Blood vessel formation requires a group of endothelial cells with heterogeneous responses to signaling inputs. During development, endothelial cells respond differentially to vascular endothelial growth factor (VEGF)-A signaling to adopt phenotypes required for network expansion. Abnormal vascular development associated with pathological conditions such as tumorigenesis or diabetic retinopathy likely results in part from loss of regulated endothelial heterogeneity. VEGF receptor Flt-1 (VEGFR-1) contributes to network formation via heterogeneous expression of the soluble isoform (sFlt-1) that in turn spatially regulates VEGF signaling to provide local sprout guidance to emerging vessel sprouts (Chappell et al, 2009). Phenotypic heterogeneity of endothelial cells in developing vessels is likely important for other aspects of vascular development, such as endothelial interactions with perivascular cells known as pericytes. Pericytes provide structural stability to maturing vessels, and perturbations in endothelial-pericyte interactions contribute to vascular pathologies. Thus, it is intriguing to speculate that endothelial phenotypic heterogeneity is modulated by Flt-1 regulation of VEGF signaling, and that aspects of this heterogeneity facilitate proper endothelial-pericyte interactions. One primary objective of this study therefore is to investigate how Flt-1 spatially regulates endothelial cell heterogeneity to establish proper vascular morphogenesis in vivo. Vascular morphology will be observed in developing mouse retinas with mosaic flt-1 expression via use of flt-1 conditional deletion mice. In vivo and in vito observations will then be used to generate a computational model for Flt-1 activity in regulating the phenotypic heterogeneity of endothelial cells and overall vessel morphology. In addition, the role of Flt-1 in spatially regulating endothelial-pericyte associations will be explored with in viro assays. In embryonic stem (ES) cell-derived vessels, VEGF signaling will be perturbed via genetic manipulation of flt-1 expression. Endothelial-pericyte interactions will be evaluated to characterize the spatial regulation of pericyte recruitment and investment. To assess the effect of altered spatial distribution of flt-1 expression on endothelial-pericyte interactions, mosaic vessels composed of wild-type (WT) and flt-1 mutant cells will be evaluated for pericyte investment. A computational model simulating how Flt-1 promotes vessel endothelial cell heterogeneity to regulate pericyte-endothelial cell interactions will be created as a tool to understand the biological consequences of disruptions in flt-1 expression (e.g. tumor setting). Observations from in vitro experiments will guide the construction and testing of this in silico model. Lastly, the mechanisms by which Flt-1 regulates pericyte-endothelial interactions in vivo will be characterized. Retinal vasculature from developing flt-1 conditional deletion mice will be evaluated for mosaic flt-1 expression and investment of pericytes. Simulations generated by the computer model for Flt-1 regulation of pericyte associations will provide a means for interpreting, analyzing, and advancing experimental observations and approaches.

Public Health Relevance

Blood vessel formation requires a group of vascular endothelial cells to engage in a variety of responses to molecular signaling inputs such that individual cells adopt different functions for network expansion. Functional differences in these endothelial cells are likely important for other aspects of vessel growth, such as their interactions with vascular support cells known as pericytes, an important cell type that surrounds vessels to maintain the structural stability and integrity of the vessel wall. Abnormal vascular development associated with pathologies such as tumor growth and metastasis or diabetic retinopathy likely results in part from losing these distinctions in endothelial cell function and from disruptions in endothelial-pericyte interactions.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Career Transition Award (K99)
Project #
1K99HL105779-01A1
Application #
8242399
Study Section
Special Emphasis Panel (ZHL1-CSR-P (O2))
Program Officer
Scott, Jane
Project Start
2012-05-15
Project End
2014-04-30
Budget Start
2012-05-15
Budget End
2013-04-30
Support Year
1
Fiscal Year
2012
Total Cost
$118,248
Indirect Cost
$8,759
Name
University of North Carolina Chapel Hill
Department
Type
Schools of Medicine
DUNS #
608195277
City
Chapel Hill
State
NC
Country
United States
Zip Code
27599
Arreola, Alexandra; Payne, Laura Beth; Julian, Morgan H et al. (2018) Von Hippel-Lindau mutations disrupt vascular patterning and maturation via Notch. JCI Insight 3:
Nesmith, Jessica E; Chappell, John C; Cluceru, Julia G et al. (2017) Blood vessel anastomosis is spatially regulated by Flt1 during angiogenesis. Development 144:889-896
Chappell, John C; Cluceru, Julia G; Nesmith, Jessica E et al. (2016) Flt-1 (VEGFR-1) coordinates discrete stages of blood vessel formation. Cardiovasc Res 111:84-93
Walpole, J; Chappell, J C; Cluceru, J G et al. (2015) Agent-based model of angiogenesis simulates capillary sprout initiation in multicellular networks. Integr Biol (Camb) 7:987-97