Adipose tissue-derived stromal vascular fraction in regenerative medicine: a brief review on biology and translation

Pablo Bora, Anish S Majumdar, Pablo Bora, Anish S Majumdar

Abstract

Adipose/fat tissue provides an abundant source of stromal vascular fraction (SVF) cells for immediate administration and can also give rise to a substantial number of cultured, multipotent adipose-derived stromal cells (ADSCs). Recently, both SVF and ADSCs have gained wide-ranging translational significance in regenerative medicine. Initially used for cosmetic breast enhancement, this mode of treatment has found use in many diseases involving immune disorders, tissue degeneration, and ischaemic conditions. In this review, we try to address several important aspects of this field, outlining the biology, technology, translation, and challenges related to SVF- and ADSC-based therapies. Starting from the basics of SVF and ADSC isolation, we touch upon recently developed technologies, addressing elements of novel methods and devices under development for point-of-care isolation of SVF. Characterisation of SVF cells and ADSCs is also an evolving area and we look into unusual expression of CD34 antigen as an interesting marker for such purposes. Based on reports involving different cells of the SVF, we draw a potential mode of action, focussing on angiogenesis since it involves multiple cells, unlike immunomodulation which is governed predominantly by ADSCs. We have looked into the latest research, experimental therapies, and clinical trials which are utilising SVF/ADSCs in conditions such as multiple sclerosis, Crohn's disease, peripheral neuropathy, osteoarthritis, diabetic foot ulcer, and so forth. However, problems have arisen with regards to the lack of proper regulatory guidelines for such therapies and, since the introduction of US Food and Drug Administration draft guidelines and the Reliable and Effective Growth for Regenerative Health Options that Improve Wellness (REGROW) Act, the debate became more public with regards to safe and efficacious use of these cells.

Keywords: CD34; Multipotent-stromal cells; Point-of-care biomedical devices; Regenerative medicine; Regulation of stem cell therapeutics; Stromal vascular fraction.

Figures

Fig. 1
Fig. 1
Potential mechanism of action of ADSCs and ECs present in SVF towards angiogenesis. Breakdown of adipose tissue releases many cell types, which together are termed SVF. The cells of the SVF can produce several bioactive soluble factors. ADSCs and EPCs, two important components of SVF, cross-talk via VEGF and PDGF-BB, respectively (among other components), to enable cell proliferation, homing towards injury, neovascularisation and other inter-connected outcomes. ADSC adipose-derived stromal cell, bFGF basic fibroblast growth factor, EC endothelial cell, EPC endothelial progenitor cell, GF growth factor, IGF-1 insulin-like growth factor-1, MMP matrix metalloproteinase, PDGF platelet-derived growth factor, RBC red blood cell, SVF stromal vascular fraction, VEGF vascular endothelial growth factor

References

    1. Bourin P, Bunnell BA, Casteilla L, et al. Stromal cells from the adipose tissue-derived stromal vascular fraction and culture expanded adipose tissue-derived stromal/stem cells: a joint statement of the International Federation for Adipose Therapeutics and Science (IFATS) and the International Society for Cellular Therapy (ISCT) Cytotherapy. 2013;15:641–8. doi: 10.1016/j.jcyt.2013.02.006.
    1. Gimble JM, Bunnell BA, Frazier T, et al. Adipose-derived stromal/stem cells. Organogenesis. 2013;9:3–10. doi: 10.4161/org.24279.
    1. Nguyen A, Guo J, Banyard DA, et al. Stromal vascular fraction: a regenerative reality? Part 1: current concepts and review of the literature. J Plast Reconstr Aesthetic Surg. 2016;69:170–9. doi: 10.1016/j.bjps.2015.10.015.
    1. Bunnell B, Flaat M, Gagliardi C, et al. Adipose-derived stem cells: isolation, expansion and differentiation. Methods. 2008;45:115–20. doi: 10.1016/j.ymeth.2008.03.006.
    1. Casteilla L. Adipose-derived stromal cells: their identity and uses in clinical trials, an update. World J Stem Cells. 2011;3:25. doi: 10.4252/wjsc.v3.i4.25.
    1. Suzuki E, Fujita D, Takahashi M, et al. Adipose tissue-derived stem cells as a therapeutic tool for cardiovascular disease. World J Cardiol. 2015;7:454–65. doi: 10.4330/wjc.v7.i11.707.
    1. Bony C, Cren M, Domergue S, et al. Adipose mesenchymal stem cells isolated after manual or water-jet-assisted liposuction display similar properties. Front Immunol. 2016;6:1–8. doi: 10.3389/fimmu.2015.00655.
    1. Mi HM, Sun YK, Yeon JK, et al. Human adipose tissue-derived mesenchymal stem cells improve postnatal neovascularization in a mouse model of hindlimb ischemia. Cell Physiol Biochem. 2006;17:279–90. doi: 10.1159/000094140.
    1. van Dijk A, Naaijkens BA, Jurgens WJFM, et al. Reduction of infarct size by intravenous injection of uncultured adipose derived stromal cells in a rat model is dependent on the time point of application. Stem Cell Res. 2011;7:219–29. doi: 10.1016/j.scr.2011.06.003.
    1. Charles-de-Sá L, Gontijo-de-Amorim NF, Maeda Takiya C, et al. Antiaging treatment of the facial skin by fat graft and adipose-derived stem cells. Plast Reconstr Surg. 2015;135:999–1009. doi: 10.1097/PRS.0000000000001123.
    1. Semon JA, Zhang X, Pandey AC, et al. Administration of murine stromal vascular fraction ameliorates chronic experimental autoimmune encephalomyelitis. Stem Cells Transl Med. 2013;2:789–96. doi: 10.5966/sctm.2013-0032.
    1. You D, Jang MJ, Kim BH, et al. Comparative study of autologous stromal vascular fraction and adipose-derived stem cells for erectile function recovery in a rat model of cavernous nerve injury. Stem Cells Transl Med. 2015;4:351–8. doi: 10.5966/sctm.2014-0161.
    1. Mohammadi R, Sanaei N, Ahsan S, et al. Stromal vascular fraction combined with silicone rubber chamber improves sciatic nerve regeneration in diabetes. Chinese J Traumatol. 2015;18:212–8. doi: 10.1016/j.cjtee.2014.10.005.
    1. Yoshimura K, Sato K, Aoi N, et al. Cell-assisted lipotransfer for cosmetic breast augmentation: supportive use of adipose-derived stem/stromal cells. Aesthetic Plast Surg. 2008;32:48–55. doi: 10.1007/s00266-007-9019-4.
    1. Pak J, Chang J-J, Lee JH, et al. Safety reporting on implantation of autologous adipose tissue-derived stem cells with platelet-rich plasma into human articular joints. BMC Musculoskelet Disord. 2013;14:337. doi: 10.1186/1471-2474-14-337.
    1. Yoshimura K, Suga H, Eto H. Adipose-derived stem/progenitor cells: roles in adipose tissue remodeling and potential use for soft tissue augmentation. Regen Med. 2009;4:265–73. doi: 10.2217/17460751.4.2.265.
    1. Matsumoto D, Sato K, Gonda K, et al. Cell-assisted lipotransfer: supportive use of human adipose-derived cells for soft tissue augmentation with lipoinjection. Tissue Eng. 2006;12:3375–82. doi: 10.1089/ten.2006.12.3375.
    1. Silva FJ, Holt DJ, Vargas V, et al. Metabolically active human brown adipose tissue derived stem cells. Stem Cells. 2014;32:572–81. doi: 10.1002/stem.1595.
    1. Gimble JM, Katz AJ, Bunnell BA. Adipose-derived stem cells for regenerative medicine. Circ Res. 2007;100:1249–60. doi: 10.1161/01.RES.0000265074.83288.09.
    1. Colen BD. A pill to shed fat? Harvard stem cell researchers say they finally can turn “bad” fat into “good.” Harvard Gaz 2014. . Accessed 8 Sept 2016.
    1. Moisan A, Lee Y-K, Zhang JD, et al. White-to-brown metabolic conversion of human adipocytes by JAK inhibition. Nat Cell Biol. 2014;17:57–67. doi: 10.1038/ncb3075.
    1. Oberbauer E, Steffenhagen C, Wurzer C, et al. Enzymatic and non-enzymatic isolation systems for adipose tissue-derived cells: current state of the art. Cell Regen. 2015;4:7. doi: 10.1186/s13619-015-0020-0.
    1. Tonnard P, Verpaele A, Peeters G, et al. Nanofat grafting: basic research and clinical applications. Plast Reconstr Surg. 2013;132:1017–26. doi: 10.1097/PRS.0b013e31829fe1b0.
    1. Aronowitz JA, Lockhart RA, Hakakian CS. Mechanical versus enzymatic isolation of stromal vascular fraction cells from adipose tissue. Springerplus. 2015;4:713. doi: 10.1186/s40064-015-1509-2.
    1. Shah FS, Wu X, Dietrich M, et al. A non-enzymatic method for isolating human adipose tissue-derived stromal stem cells. Cytotherapy. 2013;15:979–85. doi: 10.1016/j.jcyt.2013.04.001.
    1. Guo J, Nguyen A, Banyard DA, et al. Stromal vascular fraction: a regenerative reality? Part 2: mechanisms of regenerative action. J Plast Reconstr Aesthetic Surg. 2016;69:180–8. doi: 10.1016/j.bjps.2015.10.014.
    1. Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8:315–7. doi: 10.1080/14653240600855905.
    1. Zuk PA, Zhu M, Mizuno H, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001;7:211–28. doi: 10.1089/107632701300062859.
    1. SundarRaj S, Deshmukh A, Priya N, et al. Development of a system and method for automated isolation of stromal vascular fraction from adipose tissue lipoaspirate. Stem Cells Int. 2015;2015:1–11. doi: 10.1155/2015/109353.
    1. Riis S, Zachar V, Boucher S, et al. Critical steps in the isolation and expansion of adipose-derived stem cells for translational therapy. Expert Rev Mol Med. 2015;17:e11. doi: 10.1017/erm.2015.10.
    1. Tamburino S, Lombardo GA, Tarico MS, et al. The role of nanofat grafting in vulvar lichen sclerosus: a preliminary report. Arch Plast Surg. 2016;43:93. doi: 10.5999/aps.2016.43.1.93.
    1. Kemaloğlu CA. Nanofat grafting under a split-thickness skin graft for problematic wound management. Springerplus. 2016;5:138. doi: 10.1186/s40064-016-1808-2.
    1. Fraser JK, Hicok KC, Shanahan R, et al. The Celution(®) system: automated processing of adipose-derived regenerative cells in a functionally closed system. Adv Wound Care. 2014;3:38–45. doi: 10.1089/wound.2012.0408.
    1. Invitrogen™ Countess™. Invitrogen cell data sheet-ADSC. . Accessed 7 Sept 2016.
    1. Sponsored paper. Rapid analysis of human adipose-derived stem cells and 3 T3-L1 differentiation toward adipocytes using the Scepter™ 2.0 cell counter. Biotechniques. 2012;53:109–11.
    1. Ryu YJ, Cho TJ, Lee DS, et al. Phenotypic characterization and in vivo localization of human adipose-derived mesenchymal stem cells. Mol Cells. 2013;35:557–64. doi: 10.1007/s10059-013-0112-z.
    1. Asahara T, Kawamoto A, Masuda H. Concise review: circulating endothelial progenitor cells for vascular medicine. Stem Cells. 2011;29:1650–5. doi: 10.1002/stem.745.
    1. Garipcan B, Maenz S, Pham T, et al. Image analysis of endothelial microstructure and endothelial cell dimensions of human arteries—a preliminary study. Adv Eng Mater. 2011;13:B54–7. doi: 10.1002/adem.201080076.
    1. Bergman RA, Afifi AK, Heidger PM. Anatomy atlases: atlas of microscopic anatomy: section 4—blood. Plate 4.53: lymphocytes. . Accessed 7 Sept 2016.
    1. Rosenbluth MJ, Lam WA, Fletcher DA. Force microscopy of nonadherent cells: a comparison of leukemia cell deformability. Biophys J. 2006;90:2994–3003. doi: 10.1529/biophysj.105.067496.
    1. Krombach F, Münzing S, Allmeling AM, et al. Cell size of alveolar macrophages: an interspecies comparison. Environ Health Perspect. 1997;105:1261–3. doi: 10.1289/ehp.97105s51261.
    1. Duke University Medical School. Histology learning resources. . Accessed 7 Sept 2016.
    1. Duke University Medical School. Histology learning resources. . Accessed 7 Sept 2016.
    1. Proebstl D, Voisin M-B, Woodfin A, et al. Pericytes support neutrophil subendothelial cell crawling and breaching of venular walls in vivo. J Exp Med. 2012;209:1219–34. doi: 10.1084/jem.20111622.
    1. Lai N, Sims JK, Jeon NL, et al. Adipocyte induction of preadipocyte differentiation in a gradient chamber. Tissue Eng Part C Methods. 2012;18:958–67. doi: 10.1089/ten.tec.2012.0168.
    1. Traktuev DO, Merfeld-Clauss S, Li J, et al. A population of multipotent CD34-positive adipose stromal cells share pericyte and mesenchymal surface markers, reside in a periendothelial location, and stabilize endothelial networks. Circ Res. 2008;102:77–85. doi: 10.1161/CIRCRESAHA.107.159475.
    1. Traktuev DO, Prater DN, Merfeld-Clauss S, et al. Robust functional vascular network formation in vivo by cooperation of adipose progenitor and endothelial cells. Circ Res. 2009;104:1410–20. doi: 10.1161/CIRCRESAHA.108.190926.
    1. Pallua N, Serin M, Wolter TP. Characterisation of angiogenetic growth factor production in adipose tissue-derived mesenchymal cells. J Plast Surg Hand Surg. 2014;48:412–6. doi: 10.3109/2000656X.2014.903196.
    1. Grasys J, Kim B, Pallua N. Content of soluble factors and characteristics of stromal vascular fraction cells in lipoaspirates from different subcutaneous adipose tissue depots. Aesthetic Surg J. 2016;36:831–41. doi: 10.1093/asj/sjw022.
    1. Szöke K, Brinchmann JE. Concise review: therapeutic potential of adipose tissue-derived angiogenic cells. Stem Cells Transl Med. 2012;1:658–67. doi: 10.5966/sctm.2012-0069.
    1. Crisan M, Yap S, Casteilla L, et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell. 2008;3:301–13. doi: 10.1016/j.stem.2008.07.003.
    1. Maumus M, Peyrafitte J-A, D’Angelo R, et al. Native human adipose stromal cells: localization, morphology and phenotype. Int J Obes. 2011;35:1141–53. doi: 10.1038/ijo.2010.269.
    1. Lin C-S, Ning H, Lin G, et al. Is CD34 truly a negative marker for mesenchymal stromal cells? Cytotherapy. 2012;14:1159–63. doi: 10.3109/14653249.2012.729817.
    1. Suga H, Matsumoto D, Eto H, et al. Functional implications of CD34 expression in human adipose–derived stem/progenitor cells. Stem Cells Dev. 2009;18:1201–10. doi: 10.1089/scd.2009.0003.
    1. Sidney LE, Branch MJ, Dunphy SE, et al. Concise review: evidence for CD34 as a common marker for diverse progenitors. Stem Cells. 2014;32:1380–9. doi: 10.1002/stem.1661.
    1. Scherberich A, Di Maggio N, McNagny KM. A familiar stranger: CD34 expression and putative functions in SVF cells of adipose tissue. World J Stem Cells. 2013;5:1–8. doi: 10.4252/wjsc.v5.i1.1.
    1. Keith B, Simon MC. Hypoxia-inducible factors, stem cells, and cancer. Cell. 2007;129:465–72. doi: 10.1016/j.cell.2007.04.019.
    1. Das R, Jahr H, van Osch GJVM, et al. The role of hypoxia in bone marrow-derived mesenchymal stem cells: considerations for regenerative medicine approaches. Tissue Eng Part B Rev. 2010;16:159–68. doi: 10.1089/ten.teb.2009.0296.
    1. Ejtehadifar M, Shamsasenjan K, Movassaghpour A, et al. The effect of hypoxia on mesenchymal stem cell biology. Adv Pharm Bull. 2015;5:141–9. doi: 10.15171/apb.2015.021.
    1. Beegle J, Lakatos K, Kalomoiris S, et al. Hypoxic preconditioning of mesenchymal stromal cells induces metabolic changes, enhances survival, and promotes cell retention in vivo. Stem Cells. 2015;33:1818–28. doi: 10.1002/stem.1976.
    1. Haque N, Rahman MT, Abu Kasim NH, et al. Hypoxic culture conditions as a solution for mesenchymal stem cell based regenerative therapy. Sci World J. 2013;2013:1–12. doi: 10.1155/2013/632972.
    1. Grayson WL, Zhao F, Bunnell B, et al. Hypoxia enhances proliferation and tissue formation of human mesenchymal stem cells. Biochem Biophys Res Commun. 2007;358:948–53. doi: 10.1016/j.bbrc.2007.05.054.
    1. Feng Y, Zhu M, Dangelmajer S, et al. Hypoxia-cultured human adipose-derived mesenchymal stem cells are non-oncogenic and have enhanced viability, motility, and tropism to brain cancer. Cell Death Dis. 2014;5:e1567. doi: 10.1038/cddis.2014.521.
    1. Brunet De La Grange P, Barthe C, Lippert E, et al. Oxygen concentration influences mRNA processing and expression of the CD34 gene. J Cell Biochem. 2006;97:135–44. doi: 10.1002/jcb.20597.
    1. Rigotti G, Marchi A, Galie M, et al. Clinical treatment of radiotherapy tissue damage by lipoaspirate transplant: a healing process mediated by adipose-derived adult stem cells. Plast Reconstr Surg. 2007;119:1409–22. doi: 10.1097/01.prs.0000256047.47909.71.
    1. Prins H-J, Schulten EAJM, ten Bruggenkate CM, et al. Bone regeneration using the freshly isolated autologous stromal vascular fraction of adipose tissue in combination with calcium phosphate ceramics. Stem Cells Transl Med. 2016;5:1362–74. doi: 10.5966/sctm.2015-0369.
    1. Haahr MK, Jensen CH, Toyserkani NM, et al. Safety and potential effect of a single intracavernous injection of autologous adipose-derived regenerative cells in patients with erectile dysfunction following radical prostatectomy: an open-label phase I clinical trial. EBioMedicine. 2016;5:204–10. doi: 10.1016/j.ebiom.2016.01.024.
    1. Tocco I, Widgerow AD, Lalezari S, et al. Lipotransfer: the potential from bench to bedside. Ann Plast Surg. 2014;72:599–609. doi: 10.1097/SAP.0000000000000154.
    1. Doi K, Tanaka S, Iida H, et al. Stromal vascular fraction isolated from lipo-aspirates using an automated processing system: bench and bed analysis. J Tissue Eng Regen Med. 2013;7:864–70. doi: 10.1002/term.1478.
    1. Rigotti G, Charles-de-Sá L, Gontijo-de-Amorim NF, et al. Expanded stem cells, stromal-vascular fraction, and platelet-rich plasma enriched fat: comparing results of different facial rejuvenation approaches in a clinical trial. Aesthet Surg J. 2016;36:261–70. doi: 10.1093/asj/sjv231.
    1. Jackson WM, Nesti LJ, Tuan RS. Concise review: clinical translation of wound healing therapies based on mesenchymal stem cells. Stem Cells Transl Med. 2012;1:44–50. doi: 10.5966/sctm.2011-0024.
    1. Borowski DW, Gill TS, Agarwal AK, et al. Autologous adipose-tissue derived regenerative cells for the treatment of complex cryptoglandular fistula-in-ano: a report of three cases. BMJ Case Rep. 2012;2012:4–7. doi: 10.1136/bcr-2012-006988.
    1. Riordan NH, Ichim TE, Min W-P, et al. Non-expanded adipose stromal vascular fraction cell therapy for multiple sclerosis. J Transl Med. 2009;7:29. doi: 10.1186/1479-5876-7-29.
    1. Lee HC, An SG, Lee HW, et al. Safety and effect of adipose tissue-derived stem cell implantation in patients with critical limb ischemia. Circ J. 2012;76:1750–60. doi: 10.1253/circj.CJ-11-1135.
    1. Parcero JJ, Perez JA, Patel AN, et al. Autologous adipose-derived stromal stem cell implantation to resolve critical limb ischemia: case report. Cureus. 2014;6(5):e182. doi:10.7759/cureus.182. .
    1. Pers Y-M, Rackwitz L, Ferreira R, et al. Adipose mesenchymal stromal cell-based therapy for severe osteoarthritis of the knee: a phase I dose-escalation trial. Stem Cells Transl Med. 2016;5:847–56. doi: 10.5966/sctm.2015-0245.
    1. Domergue S, Bony C, Maumus M, et al. Comparison between stromal vascular fraction and adipose mesenchymal stem cells in remodeling hypertrophic scars. PLoS One. 2016;11:e0156161. doi: 10.1371/journal.pone.0156161.
    1. Frazier TP, Bowles A, Lee S, et al. Serially transplanted nonpericytic CD146– adipose stromal/stem cells in silk bioscaffolds regenerate adipose tissue in vivo. Stem Cells. 2016;34:1097–111. doi: 10.1002/stem.2325.
    1. Lin S-D, Huang S-H, Lin Y-N, et al. Injected implant of uncultured stromal vascular fraction loaded onto a collagen gel. Ann Plast Surg. 2016;76:S108–16. doi: 10.1097/SAP.0000000000000687.
    1. NEWSmax. Unregulated stem cell industry is “wild west.” NEWSmax; 2015. . Accessed 7 Sept 2016.
    1. McFarling UL. FDA moves to crack down on unproven stem cell therapies. STAT News. 2016. . Accessed 7 Sept 2016.
    1. Jabr F. In the flesh: the embedded dangers of untested stem cell cosmetics. Sci Am. 2012. Available at . Accessed 7 Sept 2016.
    1. Wilson C. Stem cell treatment causes nasal growth in woman’s back. New Sci. 2014. . Accessed 7 Sept 2016.
    1. Cyranoski D. Korean deaths spark inquiry. Nature. 2010;468:485. doi: 10.1038/468485a.
    1. McLean AK, Stewart C, Kerridge I. Untested, unproven, and unethical: the promotion and provision of autologous stem cell therapies in Australia. Stem Cell Res Ther. 2015;6:12. doi: 10.1186/s13287-015-0047-8.
    1. Ledford H. Boom in unproven cell therapies intensifies regulatory debate. Nature. 2016;537:148. doi: 10.1038/537148a.
    1. US Department of Health and Human Services (Food and Drug Administration). Human cells, tissues, and cellular- and tissue-based products (HCT/Ps) from adipose tissue : regulatory considerations; draft guidance. . Accessed 7 Sept 2016.
    1. Turner L, Knoepfler P. Selling stem cells in the USA: assessing the direct-to-consumer industry. Cell Stem Cell. 2016;19:154–7. doi: 10.1016/j.stem.2016.06.007.
    1. Kirk MS, Manchin J, Collins SM. REGROW Act. 2016. . Accessed 7 Sept 2016.
    1. Research MJFF for P. MJFF signs letter opposing the REGROW Act 2016. . Accessed 7 Sept 2016.
    1. International Society for Cellular Therapy. ISCT calls for changes to proposed US REGROW Act on cell therapies 2016. . Accessed 7 Sept 2016.
    1. The Alliance for Regenerative Medicine. ARM statement in response to U.S. Senator Kirk’s REGROW Act. . Accessed 7 Sept 2016.
    1. Knoepfler P. California stem-cell institute’s political gamble. San Fr Chron 2016. . Accessed 7 Sept 2016.
    1. Joseph A. Drive to get more patients experimental stem cell treatments stirs concern. STAT News 2016. . Accessed 7 Sept 2016.
    1. Hass R, Kasper C, Böhm S, et al. Different populations and sources of human mesenchymal stem cells (MSC): a comparison of adult and neonatal tissue-derived MSC. Cell Commun Signal. 2011;9:12. doi: 10.1186/1478-811X-9-12.

Source: PubMed

3
Iratkozz fel