit Care Med 195:1661-1670
Chen, Pin-I; Cao, Aiqin; Miyagawa, Kazuya et al. (2017) Amphetamines promote mitochondrial dysfunction and DNA damage in pulmonary hypertension. JCI Insight 2:e90427
Hopper, Rachel K; Moonen, Jan-Renier A J; Diebold, Isabel et al. (2016) In Pulmonary Arterial Hypertension, Reduced BMPR2 Promotes Endothelial-to-Mesenchymal Transition via HMGA1 and Its Target Slug. Circulation 133:1783-94
Vattulainen-Collanus, Sanna; Akinrinade, Oyediran; Li, Molong et al. (2016) Loss of PPAR? in endothelial cells leads to impaired angiogenesis. J Cell Sci 129:693-705
Spiekerkoetter, Edda; Sung, Yon K; Sudheendra, Deepti et al. (2015) Low-Dose FK506 (Tacrolimus) in End-Stage Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 192:254-7
Diebold, Isabel; Hennigs, Jan K; Miyagawa, Kazuya et al. (2015) BMPR2 preserves mitochondrial function and DNA during reoxygenation to promote endothelial cell survival and reverse pulmonary hypertension. Cell Metab 21:596-608
de Jesus Perez, Vinicio; Yuan, Ke; Alastalo, Tero-Pekka et al. (2014) Targeting the Wnt signaling pathways in pulmonary arterial hypertension. Drug Discov Today 19:1270-6

Showing the most recent 10 out of 22 publications