Abstract

BackgroundThe stromal vascular fraction (SVF) derived from adipose tissue contains adipose-derived stromal/stem cells (ASC) and can be used for regenerative applications. Thus, a validated protocol for SVF isolation, freezing, and thawing is required to manage product administration. To comply with Good Manufacturing Practice (GMP), fetal bovine serum (FBS), used to expand ASC in vitro, could be replaced by growth factors from platelet concentrates.MethodsThroughout each protocol, GMP-compliant reagents and devices were used. SVF cells were isolated from lipoaspirates by a standardized enzymatic protocol. Cells were cryopreserved in solutions containing different albumin or serum and dimethylsulfoxide (DMSO) concentrations. Before and after cryopreservation, we analyzed: cell viability (by Trypan blue); immunophenotype (by flow cytometry); colony-forming unit-fibroblast (CFU-F) formation; and differentiation potential. ASC, seeded at different densities, were expanded in presence of 10% FBS or 5% supernatant rich in growth factors (SRGF) from platelets. The differentiation potential and cell transformation grade were tested in expanded ASC.ResultsWe demonstrated that SVF can be obtained with a consistent yield (about 185 × 103 cells/ml lipoaspirate) and viability (about 82%). Lipoaspirate manipulation after overnight storage at +4 °C reduced cell viability (−11.6%). The relative abundance of ASC (CD34+CD45−CD31–) and endothelial precursors (CD34+CD45−CD31+) in the SVF product was about 59% and 42%, respectively. A period of 2 months cryostorage in autologous serum with added DMSO minimally affected post-thaw SVF cell viability as well as clonogenic and differentiation potentials. Viability was negatively affected when SVF was frozen at a cell concentration below 1.3 × 106 cells/ml. Cell viability was not significantly affected after a freezing period of 1 year.Independent of seeding density, ASC cultured in 5% SRGF exhibited higher growth rates when compared with 10% FBS. ASC expanded in both media showed unaltered identity (by flow cytometry) and were exempt from genetic lesions. Both 5% SRGF- and 10% FBS-expanded ASC efficiently differentiated to adipocytes, osteocytes, and chondrocytes.ConclusionsThis paper reports a GMP-compliant approach for freezing SVF cells isolated from adipose tissue by a standardized protocol. Moreover, an ASC expansion method in controlled culture conditions and without involvement of animal-derived additives was reported.

Highlights

  • The stromal vascular fraction (SVF) derived from adipose tissue contains adipose-derived stromal/stem cells (ASC) and can be used for regenerative applications

  • This paper reports a Good Manufacturing Practice (GMP)-compliant approach for freezing SVF cells isolated from adipose tissue by a standardized protocol

  • We evaluated cell viability by Trypan blue dye exclusion test in the fresh SVF and, as displayed in Fig. 2a, nucleated cells (NC) viability measured in a subset of fresh SVF aliquots treated by red blood cell lysis solution was significantly (p < 0.01) lowered when compared with matched untreated SVF aliquots (77.1 ± 3.9% vs 66.3 ± 4. 9%)

Read more

Summary

Introduction

The stromal vascular fraction (SVF) derived from adipose tissue contains adipose-derived stromal/stem cells (ASC) and can be used for regenerative applications. Beside its role for energy storage, is a known source of stromal precursors and stem cells These cells are enclosed in the so-called stromal vascular fraction (SVF), a heterogeneous population including hematopoietic cells, and adipose-derived stromal/stem cells (ASC). Approaches for the phenotypic characterization of SVF cells were previously suggested in position papers from the International Federation for Adipose Therapeutics and Science (IFATS) and from the International Society of Cellular Therapy (ISCT), as well as in other publications [1,2,3] Such a composite and heterogeneous pool of cells can be used for clinical applications by virtue of the pro-angiogenetic and immune modulatory activity exerted by the different cell populations [1, 2]. Successful SVF cryopreservation using different protocols has been demonstrated in previously published works [3, 5] but there is not clear consensus about the appropriate freezing approach that maximally preserves cell viability

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