1887

Abstract

Abstract

Inflammation is not merely a detrimental response to biomaterials. It can be considered as a natural agent of tissue remodeling, which is pivotal for the approach of in situ cardiovascular tissue engineering. Recently, a biodegradable synthetic scaffold was shown to remodel into a fully functional healthy blood vessel via an inflammation-mediated response. In fact, it was shown that initial infiltration of immune cells (i.e. monocytes) is indispensible for long-term remodeling of scaffolds and functionality of neo-tissues. This remodeling process is orchestrated by potent signaling molecules, of which monocyte chemotactic protein-1 (MCP-1) was identified as a key player. However, the exact mechanism remains unclear. We hypothesize that MCP-1 initiates a desired wound healing cascade by recruiting favorable monocyte and macrophage (M2 type) subpopulations in the implanted scaffold. To investigate the interactions between circulating cells and scaffolds, we have developed and validated a meso-fluidics setup that exposes small-scale 3D scaffolds to a circulating cell suspension in pulsatile flow with pressure and shear stress on the scaffold surface in the physiologic range for aortic valve and small diameter arteries. Electrospun polycaprolactone (PCL) scaffolds were loaded with fibrin gel with and without MCP-1 and placed into the fluidics setup. Human peripheral blood mononuclear cells (hPBMC) were isolated from healthy donors by density gradient centrifuging. The cells were resuspended in culture medium and circulated in the fluidics setup for up to 72 hours at a pulsatile flow (1Hz), with a peak pressure of approximately 100 mmHg and peak shear stress of 1.5 Pa on the scaffold surface. The cell suspensions were characterized at various time points by flow cytometry. The hPBMC suspension at the start consisted of CD45+ lymphocytes and monocytes. After 16 hours in the fluidics setup, the distinct monocyte subpopulation (8-20% of hPBMC) was no longer present in the medium, regardless of MCP-1 presence. This suggests monocyte activation by the scaffold, resulting in cell adhesion and monocyte-to-macrophage differentiation. Moreover, whole-mount immunostaining of the scaffolds demonstrated that addition of MCP-1 resulted in a significant increase in CD163 expression, which indicates the presence of a favorable monocyte subpopulation and macrophage polarization towards a wound healing M2 phenotype. Our results show that the PCL/fibrin scaffold evokes a response from circulating monocytes, resulting in rapid cell adhesion and infiltration. Moreover, this initial response can be modulated using MCP-1 to promote favorable M2 macrophage polarization, initiating the wound healing cascade that is necessary for long term remodeling of the synthetic scaffold.

Loading

Article metrics loading...

/content/papers/10.5339/qproc.2012.heartvalve.4.70
2012-05-01
2024-04-18
Loading full text...

Full text loading...

http://instance.metastore.ingenta.com/content/papers/10.5339/qproc.2012.heartvalve.4.70
Loading
  • Accepted: 04 June 2012
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error