Adipose-Derived Stem/Stromal Cells, Stromal Vascular Fraction, and Microfragmented Adipose Tissue

2022 ◽  
pp. 47-61
Author(s):  
Enrico Ragni ◽  
Marco Viganò ◽  
Paola De Luca ◽  
Edoardo Pedrini ◽  
Laura de Girolamo
2017 ◽  
Vol 2017 ◽  
pp. 1-14 ◽  
Author(s):  
Jonathan Rodriguez ◽  
Anne-Sophie Pratta ◽  
Nacira Abbassi ◽  
Hugo Fabre ◽  
Fanny Rodriguez ◽  
...  

Adipose-derived stem/stromal cells (ASCs) reside in the stromal vascular fraction (SVF) of adipose tissue (AT) and can be easily isolated. However, extraction of the SVF from lipoaspirate is a critical step in generating ASC, and semiautomated devices have been developed to enhance the efficacy and reproducibility of the outcomes and to decrease manipulation and contamination. In this study, we compared the reference method used in our lab for SVF isolation from lipoaspirate, with three medical devices: GID SVF-1™, Puregraft™, and Stem.pras®. Cell yield and their viability were evaluated as well as their phenotype with flow cytometry. Further on, we determined their proliferative potential using population doublings (PD), PD time (PDT), and clonogenicity assay (CFU-F). Finally, we checked their genetic stability using RT-qPCR for TERT mRNA assay and karyotyping as well as their multilineage potential including adipogenic, chondrogenic, and osteogenic differentiation. Our results demonstrate that all the devices allow the production of SVF cells with consistent yield and viability, in less time than the reference method. Expanded cells from the four methods showed no significant differences in terms of phenotype, proliferation capabilities, differentiation abilities, and genetic stability.


2019 ◽  
Vol 35 (04) ◽  
pp. 358-367 ◽  
Author(s):  
Joris A. van Dongen ◽  
Joeri van Boxtel ◽  
Martin C. Harmsen ◽  
Hieronymus P. Stevens

AbstractLipofilling, the transplantation of adipose tissue, has already been used since the end of the 19th century. For decades, lipofilling was used to restore loss of volume due to aging, trauma, or congenital defects. Later on, the indications for the use of lipofilling expanded by treating aged skin, scars, and improving wound healing. The expansion was caused by the discovery of adipose derived stromal cells (ASCs) in adipose tissue and the development of very fine harvesting and injection cannulas which made it possible to inject small adipose tissue particles in small volume areas, such as the face. ASCs are multipotent stromal cells which reside in the stromal vascular fraction (SVF) of adipose tissue and are able to differentiate in multiple cell lineages and secrete a plurality of growth factors with regenerative potentials. The discovery of ASCs led toward more experimental cell-based therapies, that is, ASCs or SVF isolated by means of enzymatic isolation procedures. Later on, enzymatic isolation procedures were forbidden in many countries by legislation and were replaced by mechanical isolation procedures, such as the Nanofat and Fractionation of Adipose Tissue (FAT) procedures. The Nanofat procedure has been extensively investigated, especially as treatment for skin rejuvenation in the face. Though, substantial evidence is lacking for using facial lipofilling or any therapeutic component, that is, ASCs or SVF for skin rejuvenation to date. In contrast, facial lipofilling to restore loss of volume seems to be promising.


2010 ◽  
Vol 5 (1) ◽  
pp. 47-58 ◽  
Author(s):  
Aušra Unguryte ◽  
Eiva Bernotiene ◽  
Algirdas Venalis

AbstractMore effective techniques should be employed for isolation of human mesenchymal stromal cells derived from adipose tissue (ADSC), seeking to make adipose tissue biopsies smaller in volume and thus less invasive. In this study, we compared properties of ADSC isolated by several different methods from the same samples of adipose tissue in order to enhance yields of potential ADSC. The mature adipocyte fraction was investigated using the ceiling culture method, including both ceiling and bottom cell fractions, and the control culture method with standard amount of medium. The results were also compared using the stromal vascular fraction from the same samples. The most efficient was the bottom cell population isolated from the mature adipocyte fraction by ceiling culture method. These cells readily differentiated into osteogenic, adipogenic and chondrogenic lineages and, similar to stromal vascular fraction cells, displayed high proliferation potential. Cultures of mature adipocyte fractions with standard amount of medium were considerably less effective. Mature adipocyte fractions yields large quantities of adipose-derived stem cells that have properties comparable with stromal vascular fraction cells suitable for tissue regeneration, especially when only small biopsies can be taken.


2018 ◽  
Vol 128 (6) ◽  
Author(s):  
Michael Griessl ◽  
Anna‐Maria Buchberger ◽  
Sybille Regn ◽  
Kilian Kreutzer ◽  
Katharina Storck

Genes ◽  
2019 ◽  
Vol 10 (6) ◽  
pp. 474 ◽  
Author(s):  
Polancec ◽  
Zenic ◽  
Hudetz ◽  
Boric ◽  
Jelec ◽  
...  

Osteoarthritis (OA) is a degenerative joint disease accompanied by pain and loss of function. Adipose tissue harbors mesenchymal stem/stromal cells (MSC), or medicinal signaling cells as suggested by Caplan (Caplan, 2017), used in autologous transplantation in many clinical settings. The aim of the study was to characterize a stromal vascular fraction from microfragmented lipoaspirate (SVF-MLA) applied for cartilage treatment in OA and compare it to that of autologous lipoaspirate (SVF-LA). Samples were first stained using a DuraClone SC prototype tube for the surface detection of CD31, CD34, CD45, CD73, CD90, CD105, CD146 and LIVE/DEAD Yellow Fixable Stain for dead cell detection, followed by DRAQ7 cell nuclear dye staining, and analyzed by flow cytometry. In SVF-LA and SVF-MLA samples, the following population phenotypes were identified within the CD45- fraction: CD31+CD34+CD73±CD90±CD105±CD146± endothelial progenitors (EP), CD31+CD34-CD73±CD90±CD105-CD146± mature endothelial cells, CD31-CD34-CD73±CD90+CD105-CD146+ pericytes, CD31-CD34+CD73±CD90+CD105-CD146+ transitional pericytes, and CD31-CD34+CD73highCD90+CD105-CD146- supra-adventitial-adipose stromal cells (SA-ASC). The immunophenotyping profile of SVF-MLA was dominated by a reduction of leukocytes and SA-ASC, and an increase in EP, evidencing a marked enrichment of this cell population in the course of adipose tissue microfragmentation. The role of EP in pericyte-primed MSC-mediated tissue healing, as well as the observed hormonal implication, is yet to be investigated.


2008 ◽  
Vol 295 (5) ◽  
pp. C1271-C1280 ◽  
Author(s):  
Rosalinda Madonna ◽  
Raffaele De Caterina

Adipose tissue development is associated with neovascularization, which might be exploited therapeutically. We investigated the neovasculogenesis antigenic profile and kinetics in adipose tissue-derived stromal cells (ADSCs) to understand the potential of ADSCs to generate new vessels. Murine and human visceral adipose tissues were processed with collagenase to obtain ADSCs from the stromal vascular fraction. Freshly isolated murine and human ADSCs featured the expression of early markers of endothelial differentiation [uptake of DiI-labeled acetylated LDL, CD133, CD34, kinase insert domain receptor (KDR)], but not markers for more mature endothelial cells (CD31 and von Willebrand factor). In methylcellulose medium, multilocular cells positive for Oil Red O staining appeared after 6 days. After 10 days, clusters of ADSCs spontaneously formed branched tubelike structures, which were strongly positive for CD34 and CD31, while losing their ability to undergo adipocyte differentiation. In Matrigel, in the presence of endothelial growth factors ADSCs formed branched tubelike structures. By clonal assays in methylcellulose we also determined the frequency of granulocyte-macrophage (CFU-GM) and erythroid (BFU-E) colony-forming units from ADSCs, compared with bone marrow-derived stromal cells (BMSCs) used as a positive control. After 4–14 days, BMSCs formed 8 ± 3 BFU-E and 40 ± 10 CFU-GM, while ADSCs never produced colonies of myeloid progenitors. The developing adipose tissue has neovasculogenic potential, based on the recruitment of local rather than circulating progenitors. Adipose tissue might therefore be a viable autonomous source of cells for postnatal neovascularization.


Sign in / Sign up

Export Citation Format

Share Document