Abstract

The therapeutic potential of mesenchymal stromal/stem cells (MSC) for treating cardiac ischemia strongly depends on their paracrine-mediated effects and their engraftment capacity in a hostile environment such as the infarcted myocardium. Adipose tissue-derived stromal vascular fraction (SVF) cells are a mixed population composed mainly of MSC and vascular cells, well known for their high angiogenic potential. A previous study showed that the angiogenic potential of SVF cells was further increased following their in vitro organization in an engineered tissue (patch) after perfusion-based bioreactor culture. This study aimed to investigate the possible changes in the cellular SVF composition, in vivo angiogenic potential, as well as engraftment capability upon in vitro culture in harsh hypoxia conditions. This mimics the possible delayed vascularization of the patch upon implantation in a low perfused myocardium. To this purpose, human SVF cells were seeded on a collagen sponge, cultured for 5 days in a perfusion-based bioreactor under normoxia or hypoxia (21% and <1% of oxygen tension, respectively) and subcutaneously implanted in nude rats for 3 and 28 days. Compared to ambient condition culture, hypoxic tension did not alter the SVF composition in vitro, showing similar numbers of MSC as well as endothelial and mural cells. Nevertheless, in vitro hypoxic culture significantly increased the release of vascular endothelial growth factor (p < 0.001) and the number of proliferating cells (p < 0.00001). Moreover, compared to ambient oxygen culture, exposure to hypoxia significantly enhanced the vessel length density in the engineered tissues following 28 days of implantation. The number of human cells and human proliferating cells in hypoxia-cultured constructs was also significantly increased after 3 and 28 days in vivo, compared to normoxia. These findings show that a possible in vivo delay in oxygen supply might not impair the vascularization potential of SVF- patches, which qualifies them for evaluation in a myocardial ischemia model.

Highlights

  • Animal studies of subacute/chronic cardiac ischemia have demonstrated that the observed benefits of mesenchymal stem/stromal cell (MSC)-based treatments in promoting angiogenesis and cardiomyocyte contractility largely depend on the effect of their secretome [9,10]

  • The static culture did induce the formation of elongated endothelial structures, only under severe hypoxia condition (Figure 1A)

  • This study showed a superior vessel length density as well as a higher percentage of engrafted human cells in stromal vascular fraction (SVF)-patches cultured under severe hypoxia compared to normoxia following 28 days of culture

Read more

Summary

Introduction

Following myocardial infarction (MI), one of the first treatments is the restoration of the macrocirculation (e.g., by a coronary artery bypass graft (CABG)). The microcirculation is often compromised by the presence of a rarefied capillary network in the areas of the infarcted myocardium [1] not restored by standard clinical revascularization interventions. There is still an unmet clinical need for the development of an additional angiogenic therapy targeting the induction of new capillaries for the treatment of subacute or chronic cardiac ischemic disease. Animal studies of subacute/chronic cardiac ischemia have demonstrated that the observed benefits of MSC-based treatments in promoting angiogenesis and cardiomyocyte contractility largely depend on the effect of their secretome (e.g., release of angiogenic, pro-survival factors) [9,10]

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call