Chronic obstructive pulmonary disease (COPD) is the third leading cause of mortality in the United States. COPD often is exacerbated by vasculopathy, which substantially worsens prognosis and limits survival. Vasculopathy is characterized by remodeling and loss of microvessels. Recent evidence has also highlighted a role for the alterations to the microvasculature during the early pathogenesis and heterogeneity of COPD, although the underlying mechanisms are not defined. The goal of this proposal is to address this knowledge gap by defining the molecular mechanisms whereby reciprocal mesenchymal progenitor-endothelial cell interactions regulate pulmonary microvascular structure and the development of COPD. In the last funding cycle, we reported the existence of a novel population of mesenchymal progenitor cells (MPC) which serve as progenitors for pericytes and therefore are required to maintain microvascular homeostasis. We further reported that Wnt/?-catenin signaling was an important regulator of this MPC function. However, how increased Wnt signaling in MPC, production of its modulator Dkk1 and the development of vasculopathy impacts surfaces for gas exchange and the pathophysiology of COPD is unknown. Therefore, understanding Wnt signaling in MPC, will be important to facilitate microvascular and tissue function. The novel premise of this proposal is that enhanced Wnt/?-catenin signaling within MPCs indirectly leads to emphysema and COPD by altering normal MPC-microvascular endothelial cell (MVEC) interactions. We hypothesize that activation of Wnt/?-catenin in MPC exacerbates the onset of COPD via 1) increased production of Dkk1 and b) a paracrine effect of Dkk1 on microvascular endothelial cell function. We will test that activation of Wnt/?- catenin in MPCs leads to COPD via increased MPC expression of Dkk1 and subsequent paracrine alteration of microvascular endothelial cell-fate specification. To test whether MPC expression of ?-catenin and Dkk1 are critical for maintenance of MPC-MVEC cross talk and tissue remodeling in COPD, we will conditionally knock down or overexpress Dkk1 and ?-catenin in MPC and expose mice to smoke or vascular injury. We will employ novel models of lineage analyses, optical coherence tomography (OCT), histological indices of angiogenesis and measures of barrier function. We will also test that maintenance of MPC-MVEC interaction via manipulation of Dkk1 signaling will attenuate loss of microvascular function and tissue structure following injury. We will manipulate Dkk1/Wnt signaling in human and mouse lung MPCs in vitro and inhibit Dkk1 signaling in murine models in vivo, to evaluate the mechanism by which MVEC function is affected. These studies will determine whether modulation of MPC, or inhibition / knockout Dkk1 signaling in MVEC is a viable target to promote microvascular function and attenuate COPD. Our proposed studies will advance the field of by defining Wnt dependent mechanisms by which MPC regulate microvascular endothelial function and identify targets to reverse vasculopathy in COPD, by repurposing of FDA approved DKK1 modulators.

Public Health Relevance

It is generally assumed that vasculopathies associated with emphysema/COPD are attributable to deficient vascular endothelium or smooth muscle cell phenotypes. We challenge this paradigm by defining an adult mesenchymal pericyte progenitor (ABCG2pos MPC) that regulates both microvascular function during tissue homeostasis and adaptive angiogenesis in response to injury, capable of exacerbating COPD. Therefore, the ABCG2posMPC compartment of lung is a previously unstudied target that will be exploited in our proposed studies to identify new pathways relevant to the pathobiology of COPD.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
2R01HL116597-05A1
Application #
9520705
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Punturieri, Antonello
Project Start
2013-08-01
Project End
2022-05-31
Budget Start
2018-06-01
Budget End
2019-05-31
Support Year
5
Fiscal Year
2018
Total Cost
Indirect Cost
Name
Vanderbilt University Medical Center
Department
Type
DUNS #
079917897
City
Nashville
State
TN
Country
United States
Zip Code
37232
Gavin, Kathleen M; Gutman, Jonathan A; Kohrt, Wendy M et al. (2016) De novo generation of adipocytes from circulating progenitor cells in mouse and human adipose tissue. FASEB J 30:1096-108
Bryant, Andrew J; Carrick, Ryan P; McConaha, Melinda E et al. (2016) Endothelial HIF signaling regulates pulmonary fibrosis-associated pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 310:L249-62
Chen, Xinping; Talati, Megha; Fessel, Joshua P et al. (2016) Estrogen Metabolite 16?-Hydroxyestrone Exacerbates Bone Morphogenetic Protein Receptor Type II-Associated Pulmonary Arterial Hypertension Through MicroRNA-29-Mediated Modulation of Cellular Metabolism. Circulation 133:82-97
Majka, Susan M; Miller, Heidi L; Helm, Karen M et al. (2014) Analysis and isolation of adipocytes by flow cytometry. Methods Enzymol 537:281-96
Nozik-Grayck, Eva; Woods, Crystal; Taylor, Joann M et al. (2014) Selective depletion of vascular EC-SOD augments chronic hypoxic pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 307:L868-76
Marriott, Shennea; Baskir, Rubin S; Gaskill, Christa et al. (2014) ABCG2pos lung mesenchymal stem cells are a novel pericyte subpopulation that contributes to fibrotic remodeling. Am J Physiol Cell Physiol 307:C684-98
West, James D; Austin, Eric D; Gaskill, Christa et al. (2014) Identification of a common Wnt-associated genetic signature across multiple cell types in pulmonary arterial hypertension. Am J Physiol Cell Physiol 307:C415-30
Meloche, Jolyane; Courchesne, Antony; Barrier, Marjorie et al. (2013) Critical role for the advanced glycation end-products receptor in pulmonary arterial hypertension etiology. J Am Heart Assoc 2:e005157
Chow, Kelsey; Fessel, Joshua P; Kaoriihida-Stansbury et al. (2013) Dysfunctional resident lung mesenchymal stem cells contribute to pulmonary microvascular remodeling. Pulm Circ 3:31-49