Calcific aortic valve (AV) disease is a major cause of cardiac-related deaths worldwide and is a strong risk factor for additional cardiovascular events. AV disease was originally thought to be a """"""""wear and tear"""""""" process but now it is known to be an active inflammatory disease leading to sclerotic and calcific lesions. Further, AV disease occurs preferentially on the fibrosa side whereas the ventricularis side remains relatively healthy. The mechanism by which shear contributes to this disease remains unknown. We hypothesize that disturbed flow conditions are present on the fibrosa side and regulate endothelial gene expression - miRNAs and mRNAs that lead to AV inflammation and calcification. Previously, we have identified shear-responsive miRNAs in human aortic valvular endothelial cells (HAVECs);however, the importance and function of these miRNAs remains unknown. Recently, we have identified side-dependent miRNAs from freshly isolated endothelial- enriched RNA in porcine AVs. From this work, we have identified miR-199a-5p, miR-214, and miR-486-5p as the top three shear- and/or side-dependent miRNAs. We hypothesize that these are mechanosensitive miRNAs in endothelium which leads to AV inflammation and calcification. To address this hypothesis, we propose the following aims. First, we will determine the role of mechanosensitive miRNAs in aortic valve endothelium in vitro and in porcine AVs. Shear- and side-dependent expression of miR-199a-5p, 214, and 486- 5p will be validated in cultured AVECs and freshly isolated, endothelial-enriched RNA from human and porcine AVs using qPCR and in situ hybridization. The role of these miRNAs in shear-sensitive endothelial function, including inflammation, migration, apoptosis, proliferation, and cell cycle will be investigated using gain- r loss-of-function studies. Next, we will determine the role of mechanosensitive miRNAs in aortic valve endothelium ex vivo using porcine AVs. Array studies with validation will be completed in porcine AVs exposed to laminar shear or oscillatory shear stress conditions to identify shear- and side-dependent miRNAs in both endothelial-enriched RNA as well as RNA from the leftover tissue (mainly interstitial cells). Following validation, the levels of the key miRNAs (miR-199a, 214, and 486-5p) will be modulated and assessed for cell function changes pertaining to inflammation and calcification. Finally, we will determine the role of mechanosensitive miRNAs in AV calcification and inflammation in vivo. AV disease will be modeled in vivo using ApoE-/- mice fed a Western diet. Development of inflammation, sclerosis (valve thickness), apoptosis, and calcification will be characterized. This mouse model of AV calcification will be used to validate the selected miRNAs from Aims 1 and 2 in an in vivo setting with the use of LNA inhibitors or miRNA precursors. Identification of these miRNAs will provide insight to molecular mechanisms by which AV disease occurs, offering potential therapeutic targets and diagnostic markers as well as important considerations in designing tissue-engineered valves.

Public Health Relevance

Calcific aortic valve (AV) disease is a major cause of cardiac-related deaths worldwide and is a strong risk factor for additional cardiovascular events. AV disease was originally thought to be a wear and tear process but now it is known to be an active inflammatory disease leading to sclerotic and calcific lesions. Further, AV disease occurs preferentially on the fibrosa side whereas the ventricularis side remains relatively healthy. The mechanism by which shear contributes to this disease remains unknown. We hypothesize that disturbed flow conditions are present on the fibrosa side and regulate endothelial gene expression - miRNAs and mRNAs that lead to AV inflammation and calcification. Identification of these miRNAs will provide insight to molecular mechanisms by which AV disease occurs, offering potential therapeutic targets and diagnostic markers as well as important considerations in designing tissue-engineered valves.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
5R01HL119798-02
Application #
8720061
Study Section
Bioengineering, Technology and Surgical Sciences Study Section (BTSS)
Program Officer
Evans, Frank
Project Start
2013-08-12
Project End
2018-05-31
Budget Start
2014-06-01
Budget End
2015-05-31
Support Year
2
Fiscal Year
2014
Total Cost
Indirect Cost
Name
Emory University
Department
Biomedical Engineering
Type
Schools of Medicine
DUNS #
City
Atlanta
State
GA
Country
United States
Zip Code
30322
Hoganson, David M; Cooper, Dane A; Rich, Kimberly N et al. (2018) Flow Preservation of Umbilical Vein for Autologous Shunt and Cardiovascular Reconstruction. Ann Thorac Surg 105:1809-1818
Schlotter, Florian; Halu, Arda; Goto, Shinji et al. (2018) Spatiotemporal Multi-Omics Mapping Generates a Molecular Atlas of the Aortic Valve and Reveals Networks Driving Disease. Circulation 138:377-393
Simmons, Craig A; Jo, Hanjoong (2018) Editorial: Special Issue on Heart Valve Mechanobiology : New Insights into Mechanical Regulation of Valve Disease and Regeneration. Cardiovasc Eng Technol 9:121-125
Hu, Shuhong; Liu, Yifei; You, Tao et al. (2018) Vascular Semaphorin 7A Upregulation by Disturbed Flow Promotes Atherosclerosis Through Endothelial ?1 Integrin. Arterioscler Thromb Vasc Biol 38:335-343
Heath, Jack M; Fernandez Esmerats, Joan; Khambouneheuang, Lucky et al. (2018) Mechanosensitive microRNA-181b Regulates Aortic Valve Endothelial Matrix Degradation by Targeting TIMP3. Cardiovasc Eng Technol 9:141-150
Sena, Brena F; Figueiredo, Jose Luiz; Aikawa, Elena (2017) Cathepsin S As an Inhibitor of Cardiovascular Inflammation and Calcification in Chronic Kidney Disease. Front Cardiovasc Med 4:88
Kim, Chan Woo; Pokutta-Paskaleva, Anastassia; Kumar, Sandeep et al. (2017) Disturbed Flow Promotes Arterial Stiffening Through Thrombospondin-1. Circulation 136:1217-1232
Kumar, Sandeep; Kang, Dong-Won; Rezvan, Amir et al. (2017) Accelerated atherosclerosis development in C57Bl6 mice by overexpressing AAV-mediated PCSK9 and partial carotid ligation. Lab Invest 97:935-945
Gogas, Bill D; Kumar, Sandeep; Benham, James et al. (2016) TCT-828 Very Late Vasomotor Responses and Gene Expression Profiles of Porcine Coronary Arteries at 4 Years after Deployment of the Everolimus-eluting Bioresorbable Vascular Scaffold and the Everolimus-eluting Metallic Xience V stent. J Am Coll Cardiol 68:B334-B335
Jha, Rajneesh; Wu, Qingling; Singh, Monalisa et al. (2016) Simulated Microgravity and 3D Culture Enhance Induction, Viability, Proliferation and Differentiation of Cardiac Progenitors from Human Pluripotent Stem Cells. Sci Rep 6:30956

Showing the most recent 10 out of 45 publications