Skip to main content

Isolation and Characterization of Adipose-Derived Stromal Cells

  • Chapter
  • First Online:
Stem Cell Processing

Abstract

Methods for adipose-derived stromal cell (ASC) isolation, characterization and the respective data generated differ between different research groups. Laboratories have developed in-house methods to isolate, expand and differentiate ASCs, which often makes data comparison difficult.

Methods for both research laboratory isolation and clinical applications are explored in this chapter. To use ASCs in the clinical setting, stem cell therapy products need to be prepared using good manufacturing practices (GMPs). This is achieved by eliminating animal-derived products from the manufacturing process. As a result, several human and chemically defined alternatives have been explored, especially during the isolation and expansion process.

ASC characterization criteria remain open ended, but should be compliant within the parameters of (1) adherence to plastic, (2) expression of a defined set of surface antigens and (3) the capacity to differentiate into various cell types.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  • Akita S, Yoshimoto H, Ohtsuru A et al (2012) Autologous adipose-derived regenerative cells are effective for chronic intractable radiation injuries. Radiat Prot Dosimetry 151:656–660

    Article  CAS  PubMed  Google Scholar 

  • Aktas M, Radke TF, Strauer BE et al (2008) Separation of adult bone marrow mononuclear cells using the automated closed separation system Sepax. Cytotherapy 10:203–211

    Article  CAS  PubMed  Google Scholar 

  • Aldridge A, Kouroupis D, Churchman S et al (2013) Assay validation for the assessment of adipogenesis of multipotential stromal cells-a direct comparison of four different methods. Cytotherapy 15:89–101

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Ali AT, Penny CB, Paiker JE et al (2006) The relationship between alkaline phosphatase activity and intracellular lipid accumulation in murine 3T3-L1 cells and human preadipocytes. Anal Biochem 354:247–254

    Article  CAS  PubMed  Google Scholar 

  • Atashi F, Modarressi A, Pepper MS (2015) The role of reactive oxygen species in mesenchymal stem cell adipogenic and osteogenic differentiation: a review. Stem Cells Dev 4:1150–1163

    Article  CAS  Google Scholar 

  • Azouna NB, Jenhani F, Regaya Z et al (2012) Phenotypical and functional characteristics of mesenchymal stem cells from bone marrow: comparison of culture using different media supplemented with human platelet lysate or fetal bovine serum. Stem Cell Res Ther 3:6

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Baer PC, Griesche N, Luttmann W et al (2010) Human adipose-derived mesenchymal stem cells in vitro: evaluation of an optimal expansion medium preserving stemness. Cytotherapy 12:96–106

    Article  CAS  PubMed  Google Scholar 

  • Baer PC (2014) Adipose derived mesenchymal stromal/stem cell: An update on their phenotype in vivo and in vitro. World J Stem Cells 6(3):256–265

    Article  PubMed  PubMed Central  Google Scholar 

  • Banyard DA, Salibian AA, Widgerow AD et al (2015) Implications for human adiposederived stem cells in plastic surgery. J Cell Mol Med 19(1):21–30

    Article  PubMed  Google Scholar 

  • Beier JP, Bitto FF, Lange C et al (2011) Myogenic differentiation of mesenchymal stem cells co-cultured with primary myoblasts. Cell Biol Int 35:397–406

    Article  CAS  PubMed  Google Scholar 

  • Bernardo ME, Avanzini MA, Perotti C et al (2006) Optimization of in vitro expansion of human multipotent mesenchymal stromal cells for cell-therapy approaches: Further insights in the search for a fetal calf serum substitute. J Cell Physiol 211(1):121–130

    Article  CAS  Google Scholar 

  • Bernardo ME, Cometa AM, Pagliara D et al (2011) Ex vivo expansion of mesenchymal stromal cells. Best Pract Res Clin Haematol 24(1):73–81

    Article  CAS  PubMed  Google Scholar 

  • Bieback K, Hecker A, Kocaömer A et al (2009) Human alternatives to fetal bovine serum for the expansion of mesenchymal stromal cells from bone marrow. Stem Cells 27(9):2331–2341

    Article  CAS  PubMed  Google Scholar 

  • Bieback K, Hecker A, Schlechter T et al (2012) Replicative aging and differentiation potential of human adipose tissue-derived mesenchymal stromal cells expanded in pooled human or fetal bovine serum. Cytotherapy 14:570–583

    Article  CAS  PubMed  Google Scholar 

  • Bitto FF, Klumpp D, Lange C et al (2013) Myogenic differentiation of mesenchymal stem cells in a newly developed neurotised av-loop model. BioMed Res Int 2013:935046

    PubMed  PubMed Central  Google Scholar 

  • Both SK, van der Muijsenberg AJ, van Bitterswijk CA (2007) A rapid and efficient method for expansion of human mesenchymal stem cells. Tissue Eng 13(1):3–9

    Article  CAS  PubMed  Google Scholar 

  • Bourin P, Bunnell BA, Casteilla L et al (2013) 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 So. Cytotherapy 15:641–648

    Article  PubMed  PubMed Central  Google Scholar 

  • Bowers RR, Lane MD (2008) Wnt signaling and adipocyte lineage commitment. Cell Cycle 7:1191–1196

    Article  CAS  PubMed  Google Scholar 

  • Brasaemle DL, Rubin B, Harten IA et al (2000) Perilipin A increases triacylglycerol storage by decreasing the rate of triacylglycerol hydrolysis. J Biol Chem 275(49):38486–38493

    Article  CAS  PubMed  Google Scholar 

  • Brzoska M, Geiger H, Gauer S et al (2005) Epithelial differentiation of human adipose tissue-derived adult stem cells. Biochem Biophys Res Commun 330(1):142–150

    Article  CAS  PubMed  Google Scholar 

  • Bunnell BA, Flaat M, Gagliardi C et al (2008) Adipose-derived stem cells: isolation, expansion and differentiation. Methods 45:115–120

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Cao J (2011) Effects of obesity on bone metabolism. J Orthop Surg Res 6:30

    Article  PubMed  PubMed Central  Google Scholar 

  • Cao F, Niu LL, Meng L et al (2004) Cardiomyocyte-like differentiation of human bone marrow mesenchymal stem cells after exposure of 5-azacytidine in vitro. Shi Yan Sheng Wu Xue Bao 37:118–124

    PubMed  Google Scholar 

  • Caplan AI (2009) Why are MSCs therapeutic? New data: new insight. J Pathol 217:318–324

    Article  CAS  PubMed  Google Scholar 

  • Carvalho PH, Daibert APF, Monteiro BS et al (2013) Diferenciação de células-tronco mesenquimais derivadas do tecido adiposo em cardiomiócitos. Arq Bras Cardiol 100:82–89

    Article  CAS  PubMed  Google Scholar 

  • Ceppo F, Berthou F, Jager J et al (2014) Implication of the Tpl2 kinase in inflammatory changes and insulin resistance induced by the interaction between adipocytes and macrophages. Endocrinology 155:951–964

    Article  PubMed  CAS  Google Scholar 

  • Chazenbalk G, Bertolotto C, Heneidi S et al (2011) Novel pathway of adipogenesis through cross-talk between adipose tissue macrophages, adipose stem cells and adipocytes: evidence of cell plasticity. PLoS One 6(3), e17834. doi:10.1371/journal.pone.0017834

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Chen HH, Decot V, Ouyang JP et al (2009) In vitro initial expansion of mesenchymal stem cells is influenced by the culture parameters used in the isolation process. Biomed Mater Eng 19:301–309

    CAS  PubMed  Google Scholar 

  • Cheng SL, Yang JW, Rifas L et al (1994) Differentiation of human bone marrow osteogenic stromal cells in vitro: induction of the osteoblast phenotype by dexamethasone. Endocrinology 134:277–286

    CAS  PubMed  Google Scholar 

  • Chieregato K, Castegnaro S, Madeo D et al (2011) Epidermal growth factor, basic fibroblast growth factor and platelet-derived growth factor-bb can substitute for fetal bovine serum and compete with human platelet-rich plasma in the ex vivo expansion of mesenchymal stromal cells derived from adipose tissue. Cytotherapy 13(8):933–943

    Article  CAS  PubMed  Google Scholar 

  • Chiou M, Xu Y, Longaker MT (2006) Mitogenic and chondrogenic effects of fibroblast growth factor-2 in adipose-derived mesenchymal cell. Biochem Biophys Res Commun 343:644–652

    Article  CAS  PubMed  Google Scholar 

  • Choi YS, Vincent LG, Lee AR et al (2012) Mechanical derivation of functional myotubes from adipose-derived stem cells. Biomaterials 33:2482–2491

    Article  CAS  PubMed  Google Scholar 

  • Crespo-Diaz R, Behfar A, Butler GW et al (2011) Platelet lysate consisting of a natural repair proteome supports human mesenchymal stem cell proliferation and chromosomal stability. Cell Transplant 20:797–811

    Article  PubMed  Google Scholar 

  • Dominici M, Le Blanc K, Mueller I et al (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8:315–317

    Article  CAS  PubMed  Google Scholar 

  • Donnenberg AD, Meyer EM, Rubin JP et al (2015) The cell-surface proteome of cultured adipose stromal cells. Cytometry 87(7):665–674

    Article  CAS  PubMed  Google Scholar 

  • Doucet C, Ernou I, Zhang Y et al (2005) Platelet lysates promote mesenchymal stem cell expansion: a safety substitute for animal serum in cell-based therapy applications. J Cell Physiol 205:228–236. doi:10.1002/jcp.20391

    Article  CAS  PubMed  Google Scholar 

  • Ersek RA, Salisbury AV (1995) Circumferential liposuction of knees, calves and ankles. Aesthetic Plast Surg 19:321–333

    Article  Google Scholar 

  • Estes BT, Diekman BO, Gimble JM, Guilak F (2010) Isolation of adipose-derived stem cells and their induction to a chondrogenic phenotype. Nat Protoc 5:1294–1311

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Fei W, Du X, Yang H (2011) Seipin, adipogenesis and lipid droplets. Trends Endocrinol Metab 22:204–210. doi:10.1016/j.tem.2011.02.004

    Article  CAS  PubMed  Google Scholar 

  • Fell HB (1925) The histogenesis of cartilage and bone in the long bones of the embryonic fowl. J Morphol 40:417–459

    Article  Google Scholar 

  • Fink T, Abildtrup L, Fogd K et al (2004) Induction of adipocyte-like phenotype in human mesenchymal stem cells by hypoxia. Stem Cells 22:1346–1355

    Article  PubMed  CAS  Google Scholar 

  • Fiorentini E, Granchi D, Leonardi E et al (2011) Effects of osteogenic differentiation inducers on in vitro expanded adult mesenchymal stromal cells. Int J Artif Organs 34:998–1011

    Article  CAS  PubMed  Google Scholar 

  • Fossett E, Khan WS, Longo UG et al (2012) Effect of age and gender on cell proliferation and cell surface characterization of synovial fat pad derived mesenchymal stem cells. J Orthop Res 30:1013–1018

    Article  PubMed  Google Scholar 

  • Fournier PF, Otteni FM (1983) Lipodissection in body sculpturing: the dry procedure. Plast Reconstr Surg 72:598–609

    Article  CAS  PubMed  Google Scholar 

  • Fowler SD, Greenspan P (1985) Application of Nile red, a fluorescent hydrophobic probe, for the detection of neutral lipid deposits in tissue sections: comparison with oil red O. J Histochem Cytochem 33:833–836

    Article  CAS  PubMed  Google Scholar 

  • Fraser JK, Wulur I, Alfonso Z et al (2006) Fat tissue: an underappreciated source of stem cells for biotechnology. Trends Biotechnol 24:150–154

    Article  CAS  PubMed  Google Scholar 

  • Freshney RI (2006) Basic principles of cell culture. Culture of cells for tissue engineering. Wiley, New Jersey. doi:10.1002/0471741817.ch1

    Google Scholar 

  • Freshney RI (2010) Culture of animal cells: a manual of basic technique and specialized applications, 6th edn. Wiley, New York

    Book  Google Scholar 

  • Gang EJ, Jeong JA, Hong SH et al (2004) Skeletal myogenic differentiation of mesenchymal stem cells isolated from human umbilical cord blood. Stem Cells 22:617–624

    Article  PubMed  Google Scholar 

  • Gimble JM, Adam JK, Bruce AB (2007) Adipose-derived stem cells for regenerative medicine. Circ Res 100:1249–1260

    Article  CAS  PubMed  Google Scholar 

  • Gimble JM, Bunnell BA, Chiu ES et al (2011) Concise review: adipose-derived stromal vascular fraction cells and stem cells: let’s not get lost in translation. Stem Cells 29:749–754

    Article  PubMed  Google Scholar 

  • Gocze PM, Freeman D (1994) Factors underlying the variability of lipid droplet fluorescence in MA-10 Leydig tumor cells. Cytometry 17:151–158

    Article  CAS  PubMed  Google Scholar 

  • Greenspan P, Mayer EP, Fowler SD (1985) Nile red: a selective fluorescent stain for intracellular lipid droplets. J Cell Biol 100:965–973

    Article  CAS  PubMed  Google Scholar 

  • Guo Y, Cordes KR, Farese RV et al (2009) Lipid droplets at a glance. J Cell Sci 122:749–752

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Guven S, Karagianni M, Schwalbe M et al (2012) Validation of an automated procedure to isolate human adipose tissue-derived cells by using the Sepax(R) technology. Tissue Eng Part C Methods 18:575–582

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Hicok KC, Hedrick MH (2011) Automated isolation and processing of adipose-derived stem and regenerative cells. Methods Mol Biol 702:87–105

    Article  CAS  PubMed  Google Scholar 

  • Houtgraaf JH, den Dekker WK, van Dalen BM et al (2012) First experience in humans using adipose tissue-derived regenerative cells in the treatment of patients with ST-segment elevation myocardial infarction. J Am Coll Cardiol 59:539–540

    Article  PubMed  Google Scholar 

  • Huang H, Song TJ, Li X et al (2009) BMP signaling pathway is required for commitment of C3H10T1/2 pluripotent stem cells to the adipocyte lineage. Proc Natl Acad Sci U S A 106:12670–12675

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Ichinose S, Tagami M, Muneta T et al (2005) Morphological examination during in vitro cartilage formation by human mesenchymal stem cells. Cell Tissue Res 322:217–226

    Article  PubMed  Google Scholar 

  • Jeon ES, Song HY, Kim MR et al (2006) Sphingosylphosphorylcholine induces proliferation of human adipose tissue-derived mesenchymal stem cells via activation of JNK. J Lipid Res 47:653–664

    Article  CAS  PubMed  Google Scholar 

  • Johnstone B, Hering TM, Caplan AI et al (1998) In vitro chondrogenesis of bone marrow-derived mesenchymal progenitor cells. Exp Cell Res 238:265–272

    Article  CAS  PubMed  Google Scholar 

  • Jones A, Kinsey SE, English A et al (2002) Isolation and characterisation of bone marrow multipotential mesenchymal progenitor cells. Arthritis Rheum 46:3349–3360

    Article  PubMed  Google Scholar 

  • Joo S, Lim HJ, Jackson JD et al (2014) Myogenic-induced mesenchymal stem cells are capable of modulating the immune response by regulatory T cells. J Tissue Eng 5:2041731414524758

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Kang YJ, Jeon ES, Song HY et al (2005) Role of c-Jun N-terminal kinase in the PDGF-induced proliferation and migration of human adipose tissue-derived mesenchymal stem cells. J Cell Biochem 95:1135–1145

    Article  CAS  PubMed  Google Scholar 

  • Kang S, Bennett CN, Gerin I et al (2007) Wnt signaling stimulates osteoblastogenesis of mesenchymal precursors by suppressing CCAAT/enhancer-binding protein α and peroxisome proliferator-activated receptor γ. J Biol Chem 282:14515–14524

    Article  CAS  PubMed  Google Scholar 

  • Kern S, Eichler H, Stoeve J et al (2006) Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells 24:1294–1301

    Article  CAS  PubMed  Google Scholar 

  • Kocaoemer A, Kern S, Klüter H et al (2007) Human AB serum and thrombin-activated platelet-rich plasma are suitable alternatives to fetal calf serum for the expansion of mesenchymal stem cells from adipose tissue. Stem Cells 25:1270–1278

    Article  CAS  PubMed  Google Scholar 

  • Koellensperger E, Bollinger N, Dexheimer V et al (2014) Choosing the right type of serum for different applications of human adipose tissue-derived stem cells: influence on proliferation and differentiation abilities. Cytotherapy 16:789–799

    Article  CAS  PubMed  Google Scholar 

  • Kølle ST, Oliveri RS, Glovinski PV et al (2013) Pooled human lysate versus fetal bovine serum—Investigating the proliferation rate, chromosome stability and angiogenic potential of human adipose tissueederived stem cells intended for clinical use. Cytotherapy 15(9):1086–1097

    Article  CAS  Google Scholar 

  • Krahmer N, Guo Y, Farese RV et al (2009) SnapShot: lipid droplets. Cell 139:10–11

    Article  CAS  Google Scholar 

  • Kyllonen L, Haimi S, Mannerstrom B et al (2013) Effects of different serum conditions on osteogenic differentiation of human adipose stem cells in vitro. Stem Cell Res Ther 4:1–17

    Article  CAS  Google Scholar 

  • Lennon DP, Haynesworth SE, Young RG et al (1995) A chemically defined medium supports in vitro proliferation and maintains the osteochondral potential of rat marrow-derived mesenchymal stem cells. Exp Cell Res 219:211–222

    Article  CAS  PubMed  Google Scholar 

  • Lennon DP, Haynesworth SE, Bruder SP et al (1996) Human and animal mesenchymal progenitor cells from bone marrow: identification of serum for optimal selection and proliferation. Vitro Cell Dev Biol Animal 32(10):602–611

    Article  Google Scholar 

  • Lo Surdo JL, Millis B, Bauer SR (2013) Automated microscopy as a quantitative method to measure differences in adipogenic differentiation in preparations of human mesenchymal stromal cells. Cytotherapy 15:1527–1540

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Lode A, Bernhardt A, Gelinsky M (2008) Cultivation of human bone marrow stromal cells on three-dimensional scaffolds of mineralized collagen: influence of seeding density on colonization, proliferation and osteogenic differentiation. J Tissue Eng Regen Med 2:400–407

    Article  CAS  PubMed  Google Scholar 

  • Lu X, Alshemali S, de Wynter EA et al (2010) Mesenchymal stem cells from CD34(-) human umbilical cord blood. Transfus Med 20:178–184

    Google Scholar 

  • Lund P, Pilgaard L, Duroux M et al (2009) Effect of growth media and serum replacements on the proliferation and differentiation of adipose-derived stem cells. Cytotherapy 11:189–197

    Article  CAS  PubMed  Google Scholar 

  • McNeil M, Daffe M, Brennan PJ (1991) Location of the mycolyl ester substituents in the cell walls of mycobacteria. J Biol Chem 266:13217–13223

    CAS  PubMed  Google Scholar 

  • Menssen A, Häupl T, Sittinger M et al (2011) Differential gene expression profiling of human bone marrowderived mesenchymal stem cells during adipogenic development. BMC Genomics 12:461–477

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Mizuno H (2009) Adipose-derived stem cells for tissue repair and regeneration: ten years of research and a literature review. J Nippon Med Sch 76:56–66

    Article  PubMed  Google Scholar 

  • Müller I, Kordowich S, Holzwarth C et al (2006) Animal serum-free culture conditions for isolation and expansion of multipotent mesenchymal stromal cells from human BM. Cytotherapy 8:437–444

    Article  PubMed  Google Scholar 

  • Naumann A, Dennis JE, Awadallah A et al (2002) Immunochemical and mechanical characterization of cartilage subtypes in rabbit. J Histochem Cytochem 50:1049–1058

    Article  CAS  PubMed  Google Scholar 

  • Ning H, Lin G, Lue TF et al (2006) Neuron-like differentiation of adipose tissue-derived stromal cells and vascular smooth muscle cells. Differentiation 74:510–518

    Article  CAS  PubMed  Google Scholar 

  • Ntambi JM, Young-Cheul K (2000) Adipocyte differentiation and gene expression. J Nutr 130:3122S–3126S

    CAS  PubMed  Google Scholar 

  • Nuttall ME, Shah F, Singh V et al (2014) Adipocytes and the regulation of bone remodeling: A balancing act. Calcif Tissue Int 94(1):78–87

    Article  CAS  PubMed  Google Scholar 

  • Patrikoski M, Juntunen M, Boucher S et al (2013) Development of fully defined xeno-free culture system for the preparation and propagation of cell therapy compliant human adipose stem cells. Stem Cell Res Ther 4:1–15

    Article  CAS  Google Scholar 

  • Rajala K, Lindroos B, Hussein SM et al (2010) A defined and xeno-free culture method enabling the establishment of clinical-grade human embryonic, induced pluripotent and adipose stem cells. PLoS One 5:1–14

    Google Scholar 

  • Ramírez-Zacarías JL, Castro-Muñozledo F, Kuri-Harcuch W (1992) Quantitation of adipose conversion and triglycerides by staining intracytoplasmic lipids with oil red O. Histochem Cell Biol 97:493–497

    Google Scholar 

  • Rodbell M (1966) Metabolism of isolated fat cells II. The similar effects of phospholipase c (clostridium perfringens alpha toxin) and of insulin on glucose and amino acid metabolism. J Biol Chem 241:130–139

    CAS  PubMed  Google Scholar 

  • Rodbell M, Jones AB (1966) Metabolism of isolated fat cells. III. The similar inhibitory action of phospholipase c (clostridium perfringens alpha toxin) and of insulin on lipolysis stimulated by lipolytic hormones and theophylline. J Biol Chem 241:140–142

    CAS  PubMed  Google Scholar 

  • Romagnoli C, Brandi ML (2014) Adipose mesenchymal stem cells in the field of bone tissue engineering. World J Stem Cells 6(2):144–152

    Article  PubMed  PubMed Central  Google Scholar 

  • Rosen CJ, Bouxsein ML (2006) Mechanisms of disease: is osteoporosis the obesity of bone? Nat Clin Pract Rheumatol 2(1):35–43

    Article  CAS  PubMed  Google Scholar 

  • Rubin CS, Hirsch A, Fung C et al (1978) Development of hormone receptors and hormonal responsiveness in vitro. Insulin receptors and insulin sensitivity in the preadipocyte and adipocyte forms of 3T3-L1 cells. J Biol Chem 253:7570–7578

    CAS  PubMed  Google Scholar 

  • Russell TR, Ho R (1976) Conversion of 3T3 fibroblasts into adipose cells: triggering of differentiation by prostaglandin F2alpha and 1-methyl-3-isobutyl xanthine. Proc Natl Acad Sci U S A 73:4516–4520

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Schaedlich K, Knelangen JM, Santos AN et al (2010) A simple method to sort ESC-derived adipocytes. Cytometry 77A:990–995

    Article  CAS  Google Scholar 

  • Schallmoser K, Bartmann C, Rohde E et al (2007) Human platelet lysate can replace fetal bovine serum for clinical-scale expansion of functional mesenchymal stromal cells. Transfusion 47:1436–1446

    Article  CAS  PubMed  Google Scholar 

  • Schallmoser K, Bartmann C, Rohde E et al (2010) Replicative senescence-associated gene expression changes in mesenchymal stromal cells are similar under different culture conditions. Haematologica 95(6):867–874

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Sekiya I, Colter DC, Prockop DJ (2001) BMP-6 enhances chondrogenesis in a subpopulation of human marrow stromal cells. Biochem Biophys Res Commun 284:411–418

    Article  CAS  PubMed  Google Scholar 

  • Sekiya I, Larson BL, Vuoristo JT et al (2005) Comparison of effect of BMP-2, −4, and −6 on in vitro cartilage formation of human adult stem cells from bone marrow stroma. Cell Tissue Res 320(2):269–276

    Article  CAS  PubMed  Google Scholar 

  • Smyth MJ, Wharton W (1992) Differentiation of A31T6 preadipocytes to adipocytes: a flow cytometric analysis. Exp Cell Res 199:29–38

    Article  CAS  PubMed  Google Scholar 

  • Song HY, Jeon ES, Jung JS et al (2005) Oncostatin M induces proliferation of human adipose tissue derived mesenchymal stem cells. Int J Biochem Cell Biol 37:2357–2365

    Article  CAS  PubMed  Google Scholar 

  • Sotiropoulou PA, Perez SA, Salagianni M et al (2006) Characterization of the optimal culture conditions for clinical scale production of human mesenchymal stem cells. Stem Cells 24:462–471

    Article  PubMed  Google Scholar 

  • Spiegelman BM, Green H (1981) Cyclic AMP-mediated control of lipogenic enzyme synthesis during adipose differentiation of 3T3 cells. Cell 24:503–510

    Article  CAS  PubMed  Google Scholar 

  • Stedman TL (2006) Stedman’s medical dictionary. Lippincott Williams & Wilkins, Baltimore, MD

    Google Scholar 

  • Stern-Straeter J, Bonaterra GA, Juritz S et al (2014) Evaluation of the effects of different culture media on the myogenic differentiation potential of adipose tissue- or bone marrow-derived human mesenchymal stem cells. Int J Mol Med 33:160–170

    CAS  PubMed  Google Scholar 

  • Strem BM, Hicok KC, Zhu M et al (2005) Multipotential differentiation of adipose tissue-derived stem cells. Keio J Med 54:132–141

    Article  CAS  PubMed  Google Scholar 

  • Stute N, Holtz K, Bubenheim M et al (2004) Autologous serum for isolation and expansion of human mesenchymal stem cells for clinical use. Exp Hematol 32:1212–1225

    Article  CAS  PubMed  Google Scholar 

  • Tang QQ, Lane MD (2012) Adipogenesis: from stem cell to adipocyte. Annu Rev Biochem 81:715–736

    Article  CAS  PubMed  Google Scholar 

  • Tang QQ, Otto TC, Lane MD (2004) Commitment of C3H10T1/2 pluripotent stem cells to the adipocyte lineage. Proc Natl Acad Sci U S A 101(26):9607–9611

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Van Der Valk J, Mellor D, Brands R et al (2004) The humane collection of fetal bovine serum and possibilities for serum-free cell and tissue culture. Toxicol In Vitro 18(1):1–12

    Article  PubMed  CAS  Google Scholar 

  • Van Der Valk J, Brunner D, De Smet K et al (2010) Optimization of chemically defined cell culture media—replacing fetal bovine serum in mammalian in vitro methods. Toxicol In Vitro 24(4):1053–1063

    Article  PubMed  CAS  Google Scholar 

  • van Vollenstee FA, Jackson C, Hoffmann D et al (2016) Human adipose derived mesenchymal stromal cells transduced with GFP lentiviral vectors: assessment of immunophenotype and differentiation capacity in vitro. Cytotechnology. doi:10.1007/s10616-016-9945-6

    PubMed  PubMed Central  Google Scholar 

  • Von Heimburg D, Hemmrich K, Haydarlioglu S et al (2004) Comparison of viable cell yield from excised versus aspirated adipose tissue. Cells Tissues Organs 178:87–92

    Article  Google Scholar 

  • Yang S, Pilgaard L, Chase LG et al (2012) Defined xenogeneic-free and hypoxic environment provides superior conditions for long-term expansion of human adipose-derived stem cells. Tissue Eng Part C Methods 18:593–602

    Article  CAS  PubMed  Google Scholar 

  • Zimmerlin L, Donnenberg VS, Rubin JP et al (2013) Mesenchymal markers on human adipose stem/progenitor cells. Cytometry 83(1):134–140

    Article  PubMed  CAS  Google Scholar 

  • Zingsem J, Strasser E, Weisbach V et al (2003) Cord blood processing with an automated and functionally closed system. Transfusion 43:806–813

    Article  PubMed  Google Scholar 

  • Zinno F, Landi F, Scerpa MC et al (2011) Processing of hematopoietic stem cells from peripheral blood before cryopreservation: use of a closed automated system. Transfusion 51:2656–2663

    Article  PubMed  Google Scholar 

  • Zuk PA, Zhu M, Mizuno H et al (2001) Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng 7:211–228

    Article  CAS  PubMed  Google Scholar 

  • Zuk PA, Zhu M, Ashjian P et al (2002) Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell 13:4279–4295

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Zuscik MJ, Hilton MJ, Zhang X et al (2008) Regulation of chondrogenesis and chondrocyte differentiation by stress. J Clin Invest 118:429–438

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Zych J, Stimamiglio MA, Senegaglia AC et al (2013) The epigenetic modifiers 5-aza-2’-deoxycytidine and trichostatin A influence adipocyte differentiation in human mesenchymal stem cells. Braz J Med Biol Res 46:405–416

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Michael S. Pepper .

Editor information

Editors and Affiliations

Appendix: Brief Description of Current Methods

Appendix: Brief Description of Current Methods

7.1.1 Manual Isolation Procedure

The isolation protocol is adapted from the procedures described by Zuk et al. (2001), Bunnell et al. (2008) and Estes et al. (2010) (Fig. A.1).

Fig. A.1
figure 6

Outline of the procedure used f or isolating adipose-derived stromal cells. Lipoaspirate samples are processed by enzymatic digestion, and the stromal vascular fraction (SVF) is collected. Adipose-derived stromal cells from the SVF adhere to the plastic culture dish, and non-adherent cells are washed away after 24 h

The method requires the transfer of lipoaspirate material into 50 ml tubes (30 ml lipoaspirate/tube), followed by the addition of 20 ml phosphate buffer saline (PBS), supplemented with antibiotics (Fig. A.1). Depending on the future application of the adipose-derived stromal cell (ASC) isolates, culture medium may contain either FBS or human derivatives and antibiotics, usually penicillin and streptomycin (pen/strep). The adipose tissue is separated from peripheral blood contaminants by centrifugation at 1660 g for 3 min. The top oil layer is aspirated with a suction-assisted glass pipette system, and the compact lipoaspirate is carefully transferred to a sterile 50 ml tube. It is recommended that the washing steps are repeated until the compacted lipoaspirate material is golden yellow in colour without any visible evidence of peripheral blood contamination. The volume of the compacted lipoaspirate material should be recorded once most of the blood contamination has been removed.

To release resident ASCs in adipose tissue from the fibrous network, the lipoaspirate is enzymatically digested in culture plates. The most popular approach makes use of the enzyme, collagenase type I. Other enzymatic alternatives include dispase and trypsin. A filter-sterilized 0.1 % collagenase type I solution is prepared using PBS supplemented with 2 % antibiotics. The volume of collagen solution required is dependent on the volume of the compacted adipose tissue previously recorded. The ratio of compacted adipose tissue volume to the volume of collagen solution should be at least 2:1, meaning that the final volume of collagen digesting solution added to the adipose tissue in the culture plates should be half that of the washed compacted adipose tissue volume. The ratio of collagenase to adipose tissue should be optimized by each laboratory as well as for each isolation technique used. A sterile plastic pipette is used to mix the adipose tissue well, before incubation at 37 °C, 5 % CO2 for 45 min. The sample may either be continuously agitated using an automated rotating system or agitated manually every 15 min with a plastic pipette to aid the mechanical breakdown of the fibrous tissue.

The collagenase-digested sample is then transferred to sterile 50 ml tubes. The tubes are shaken vigorously and centrifuged at 500 g for 5 min, resulting in the SVF pellet settling to the bottom of the tube. The compacted adipose tissue and collagenase solution supernatants are carefully aspirated, and the collagenase activity is neutralized by adding 2 ml of stromal medium to the AD-SVF pellet. Stromal medium may consist of either Dulbecco’s Modified Eagle Medium (DMEM) containing GlutaMax™, 4.5 g/L D-glucose and pyruvate or alpha-Modified Eagle Medium (α-MEM) containing GlutaMax™, supplemented with 10 % serum and 1 % antibiotics.

The AD-SVF pellets are pooled into a single tube (15 or 50 ml) and centrifuged at 265 g for 5 min, followed by aspiration of the supernatant. Red blood cells present in the AD-SVF pellet are lysed either by the addition of an ammonium chloride-based lysing solution or an enzymatic-based lysing solution like VersaLyse™ (Beckman Coulter, Miami, USA). After a 10–15 min incubation period at room temperature, the lysing reaction is stopped by filling the tube with PBS supplemented with antibiotics and centrifuged at 184 g for 5 min. The supernatant is removed and the pellet resuspended in stromal medium before filtering the cellular suspension through a 70 μm cell strainer to remove any larger, undigested fragments.

In order to seed the cells at the correct seeding density, an absolute cell count should be performed. Cell counts may be performed by using either a manual approach in which the cells are counted using a haemocytometer or an automated cell counting device such as a flow cytometer. Details are provided below.

The AD-SVF is seeded at an initial seeding density of 5 × 105 cells per cm2. In order to determine the volume of cell suspension required for initial seeding, the following formula should be used:

$$ \begin{array}{l}\mathrm{Volume}\ \left(\mu \mathrm{l}\right)=\left(\frac{\mathrm{initial}\ \mathrm{seeding}\ \mathrm{density}\times \mathrm{seeding}\ \mathrm{surface}\ \mathrm{area}\ \mathrm{of}\ \mathrm{tissue}\ \mathrm{culture}\ \mathrm{flask}\ \mathrm{or}\ \mathrm{well}}{\mathrm{total}\ \mathrm{number}\ \mathrm{of}\ \mathrm{viable}\ \mathrm{cells}}\right)\\ {}\kern8.25em \times \mathrm{cell}\ \mathrm{suspension}\ \mathrm{volume}\ \mathrm{in}\ \mu \mathrm{l}\end{array} $$

After the cells are seeded, the cultures flasks are swirled gently to ensure uniform distribution. The culture flasks are maintained in an incubator under standard cell culture conditions (humidity, 5 % CO2 and 37 °C). The cultures are washed twice after 24 h with PBS supplemented with antibiotics to remove non-adherent cells, cellular debris and non-viable cells. Fresh stromal medium is added to the culture flasks and incubated under standard conditions until cells are 80–90 % confluent, implying that cells cover 80–90 % of the culture flask surface area. In order to determine the volume of cell suspension required for reseeding after thawing frozen samples, the following formula should be used:

$$ \begin{array}{l}\mathrm{Total}\ \mathrm{number}\ \mathrm{of}\ \mathrm{viable}\ \mathrm{cell}\mathrm{s}\ \mathrm{in}\ \mathrm{cell}\ \mathrm{s}\mathrm{uspension}\div \hfill \\ {}\kern1em \left(\mathrm{reseeding}\ \mathrm{density}\times \mathrm{reseeding}\ \mathrm{s}\mathrm{urface}\ \mathrm{area}\ \mathrm{of}\ \mathrm{tissue}\ \mathrm{culture}\ \mathrm{flask}\ \mathrm{or}\ \mathrm{well}\right)\hfill \end{array} $$

7.1.2 Methods Used to Obtain Absolute Cell Counts

7.1.2.1 Trypan Blue (0.4 %) Dye Exclusion Method

Trypan blue is a non-membrane permeable vital stain that is used to assess the viability of cells. Trypan blue is not able to cross the cell membrane of intact, viable cells and therefore only stains cells with compromised cell membranes. Dead cells display a distinct blue colour after staining, due to the accumulation of trypan blue in the cytoplasm of cells with compromised cell membranes The recommended procedure is as follows: (1) prepare the Neubauer counting chamber (haemocytometer) by carefully placing the cover slip on the counting grid; (2) carefully mix 80 μl of a 0.4 % trypan blue with 100 μl PBS and 20 μl of the suspension; (3) carefully load 10 μl of the solution onto both sides of the Neubauer counting chamber (haemocytometer); and (4) count the viable (unstained) cells as well as dead cells (stained) using a microscope (10 times objective lens). The following formula is used to determine the absolute cell concentration (Fig. A.1):

Absolute cell concentration (cells/ml) \( =\left(\frac{Q1+Q2+Q3+\dots +Q8}{8}\right)\times \), where Q1–Q8 refer to eight quadrants on the haemocytometer. The factor 10 is to correct for the dilution of the sample with the 0.4 % trypan blue solution.

There are several commercial automated counting devices on the market that make use of trypan blue (0.4 %) dye exclusion assay principles. Examples of such devices are the Vi-Cell XRâ„¢ automated cell counter (Beckman Coulter, Miami, USA), Countessâ„¢ automated cell counter (Invitrogen, Carlsbad, USA) and TC20â„¢ automated cell counter (Bio-Rad, Hercules, USA).

7.1.2.2 Absolute Cell Count Determination Using a Benchtop Flow Cytometer (Beckman Coulter Flow Cytometers)

An example of the strategy that is followed to obtain an absolute count on a benchtop flow cytometer is illustrated in Fig. A.2. The cell population of interest is identified by using a side scatter linear (SS lin) and a forward scatter linear (FS lin) histogram by placing a region around the cell population of interest only, excluding the counting beads and debris. The counting beads are enumerated in the CAL region. In order to obtain an accurate absolute count, it is important to count a sufficient number of cells as well as counting beads (>1000 events if possible) as well as to perform the count as soon as possible after the counting beads are mixed with the sample.

Fig. A.2
figure 7

(a) A side scatter linear and forward scatter linear dot histogram displaying all the events measured by the flow cytometer. The flow beads (pink) and a gate were used to encircle the cell population that was counted until the CAL factor was reached. The gate-labelled cell population displays the cell population count that was expressed as the number of cells per μl cell suspension. (b) A histogram displaying the flow beads with a region of interest placed over the peak of the flow cytometry counting beads labelled as CAL. In this example, the specific calibration factor (assayed bead concentration) was 986

7.1.3 Induction of Adipogenesis In Vitro

ASCs are seeded at a density of 5 × 103 cells/cm2 in a six-well plate and maintained under standard culturing conditions (37 °C and 5 % CO2) with complete stromal medium (α-MEM or DMEM, containing 10 % FBS and 1 % pen/strep). When 70–80 % confluency is achieved, the stromal medium is removed and replaced with adipogenic induction medium (Table A.1) in three of the wells of the 6-well plate. DMEM supplemented with serum and antibiotics is added to the remaining three wells. These wells serve as non-induced controls. The cultures are maintained for 21 days under standard culturing conditions of 37 °C, 5 % CO2. During this period the induction and control media are replaced every second day.

Table A.1 Composition of induction media for adipogenic, osteogenic, chondrogenic and myogenic differentiation in vitro

7.1.3.1 Qualitative Assessment of Adipogenesis In Vitro Using Oil Red O

After 21 days of induction, the 6-well plates containing both induced and non-induced cultures are fixed by adding a 4 % formaldehyde solution for 1 h. A 0.5 % Oil Red O stock solution is prepared in isopropanol. An Oil Red O working solution is prepared from the stock solution by mixing three parts of the stock solution with two parts of double-distilled water (ddH2O) (volume/volume). After removal (aspiration) of the fixative solution, the cultures are allowed to dry at room temperature before adding 1 ml of Oil Red O working solution to both the adipogenic induced and non-induced cultures, followed by a 20 min incubation at room temperature. The stain is removed, and the wells are thoroughly washed with ddH2O until no pink discoloration of the freshly added ddH2O is visible to the naked eye.

The cultures are then counterstained with Toluidine Blue O. A 0.01 % Toluidine Blue O counterstain solution is prepared (pH 11) by adding 0.005 g Toluidine Blue O and 0.005 g Na2CO3 to 50 ml ddH2O. The Toluidine Blue O counterstain is added to the Oil Red O-stained cultures for 5 min at room temperature, after which the excess stain is washed away with ddH2O. 1 ml ddH2O is added to each well before microscopy analysis.

7.1.4 Induction of Osteogenesis In Vitro

Immunophenotyped ASCs are seeded at a density of 5 × 103 cells/cm2 in a six-well plate and maintained under standard culture conditions (37 °C and 5 % CO2) with stromal medium (α-MEM, containing 10 % FBS and 1 % pen/strep) until about 60–70 % confluency. Osteogenic induction medium (Table A.1) is introduced to half the wells and DMEM supplemented with serum and antibiotics to the other half to serve as non-induced (control) cultures. The cultures are maintained for 21 days under standard culture conditions of 37 °C, 5 % CO2. The induction and non-induction media are replaced every second day.

7.1.4.1 Qualitative Assessment of Osteogenesis In Vitro

Twenty-one days after induction of osteogenic diffe rentiation, the cells are fixed by addition of a 4 % formaldehyde fixative solution for 1 h. A 2 % Alizarin Red S classical stain is used to detect the calcium in the mineral matrix from mature osteocytes. An alizarin stock solution is prepared by adding 2 g Alizarin Red S powder to 100 ml of ddH2O. The solution is mixed thoroughly using a magnetic stirrer until solutes are dissolved before filtering through filter paper.

The induced and non-induced cultures are pre-washed with 2 ml PBS at pH 4.2 for 5 min before introducing 2 ml of the 2 % Alizarin Red S stain and incubating the cultures for 10 min at room temperature. The cultures are washed thoroughly with ddH2O to remove the excess stain. 1 ml ddH2O is added to each well before microscopy analysis.

7.1.5 Induction of Chondrogenesis In Vitro

A suspension culture technique is usually used for the differentiation of ASCs into chondrocytes. ASCs are seeded in a T25 flask at a density of 5 × 103 cells/cm2 and maintained under standard culture conditions until about 60 % confluence. The cells are enzymatically removed from the flask (0.25 % trypsin-EDTA) followed by the neutralization of the enzymatic action with the addition of stromal medium (α-MEM, containing serum and antibiotics).

The cell suspension is transferred into a 15 ml tube, and the sample is centrifuged for 5 min at 400 g. The substrate is carefully aspirated until only the ASC pellet remains in the tube. The ASC pellet is suspended in chondrogenic induction medium (Table A.1), for the chondrogenic induced cultures or DMEM supplemented with serum only and for the non-induced cultures, and centrifuged at 400 g for 10 min. The tubes are carefully placed into the incubator without disrupting the pellet. The tube caps are slightly loosened to allow for gas exchange to occur. Cultures are incubated under standard conditions of 37 °C, 5 % CO2 for 21 days. The induction and control media (0.5 ml) are replaced every second day. After 24 h, the ASC pellets contract into a sphere. The cells that have not been incorporated into the sphere after 48 h are removed from the suspension cultures during medium replacement.

7.1.5.1 Qualitative Assessment of Chondrogenesis In Vitro

Each induced and non-induced chondrocyte sphere is serially dehydrated in 30, 50, 70 and 90 % ethanol, followed by three changes of absolute ethanol for 15 min per dehydration step. The dehydrated chondrocyte spheres are infiltrated with 50 % LR White Resin in absolute ethanol for 1 h, followed by an infiltration in a 100 % LR White Resin overnight. To embed the tissue spheres, they are transferred into resin capsules with 100 % LR White Resin and polymerized for 24 h at 60 °C.

Ten to 15 serial transverse sections of between 0.5 and 1.0 μm (optimal 0.5 μm) are prepared using a microtome, and the sections are stained with 1 % Toluidine Blue O stain for 30 s. The glass slides containing stained sections are gently rinsed with ddH2O. One drop of ddH2O is added on the glass plate and covered using a cover slip before microscopy analysis is performed.

Rights and permissions

Reprints and permissions

Copyright information

© 2016 Springer International Publishing Switzerland

About this chapter

Cite this chapter

van Vollenstee, F.A. et al. (2016). Isolation and Characterization of Adipose-Derived Stromal Cells. In: Pham, P. (eds) Stem Cell Processing . Stem Cells in Clinical Applications. Springer, Cham. https://doi.org/10.1007/978-3-319-40073-0_7

Download citation

Publish with us

Policies and ethics