Exploring the Interactions between Mesenchymal Stem Cells and Endothelial Cells in Engineered Perivascular Niches

Authors:
Bita Carrion
In:
Source: Other
Publication Date: (2013)
Issue: 1: 1-159
Research Area:
Basic Research
Cells used in publication:
Fibroblast, lung, human normal (NHLF)
Species: human
Tissue Origin: lung
Abstract
The long term survival of engineered tissue constructs is strongly dependent on oxygen delivery and nutrient exchange provided by the vasculature. Therapeutic approaches to induce neovascularization comprise cell-based therapies using endothelial cells (ECs) co-cultured with stromal cells to form long lasting functional blood vessels. This dissertation investigates the role of cross-talk between ECs and stromal cells in governing the processes of neovascularization in fibrin matrices, representative of the wound healing environment in vivo. Special attention was paid to a specific molecular interaction between the a6ß1 integrin adhesion receptor on bone marrow derived stromal cells (BMSCs) and EC-deposited laminin. Prior studies have shown that co-delivery of ECs and stromal cells supports the formation of robust blood vessel networks. To validate this phenomenon in vitro, we utilized an established three dimensional (3D) microfluidic device as a model system. ECs suspended within 2.5 mg/mL fibrin gels patterned in the device adjacent to stromal cells (either fibroblasts or BMSCs) executed a morphogenetic process akin to vasculogenesis, forming a primitive vascular plexus and maturing into a robust capillary network with hollow well-defined lumens. Both BMSCs and fibroblasts associated with the ECs like pericytes, but promoted capillary morphogenesis with distinct kinetics. Furthermore, biochemical assays revealed that the perivascular association of BMSCs required their a6ß1 integrin receptor, presumably to mediate an interaction with EC-deposited laminin. xi To further investigate the a6ß1 integrin-laminin interactions, we used a 3D in vitro model of angiogenesis in which ECs coated on microcarrier beads were co-cultured with BMSCs within a fibrin matrix. Using RNA interference, we demonstrated that a6 integrin inhibition in BMSCs reduced capillary sprouting, and blocked their ability to associate with nascent blood vessels. Furthermore, we demonstrated that the BMSCs with attenuated a6 integrin expression levels proliferate at a significantly lower rate relative to either control cells expressing non-targeting shRNA or wild type BMSCs. Despite the addition of cells to compensate for this deficit in proliferation, deficient sprouting persists. These data collectively underscore the importance of understanding integrin-mediated interactions between EC-deposited basement membranes and supporting stromal cells during the complex process of vessel formation. The results presented in this dissertation could have significant implications for both physiological and pathological neovascularization.