A cornerstone of several common therapies for human diseases is the surgical use of a vein as an arterial conduit. Autologous vein bypass grafts are commonly performed for advanced cardiovascular and peripheral vascular disease;similarly, surgeons also perform arteriovenous fistulae (AVF), the preferred access for hemodialysis. The poor patency of both vein grafts and AVF, requiring additional re-do procedures and surgery, reflects our imperfect understanding of the biology of venous remodeling that leads to successful venous adaptation to the arterial environment. This knowledge gap creates an unmet medical need for novel approaches to enhance venous adaptation, maturation, and successful long-term use of venous conduits. The tyrosine kinase receptor Eph-B4 is an embryonic determinant of veins. Diminished Eph-B4 expression is associated with vein graft adaptation in humans and mice, and reduced Eph-B4 activity is essential for venous adaptation in adult mouse vein grafts. We present exciting new data that: 1) our innovative mouse model of AVF faithfully recapitulates human AVF maturation, including a subset that fail to mature;2) in both humans and mice, Eph-B4 expression increases during AVF adaptation to the arterial environment, unlike the decreased Eph-B4 expression during vein graft adaptation;3) Eph-B4 function is essential for AVF adaptation;and 4) Eph-B4 tyrosine-774 is a critical site of Eph-B4 phosphorylation and downstream signaling. We hypothesize that vein graft adaptation leads to loss of vessel identity and AVF maturation leads to merged arterial-venous identity. Our data also suggest a mechanism, e.g. that Eph-B4 is a previously unrecognized but critical endothelial transducer of hemodynamic loads to veins. We hypothesize that differences in the hemodynamic environments of vein grafts and AVF differentially regulate Eph-B4 tyrosine-774 phosphorylation and/or Eph-B4 expression. We will use a combination of in vitro and in vivo models, using a bioreactor that can individually control separate hemodynamic loads on both large and small vessels and endothelial monolayers, as well as using in vivo models of vein grafts and AVF, to test our hypothesis with the following specific aims:
Aim I : Determine how hemodynamic loads differentially regulate Eph-B4 phosphorylation and function in whole vessels, including analysis of successful and failed AVF.
Aim II : Determine how different magnitudes of shear stress regulate Eph-B4 phosphorylation and function in endothelial cells.
Aim III : Determine how Eph-B4 tyrosine-774 phosphorylation regulates venous endothelial cell function. The work in this proposal will establish our innovative and paradigm-changing hypothesis that Eph-B4 is a novel and critical transducer of mechanical loads to the blood vessel and that Eph-B4 is differentially responsive to different types of loads, even in the absence of Ephrin-B2. Eph-B4 activity or lack thereof, defines the phenotype of the blood vessel and its function. Abnormal regulation of vessel identity during vein graft adaptation and AVF maturation can be manipulated to improve human clinical therapies.

Public Health Relevance

A cornerstone of several common therapies for human diseases is the surgical use of a vein as an arterial conduit, such as the autologous vein bypass grafts that are commonly performed for advanced cardiovascular and peripheral vascular disease as well as arteriovenous fistulae (AVF), the preferred access for hemodialysis. The recent disappointing failure of the PREVENT trials emphasizes the continuing poor long- term patency of vein grafts and short-term patency of AVF that lead to patient suffering and extensive expenditure of precious healthcare dollars and other resources for additional re-do procedures and surgery. The work to be accomplished within this proposal addresses the knowledge gap in the biology of venous remodeling that has created a serious unmet medical need for novel approaches to enhance venous adaptation to the arterial environment, maturation, and successful long-term use of venous conduits.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
High Priority, Short Term Project Award (R56)
Project #
2R56HL095498-06
Application #
8903555
Study Section
Bioengineering, Technology and Surgical Sciences Study Section (BTSS)
Program Officer
Reid, Diane M
Project Start
2014-09-01
Project End
2015-08-31
Budget Start
2014-09-01
Budget End
2015-08-31
Support Year
6
Fiscal Year
2014
Total Cost
Indirect Cost
Name
Yale University
Department
Surgery
Type
Schools of Medicine
DUNS #
City
New Haven
State
CT
Country
United States
Zip Code
06510
Yu, Jun; Dardik, Alan (2018) A Murine Model of Hind Limb Ischemia to Study Angiogenesis and Arteriogenesis. Methods Mol Biol 1717:135-143
Protack, Clinton D; Foster, Trenton R; Hashimoto, Takuya et al. (2017) Eph-B4 regulates adaptive venous remodeling to improve arteriovenous fistula patency. Sci Rep 7:15386
Bai, Hualong; Li, Xin; Hashimoto, Takuya et al. (2017) Patch Angioplasty in the Rat Aorta or Inferior Vena Cava. J Vis Exp :
Wang, Mo; Collins, Michael J; Foster, Trenton R et al. (2017) Eph-B4 mediates vein graft adaptation by regulation of endothelial nitric oxide synthase. J Vasc Surg 65:179-189
Kuwahara, Go; Hashimoto, Takuya; Tsuneki, Masayuki et al. (2017) CD44 Promotes Inflammation and Extracellular Matrix Production During Arteriovenous Fistula Maturation. Arterioscler Thromb Vasc Biol 37:1147-1156
Bai, Hualong; Lee, Jung Seok; Chen, Elizabeth et al. (2017) Covalent modification of pericardial patches for sustained rapamycin delivery inhibits venous neointimal hyperplasia. Sci Rep 7:40142
Assi, Roland; Foster, Trenton R; He, Hao et al. (2016) Delivery of mesenchymal stem cells in biomimetic engineered scaffolds promotes healing of diabetic ulcers. Regen Med 11:245-60
Bai, Hualong; Wang, Mo; Foster, Trenton R et al. (2016) Pericardial patch venoplasty heals via attraction of venous progenitor cells. Physiol Rep 4:
Hu, Haidi; Patel, Sandeep; Hanisch, Jesse J et al. (2016) Future research directions to improve fistula maturation and reduce access failure. Semin Vasc Surg 29:153-171
Hashimoto, Takuya; Yamamoto, Kota; Foster, Trenton et al. (2016) Intraluminal Drug Delivery to the Mouse Arteriovenous Fistula Endothelium. J Vis Exp :e53905

Showing the most recent 10 out of 12 publications