culture media for the differentiation of mesenchymal stromal cells

15
Review Culture media for the differentiation of mesenchymal stromal cells Corina Vater, Philip Kasten, Maik Stiehler Department of Orthopedic Surgery, University Hospital Carl Gustav Carus, Dresden, Germany article info Article history: Received 7 April 2010 Received in revised form 20 July 2010 Accepted 27 July 2010 Available online 3 August 2010 Keywords: Mesenchymal stromal cell Cell culture medium Differentiation Proliferation Osteoblast abstract Mesenchymal stromal cells (MSCs) can be isolated from various tissues such as bone marrow aspirates, fat or umbilical cord blood. These cells have the ability to proliferate in vitro and differentiate into a series of mesoderm-type lineages, including osteoblasts, chondrocytes, adipocytes, myocytes and vascular cells. Due to this ability, MSCs provide an appealing source of progenitor cells which may be used in the field of tissue regeneration for both research and clinical purposes. The key factors for successful MSC prolifer- ation and differentiation in vitro are the culture conditions. Hence, we here summarize the culture media and their compositions currently available for the differentiation of MSCs towards osteogenic, chondro- genic, adipogenic, endothelial and vascular smooth muscle phenotypes. However, optimal combination of growth factors, cytokines and serum supplements and their concentration within the media is essential for the in vitro culture and differentiation of MSCs and thereby for their application in advanced tissue engineering. Ó 2010 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved. 1. Introduction Since Owen and Friedenstein’s work [1] it has been assumed that bone marrow contains special cells that can be expanded in vitro and differentiated into various mesoderm-type lineages, including bone, fat, cartilage, muscle, tendon, haematopoiesis-sup- porting stroma and vasculature [2–6]. We presently refer to these nonhematopoietic cells as mesenchymal stromal cell (MSC; Fig. 1) [7]. In addition, researchers have recently transdifferentiated MSCs into non-mesodermal cell types such as neuronal-like cells [8–11] and pancreatic cell progenitors [12–15]. To date, MSCs have been isolated not only from bone marrow but also from many other tissues and organs, including adipose tis- sue, umbilical cord blood, placental tissue, liver, spleen, testes, menstrual blood, amniotic fluid, pancreas and periosteum [16– 20]. When cultured in vitro on polystyrene surfaces, MSCs reveal morphological heterogeneity. These cells can be narrow spindle- shaped, large polygonal or even cuboidal-shaped when growing into a confluent monolayer [21]. In adult human, MSCs lack the hematopoietic surface antigens, e.g. CD11, CD14, CD34 and CD45 [22]. Meanwhile numerous molecular markers have been found on MSC surface, but none of them is specific to MSCs. Despite this, molecules such as CD44, CD73, CD90, STRO-1 and CD105/SH2 [3,23–25] are still currently used to identify MSCs. MSCs are considered to be nonimmunogenic since these cells have been transplanted into allogeneic hosts even without using any immunosuppressive drugs [22,26]. Furthermore, it has been reported that MSCs actually possess immunosuppressive proper- ties by modulating the function of T-cells [27,28], dendritic cells and B-cells [29–32]. For the characterization of MSC plasticity, their ability to differ- entiate in vitro into osteoblasts, chondrocytes and adipocytes is currently treated as the gold standard. This, in combination with the advantages that MSCs have no immunogenicity and can be eas- ily isolated from different tissues and expanded in vitro, enables MSCs to be a promising source of stem cells. Hence MSCs have 1742-7061/$ - see front matter Ó 2010 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved. doi:10.1016/j.actbio.2010.07.037 Abbreviations: 1,25-D3, 1,a25-dihydroxyvitamin D3; a2Col6, a chain 2 of type 6 collagen; ALP, alkaline phosphatase; asc, ascorbic acid; Asc-2-P, ascorbic acid 2-phosphate; AT, adipose tissue; BM, bone marrow; BM-MSCs, bone marrow- derived mesenchymal stromal cells; BMP, bone morphogenetic protein; BSP, bone sialoprotein; C/EBP, CCAAT-enhancer-binding proteins; cAMP, cyclic adenosine monophosphate; Col10, collagen type 10; Col11, collagen type 11; Col1a1, collagen type 1 alpha-1; Col2, collagen type 2; Col9, collagen type 9; CREB/p300, cAMP response element-binding protein/E1A binding protein p300; dex, dexamethasone; EC, endothelial cell; ECM, extracellular matrix; FABP4/aP2, fatty acid-binding pro- tein-4; FCS, fetal calf serum; FGF, fibroblast growth factor; Gata-6, GATA-binding protein 6; GPDH, glycerol-3-phosphate dehydrogenase; IBMX, 3-isobutyl-1-meth- ylxanthine; IGF, insulin-like growth factor; LDL, low-density lipoprotein; LPL, lipoprotein lipase; mRNA, messenger ribonucleic acid; MSC, mesenchymal stromal cell; OC, osteocalcin; ON, osteonectin; OPN, osteopontin; Osx, osterix; PDGF, platelet-derived growth factor; PPARc2, peroxisome proliferation-activated recep- tor c2; PRP, platelet-rich plasma; Runx-2, runt-related transcription factor-2; SCID, severe combined immunodeficiency; SM22-a, transgelin; SMMHC, smooth muscle myosin heavy chain; Sox9, SRY-related high-mobility group box 9; SRF, serum response factor; T3, triiodothyonine; TGF-b, transforming growth factor-beta; UCB, umbilical cord blood; UCWJ, umbilical cord Wharton’s jelly; VEGF, vascular endothelial growth factor; vitD3, vitamin D3; VSMC, vascular smooth muscle cell; a-SMA, a-smooth muscle actin; b-GP, beta-glycerophosphate. Corresponding author. Address: Department of Orthopedic Surgery, University Hospital Carl Gustav Carus, Fetscherstr. 74, D-01307 Dresden, Germany. Tel.: +49 (0)351 458 3137; fax: +49 (0)351 449 210 419. E-mail address: [email protected] (M. Stiehler). Acta Biomaterialia 7 (2011) 463–477 Contents lists available at ScienceDirect Acta Biomaterialia journal homepage: www.elsevier.com/locate/actabiomat

Upload: avinash-patil

Post on 18-Apr-2015

109 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Culture Media for the Differentiation of Mesenchymal Stromal Cells

Acta Biomaterialia 7 (2011) 463–477

Contents lists available at ScienceDirect

Acta Biomaterialia

journal homepage: www.elsevier .com/locate /actabiomat

Review

Culture media for the differentiation of mesenchymal stromal cells

Corina Vater, Philip Kasten, Maik Stiehler ⇑Department of Orthopedic Surgery, University Hospital Carl Gustav Carus, Dresden, Germany

a r t i c l e i n f o a b s t r a c t

Article history:Received 7 April 2010Received in revised form 20 July 2010Accepted 27 July 2010Available online 3 August 2010

Keywords:Mesenchymal stromal cellCell culture mediumDifferentiationProliferationOsteoblast

1742-7061/$ - see front matter � 2010 Acta Materialdoi:10.1016/j.actbio.2010.07.037

Abbreviations: 1,25-D3, 1,a25-dihydroxyvitamin Dcollagen; ALP, alkaline phosphatase; asc, ascorbic2-phosphate; AT, adipose tissue; BM, bone marrowderived mesenchymal stromal cells; BMP, bone morpsialoprotein; C/EBP, CCAAT-enhancer-binding protemonophosphate; Col10, collagen type 10; Col11, collatype 1 alpha-1; Col2, collagen type 2; Col9, collageresponse element-binding protein/E1A binding proteiEC, endothelial cell; ECM, extracellular matrix; FABP4tein-4; FCS, fetal calf serum; FGF, fibroblast growth fprotein 6; GPDH, glycerol-3-phosphate dehydrogenasylxanthine; IGF, insulin-like growth factor; LDL, lolipoprotein lipase; mRNA, messenger ribonucleic acidcell; OC, osteocalcin; ON, osteonectin; OPN, osteoplatelet-derived growth factor; PPARc2, peroxisome ptor c2; PRP, platelet-rich plasma; Runx-2, runt-relatedsevere combined immunodeficiency; SM22-a, transgemyosin heavy chain; Sox9, SRY-related high-mobiliresponse factor; T3, triiodothyonine; TGF-b, transformumbilical cord blood; UCWJ, umbilical cord Whaendothelial growth factor; vitD3, vitamin D3; VSMC,a-SMA, a-smooth muscle actin; b-GP, beta-glyceroph⇑ Corresponding author. Address: Department of O

Hospital Carl Gustav Carus, Fetscherstr. 74, D-01307(0)351 458 3137; fax: +49 (0)351 449 210 419.

E-mail address: maik.stiehler@uniklinikum-dresde

Mesenchymal stromal cells (MSCs) can be isolated from various tissues such as bone marrow aspirates,fat or umbilical cord blood. These cells have the ability to proliferate in vitro and differentiate into a seriesof mesoderm-type lineages, including osteoblasts, chondrocytes, adipocytes, myocytes and vascular cells.Due to this ability, MSCs provide an appealing source of progenitor cells which may be used in the field oftissue regeneration for both research and clinical purposes. The key factors for successful MSC prolifer-ation and differentiation in vitro are the culture conditions. Hence, we here summarize the culture mediaand their compositions currently available for the differentiation of MSCs towards osteogenic, chondro-genic, adipogenic, endothelial and vascular smooth muscle phenotypes. However, optimal combination ofgrowth factors, cytokines and serum supplements and their concentration within the media is essentialfor the in vitro culture and differentiation of MSCs and thereby for their application in advanced tissueengineering.

� 2010 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.

1. Introduction porting stroma and vasculature [2–6]. We presently refer to these

Since Owen and Friedenstein’s work [1] it has been assumedthat bone marrow contains special cells that can be expandedin vitro and differentiated into various mesoderm-type lineages,including bone, fat, cartilage, muscle, tendon, haematopoiesis-sup-

ia Inc. Published by Elsevier Ltd. A

3; a2Col6, a chain 2 of type 6acid; Asc-2-P, ascorbic acid; BM-MSCs, bone marrow-

hogenetic protein; BSP, boneins; cAMP, cyclic adenosinegen type 11; Col1a1, collagenn type 9; CREB/p300, cAMPn p300; dex, dexamethasone;/aP2, fatty acid-binding pro-actor; Gata-6, GATA-bindinge; IBMX, 3-isobutyl-1-meth-w-density lipoprotein; LPL,

; MSC, mesenchymal stromalpontin; Osx, osterix; PDGF,roliferation-activated recep-transcription factor-2; SCID,

lin; SMMHC, smooth musclety group box 9; SRF, seruming growth factor-beta; UCB,rton’s jelly; VEGF, vascularvascular smooth muscle cell;osphate.rthopedic Surgery, UniversityDresden, Germany. Tel.: +49

n.de (M. Stiehler).

nonhematopoietic cells as mesenchymal stromal cell (MSC; Fig. 1)[7]. In addition, researchers have recently transdifferentiated MSCsinto non-mesodermal cell types such as neuronal-like cells [8–11]and pancreatic cell progenitors [12–15].

To date, MSCs have been isolated not only from bone marrowbut also from many other tissues and organs, including adipose tis-sue, umbilical cord blood, placental tissue, liver, spleen, testes,menstrual blood, amniotic fluid, pancreas and periosteum [16–20]. When cultured in vitro on polystyrene surfaces, MSCs revealmorphological heterogeneity. These cells can be narrow spindle-shaped, large polygonal or even cuboidal-shaped when growinginto a confluent monolayer [21]. In adult human, MSCs lack thehematopoietic surface antigens, e.g. CD11, CD14, CD34 and CD45[22]. Meanwhile numerous molecular markers have been foundon MSC surface, but none of them is specific to MSCs. Despite this,molecules such as CD44, CD73, CD90, STRO-1 and CD105/SH2[3,23–25] are still currently used to identify MSCs.

MSCs are considered to be nonimmunogenic since these cellshave been transplanted into allogeneic hosts even without usingany immunosuppressive drugs [22,26]. Furthermore, it has beenreported that MSCs actually possess immunosuppressive proper-ties by modulating the function of T-cells [27,28], dendritic cellsand B-cells [29–32].

For the characterization of MSC plasticity, their ability to differ-entiate in vitro into osteoblasts, chondrocytes and adipocytes iscurrently treated as the gold standard. This, in combination withthe advantages that MSCs have no immunogenicity and can be eas-ily isolated from different tissues and expanded in vitro, enablesMSCs to be a promising source of stem cells. Hence MSCs have

ll rights reserved.

Page 2: Culture Media for the Differentiation of Mesenchymal Stromal Cells

Fig. 1. Morphology of human immortalized single cell-derived BM-MSCs at different stages of confluency (A: 10%; B: 90%). Cells were cultured in a-MEM containing 1%penicillin/streptomycin and 10% FCS (BM-MSCs, bone marrow-derived MSCs).

464 C. Vater et al. / Acta Biomaterialia 7 (2011) 463–477

been used in the therapy of diseases such as extended osseous de-fects [33], acute myocardial infarction [34], leukemia [35] and dia-betes [36]. In addition, the homing capability endows MSCs withfurther potential applications. For example, MSCs may be usedfor supporting tissue regeneration [37], correcting congenital dis-orders (e.g. osteogenesis imperfecta [38]) and controlling chronicinflammatory diseases [39,40], and have even employed as vehi-cles for the delivery of biological agents [22] and as probes in thebiocompatibility test of new implant materials. A prerequisite forthe therapeutic application of MSCs is to develop efficient andstandardized protocols so that MSCs can be induced to differenti-ate along the way as required. Therefore, we here present an over-view of the optimized protocols for MSC differentiation towardsthe osteogenic, chondrogenic, adipogenic, endothelial and vascularsmooth muscle phenotypes.

2. Osteogenic differentiation

2.1. Background

Bone diseases are major socioeconomic issues. The WorldHealth Organization has acknowledged this fact by declaring theyears 2000–2010 ‘‘The Bone and Joint Decade”. The developmentof innovative bone-healing strategies is a prerequisite for the suc-cessful treatment of a variety of patients suffering from local bonedefects caused by trauma, tumour, infection, degenerative jointdisease, congenital crippling disorders or periprosthetic bone loss.Furthermore, bone graft material is frequently needed for spinalfusion, joint revision surgery, corrective osteotomy proceduresand bone reconstruction in the field of oral and maxillofacial sur-gery. Bone grafting is one of the most common orthopaedic proce-dures with autologous bone graft providing osteoinductive growthfactors, bone-forming cells and structural support for new bone in-growth. However, the use of autologous bone graft is associatedwith the disadvantages of limited graft availability and donor sitemorbidity, e.g. pain, infection, pelvic fractures or neurovascular in-jury. The implantation of sterilized bone allograft material – usu-ally derived from femoral heads during joint replacementprocedures – as a widely used alternative bone-filling materialmay result in failure rates of up to 30% due to insufficient osseoin-tegration of the graft, requiring further surgical intervention [41].Insufficient bone-healing therefore remains a challenging issue.In this context, innovative cell-based strategies using MSCs arepromising for both site-specific and systemic bone regeneration.

2.2. Morphology and differentiation markers

When being differentiated into osteoblasts, MSCs transformfrom a fibroblastic to a cuboidal shape, produce extracellular ma-

trix (ECM), mainly composed of collagen type I, and in a later stageform aggregates or nodules that can be stained positively by aliza-rin red and von Kossa techniques. Increased expression of alkalinephosphatase (ALP; Fig. 2A) and calcium accumulation are observedin MSCs during osteogenic differentiation [3,22]. The enzymaticactivity of ALP as well as the calcium content can be quantifiedby colorimetric assays [42]. At the molecular level, osteogenic dif-ferentiation of MSCs is controlled by interactions between distincthormones and transcription factors.

Runt-related transcription factor-2 (Runx-2) effectuates theexpression of bone-specific genes, e.g. osterix (Osx), collagen type1 alpha-1 (Col1a1), osteocalcin (OC) and bone sialoprotein (BSP),by binding to the promoters of these genes [43–48]. Generally,Runx-2, ALP, Col1a1, transforming growth factor-beta 1 (TGF-b1),osteonectin (ON) and bone morphogenetic protein-2 (BMP-2) areknown to be early markers of osteoblastic differentiation, whereasOC and osteopontin (OPN) are expressed later in the differentiationprocess [49–52].

2.3. Differentiation protocols

The classical method for osteogenic differentiation of MSCsin vitro involves incubating a confluent monolayer of MSCs withcombinations of dexamethasone (dex), beta-glycerophosphate (b-GP) and ascorbic acid (asc) for several weeks. In addition, combina-tions of vitamin D3 (vitD3), transforming growth factor-beta (TGF-b) and bone morphogenetic proteins (BMPs) are used for osteo-genic differentiation. In the following these supplements will bedescribed in detail.

Dex is a synthetic glucocorticoid and has been reported to be anessential requirement for osteoprogenitor cell differentiation inMSCs [53,54]. While MSCs that were cultured in basal mediumwithout osteogenic supplements express increased levels of ALP,they fail to express mineralized ECM as well as other osteogenicmarkers such as Col1 [55]. Although the precise mechanisms of ac-tion of dex on stem cell differentiation and skeletal function are notknown, it is supposed that dex induces transcriptional effects. Inrat osteoblast-like cells, for instance, dex induces transcription ofBSP by binding on a glucocorticoid response element in the pro-moter region of the BSP gene [56]. On the other hand, dex improvesthe expression of the b-catenin-like molecule TAZ (transcriptionalcoactivator with PDZ-binding motif) as well as integrin a5, whichboth promote osteoblastic differentiation of MSCs by activatingRunx-2-dependent gene transcription [57,58]. However, glucocor-ticoids in supraphysiological amounts have deleterious effects onbone in vivo, resulting in inhibition of osteoblast function [59]. Ina study by Walsh et al. MSCs were cultured in the presence andabsence of dex at concentrations between 10 pM and 1 lM forup to 28 days [60]. The authors suggest that the critical effective

Page 3: Culture Media for the Differentiation of Mesenchymal Stromal Cells

Fig. 2. Differentiation potential of MSCs. For osteogenic differentiation human BM-MSCs were seeded on glass slides and cultured in a-MEM containing 1% penicillin/streptomycin, 10% FCS, 10 nM dex, 3.5 mM b-GP and 10 nM 1,25-D3. After 21 days of cultivation immunofluorescent staining for alkaline phosphatase (shown in green) andfluorescence-labeled phalloidin staining for actin (shown in red) was performed (A). To induce chondrogenic differentiation porcine BM-MSCs were cultured in pellet inDMEM containing 1% ITS, 1% penicillin/streptomycin, 10 ng ml�1 TGF-b3, 10 nM dex and 210 lM ascorbic acid. After 21 days, differentiation into the chondrogenic lineagewas visualized by immunohistochemical staining for ColX (shown in brown) (B). Human immortalized single-cell-derived BM-MSCs were seeded on tissue culturepolystyrene and induced to differentiate into adipocytes by culturing the cells in DMEM containing 10% FCS, 1 mM dex, 500 lM IBMX, 1 lg ml�1 insulin, and 100 lMindomethacin for 21 days. The appearance of intracellular lipid-rich vacuoles was confirmed by oil red O staining (shown in red) (C). Tubular-like structures formed by humanimmortalized single-cell-derived BM-MSCs in fibrin-based gels after 7 days of endothelial differentiation in a-MEM containing 1% penicillin/streptomycin, 10% FCS and50 ng ml�1 VEGF was visualized by bright-field microscopy (D). Immunofluorescent staining for calponin (shown in green) and fluorescent staining for actin (shown in red)and nuclei (shown in blue) in porcine BM-MSCs differentiated towards the vascular smooth muscle phenotype (E). Therefore cells were cultured in a-MEM containing 1%penicillin/streptomycin, 10% FCS and 10 ng ml�1 TGF-b1.

C. Vater et al. / Acta Biomaterialia 7 (2011) 463–477 465

concentration of dex is 10 nM, corresponding to its physiologicalconcentration. At higher concentrations (of the order of 100 nM)dex inhibits osteoblast differentiation in MSCs and leads to gluco-corticoid-induced osteoporosis [57]. In addition, proliferation isnegatively affected, mainly due to the inhibitory effect of glucocor-ticoids on collagen synthesis [61]. However, when MSCs are cul-tured in the presence of asc, the effects of glucocorticoids oncollagen production are markedly masked [62]. Song et al. even re-ported on a reductive effect of dex on density-related apoptosis inMSC cultures [63]. When implanted subcutaneously into SCIDmice, pretreatment of MSCs with 100 nM dex for up to 5 weeks re-sulted in enhanced bone tissue formation compared with un-treated controls [42]. For complete induction of osteogenicdifferentiation of in vitro cultivated MSCs at least 3 weeks of con-tinuous treatment with dex is required [42].

For matrix mineralization the presence of both calcium andphosphate ions is essential. b-GP, which is enzymatically hydro-

lyzed by alkaline phosphatase, serves as a crucial source of inor-ganic phosphate [64]. Chung et al. showed that cultivation ofosteoblast-like cells in culture medium containing b-GP leads tomineral formation, lactate generation, increased ALP activity, aswell as protein and phospholipid synthesis, indicating enhancedosteogenic differentiation [65]. Usually 5–10 mM b-GP is used forosteogenic differentiation of MSCs [55,65,66].

Asc plays an important role as a cofactor for the hydroxylationof proline and lysine residues in collagens, which are the mostabundant group of ECM proteins in the body [67,68]. One difficultyconcerning the handling of asc is its instability in solution, espe-cially under standard culture conditions (pH 7.5, 37 �C, humidifiedatmosphere, 5% CO2) [69,70]. Thus it is recommended to use thelong-acting vitamin C derivative ascorbic acid 2-phosphate (Asc-2-P) which was found to be stable under conventional culture con-ditions [71]. Recent studies showed that in the presence of Asc-2-PMSCs demonstrate upregulation of genes related to cell cycle and

Page 4: Culture Media for the Differentiation of Mesenchymal Stromal Cells

466 C. Vater et al. / Acta Biomaterialia 7 (2011) 463–477

mitosis, whereas absence of Asc-2-P leads to reduced ALP expres-sion and inhibition of calcium accumulation [72]. The mitogenic ef-fect of Asc-2-P can be ascribed either to the role of the Asc-2-P-induced ECM as a growth factor reservoir or to a direct activationof mitogenic pathways. In the absence of collagens, integrin signal-ling is impaired and downstream signals that are needed for mito-gen-activated protein kinase activation are absent. No inhibitoryeffect on cell growth could be found when supplementing the cul-ture media with up to 10 mM ascorbic acid. Usually, concentra-tions ranging from 50 up to 500 lM are used to induceosteogenic phenotype of MSCs [42,69,73].

vitD3 is a secosteroid hormone which is essential for the func-tion of osteoblasts. Its active form, 1a,25-dihydroxyvitamin D3(1,25-D3), demonstrates potent antiproliferative effects by block-ing the transition from the G1- to the S-phase during the cell cycle.Thereby osteogenic differentiation of MSCs is supported [74,75].Non-genomic effects of 1,25-D3, which occur through the interac-tion of the hormone with a membrane-bound receptor, include anincrease in cyclic adenosine monophosphate (cAMP) and the open-ing of calcium channels [76]. Through interaction with a nuclearvitamin D receptor, 1,25-D3 has been shown to stimulate theexpression of bone-related transcription factors, i.e. Runx-2 andOsx, in addition to osteoblast differentiation markers, e.g. ALP,Col1a1, OC and OPN [70]. Although 1,25-D3 synergized with bothdex and bone morphogenetic protein-2 promoted expression ofosteoblastic markers, this agent alone was unable to induce matrixmineralization [59,77]. In contrast to this, Jaiswal et al. reportedthat dex may reduce vitamin D receptor expression in osteoblasticcells [78], leading to a reduced uptake ability for 1,25-D3 andtherefore decreased expression of differentiation markers such asOC.

As well as using dex, b-GP, Asc-2-P and 1,25-D3, the addition ofdistinct growth factors can enhance osteogenic differentiation ofMSCs in vitro. TGF-b exists in three isoforms, whereas TGF-b1 isthe major form found in bone [79,80]. TGF-b1 influences cellgrowth and plays an essential role in the control of bone formationby modulating the synthesis and degradation of several bone ma-trix components, e.g. collagen type 1 and non-collagenous proteins[81,82]. Notably, although TGF-b1 stimulates the expression ofRunx-2, it inhibits osteoblast differentiation in the late stages[77,83]. BMPs are also members of the TGF-superfamily and can,in contrast to TGF-b1, induce ectopic bone formation in developedtissues [84,85]. Recent reports investigating the role of BMPs inosteogenesis [86–88] showed that there may be species-specificdifferences in the effect of BMPs in vitro. In both mice and rats,BMPs promote osteoblast differentiation [89–91]. Interestingly,there is body of evidence that BMP-2, BMP-4 and BMP-7 fail to in-duce osteogenic differentiation of human MSCs [87,92]. In contrast,

Table 1Representative protocols used for in vitro osteogenic differentiation of MSCs (asc, ascorbidexamethasone; FCS, fetal calf serum; ITS, insulin-transferrin-selenious acid; TGF-b, transfomarrow; UCB, umbilical cord blood; UCWJ, umbilical cord Wharton’s jelly).

Reference FCS dex asc

Oswald et al. [5] 10% 100 nM 200 lMJørgensen et al. [59] 10% 100 nM –

Friedman et al. [262] – 100 nM 25 lg ml–1

Stiehler et al. [263] 10% 100 nM 290 nMHildebrandt et al. [55] – 10 nM 100 nM 300 lMPytlík et al. [73] 10% 100 nM 500 lMSong et al. [42] 10% 100 nM 50 lMChen et al. [66] 10% 100 nM 200 lM

several studies demonstrated that in cells of the osteoblast lineage,BMP-2, BMP-4 and BMP-7 are capable of inducing expression ofALP, Col1a1, OPN, BSP and other non-collagenous bone proteinsfound in osteoid [55,91,93–96].

Representative protocols used for in vitro osteogenic differenti-ation of MSCs including dex, b-GP, Asc-2-P, vitD3, TGF-b and BMPsas well as commercially available culture media are summarized inTables 1 and 2.

3. Chondrogenic differentiation

3.1. Background

Cartilage defects have only a limited intrinsic healing capacity.For instance, partial thickness defects that do not penetrate thesubchondral bone usually do not repair spontaneously [97]. Smallfull-thickness defects can repair spontaneously, resulting inhyaline-like cartilage. However, larger defects only regenerate byproduction of fibrous tissue or fibrocartilage, which as biomechan-ically inferior compared with physiological hyaline cartilage. Thecell-based regeneration of (osteo)chondral defects presents a ma-jor challenge. Although, in principle, autologous chondrocytescan be used for cartilage tissue-engineering applications, their lim-ited availability, quantity and viability are major drawbacks [98].In this context, MSCs are promising candidates for cartilage regen-eration [99].

3.2. Morphology and differentiation markers

During chondrogenic differentiation MSCs change from a char-acteristic fibroblast-like morphology to a large round shape. Thecells are surrounded by abundant ECM, consisting of a highly orga-nized network of collagen (predominantly type 2 collagen) andaggregating proteoglycans and glycosaminoglycans of high molec-ular weight [100–102] that can be detected by, for example, Alcianblue, Toluidine blue and Safranin O stainings and quantified byspecific assays [72,103].

The protein SRY-related high-mobility group box 9 (Sox9) is anearly transcription factor of chondrogenesis and controls theexpression of the genes collagen type 2 (Col2), collagen type 9(Col9), collagen type 10 (Col10; Fig. 2B), collagen type 11 (Col11),aggrecan and cartilage link protein [104–108]. By binding to thepromoter of these genes, Sox9 forms transactivating complexeswith other proteins, e.g. Sox5/Sox6, cAMP response element-bind-ing protein/E1A binding protein p300 (CREB/p300) and c-musculo-aponeurotic fibrosarcoma [104,109,110]. Col10 is known to besynthesized during chondrocyte hypertrophy in the growth plate

c acid; bFGF, basic fibroblast growth factor; BMP, bone morphogenetic protein; dex,rming growth factor-beta; vitD3, vitamin D3; b-GP, beta-glycerophosphate; BM, bone

b-GP Other Cell source

10 mM – Human BM– vitD3 (1 nM)

BMP-2 (100 ng ml�1)Human BM

5 mM ITS (1%)bFGF (25 ng ml�1)

FGF-8/FGF-10 (50 ng ml�1)TGF-b (200 pM)BMP-2/4/6/7/14 (20 nM)

Human BM

5 mM vitD3 (10 nM) Human BM5 mM BMP-2 (100 nM) Human UCB10 mM – Human BM10 mM – Human BM10 mM – Human BM/UCWJ

Page 5: Culture Media for the Differentiation of Mesenchymal Stromal Cells

Table 2Media commercially available for osteogenic differentiation of MSCs.

Company Product

Invitrogen (Carlsbad, USA) STEMPRO� Osteogenesis Differentiation KitTrevigen (Gaithersburg, USA) Mesenchymal Stem Cell Osteogenic Differentiation KitThermo Scientific (Waltham, USA) HyClone™ AdvanceSTEM™ Osteogenic Differentiation KitR&D Systems (Minneapolis, USA) Human/Mouse StemXVivo Osteogenic/Adipogenic

Base Media Human/Mouse StemXVivo Osteogenic SupplementMiltenyi Biotec (Bergisch Gladbach, Germany) NH OsteoDiff MediumCellular Engineering Technologies Inc. (Coralville, USA) Osteogenic Differentiation MediaProvitro GmbH (Berlin, Germany) hMSC osteogenesis induction mediumPromoCell GmbH (Heidelberg, Germany) Mesenchymal Stem Cell Osteogenic Differentiation Medium

C. Vater et al. / Acta Biomaterialia 7 (2011) 463–477 467

of long bones and is used to identify terminally differentiatedhypertrophic chondrocytes [111–114].

3.3. Differentiation protocols

In 1998 Johnstone et al. first described a defined cell culturemedium for in vitro chondrogenesis of MSCs [115]. The two mainprinciples involved in enhancing chondrogenic differentiationin vitro are close cell-to-cell contact, usually achieved by cell pelletor micromass culture [115,116], and the addition of chondrogenicbioactive factors, e.g. dex, asc, TGF-b, BMPs, fibroblast growth fac-tor (FGF) and insulin-like growth factor (IGF). Among these dex, ascand TGF-b have been shown to be most effective [102,117–120].The effects of these factors on MSCs will be discussed in more de-tail below.

Glucocorticoids are used for the in vitro differentiation of MSCsinto multiple lineages, including stroma, fat, bone and cartilage[121]. Glucocorticoid function is mediated by the cytoplasmic glu-cocorticoid receptor, which influences various differentiation pro-cesses by inducing transcriptional actions. In a study by Derfoulet al., dex upregulated gene expression and protein levels of severalcartilage matrix markers, in particular Col11 [121]. However, incontrast to combination with TGF-b, dex alone has little effectson the expression of chondrogenic markers such as aggrecan, car-tilage oligomeric matrix protein and Col2. For human primary bonemarrow-derived MSCs, successful chondrogenic differentiationwas shown in medium containing 100 nM dex [122].

Asc and its derivative Asc-2-P causes collagen hydroxylation viamodification of proline and lysine residues. Its addition to the cul-ture medium leads to increased MSC proliferation and enhancesproduction of collagen type 2, which is one of the most importantECM proteins in cartilage [120].

TGF-b interacts with a heteromeric receptor complex compris-ing two structurally related serine–threonine kinases, types I andII receptors, and transforms its signals intracellularly via Smad pro-teins [123,124]. Together with dex, TGF-b represents an indispens-able factor for chondrogenic differentiation of human MSCs [118].Although Barry et al. state that TGF-b2 and TGF-b3 are more effec-tive than TGF-b1 in promoting glycosaminoglycan and type 2 col-lagen accumulation [122], others have concluded that any of theTGF-b subtypes are equally active chondrogenic factors and thatlot- rather than subtype-specific differences seem to exist [125].Whilst chondrogenic differentiation of MSCs in monolayer cultureappears to be dependent on TGF-b concentration and on cultiva-tion time, a concentration of 10 ng ml�1 TGF-b is sufficient for suc-cessful MSC differentiation in pellet culture [122,126]. AlthoughTGF-b supports early- and intermediate-stage chondrogenesis, itis known to retain chondrocytes in the prehypertrophic state.Therefore TGF-b most likely represses terminal in vitro differentia-tion of MSCs [127].

BMPs play an important role during bone morphogenesis by ini-tiating chondro-progenitor cell determination and differentiation[128]. Although BMP-2, BMP-4, BMP-6 and BMP-7 act synergisti-

cally with TGF-b by enhancing ECM deposition, they are not suffi-cient to stimulate in vitro chondrogenesis of human MSCs inconventional pellet culture with dex-supplemented medium[129,130]. In a study by Richter et al. MSCs from bone marrow re-quired only dex and TGF-b for successful chondrogenic differentia-tion, whereas the differentiation of MSCs derived from adiposetissue or synovial tissue was dependent on the additional supple-mentation with BMP-6 [122]. Sekiya et al. demonstrated that thechondrogenic effect of BMP-2 on bone-marrow-derived MSCs isgreater than that of BMP-4 and BMP-6 as measured by increasedpellet weight, superior production of proteoglycans and Col2 [131].

In vitro treatment of MSCs with FGF, dex, or combinations ofboth, increased pellet content of collagen type 2 and glycosamino-glycans as well as mRNA expression of aggrecan [132,133]. Jingushiet al. found that repeated injections of FGF into the fracture site inrats induced cartilaginous callus formation enlargement [117].

The chondrogenic differentiation potential of MSCs treated withIGF has been investigated extensively. IGF stimulates MSC prolifer-ation, regulates cell apoptosis, induces the expression of chondro-cyte markers, increases the synthesis of matrix proteins includingcollagen type 2 and proteoglycans, and promotes the survival,development and maturation of chondrocytes in vitro [102,134–136]. IGF-preconditioned autologous bone-marrow-derived MSCsenhanced chondrogenesis in full-thickness cartilage lesions in thefemoropatellar articulations of young mature horses [137].Although some studies did not show any effect of isolated IGFapplication on in vivo MSC differentiation, Indrawattana et al.showed that IGF is capable of inducing chondrogenic differentia-tion on a level comparable to TGF-b [138–140]. However, Matsudaet al. reported that the combination of TGF-b, dex and IGF was themost effective cocktail to stimulate differentiation of MSCs tochondrocytes [141].

Usually chondrogenic differentiation of MSCs is performed inserum-free media in order to reduce serum-induced cellular apop-tosis [142]. In this context, platelet-rich plasma (PRP) seems to be apromising supplementary agent. PRP is defined as plasma enrichedfor platelets and contains effective growth factors such as TGF-b1and vascular endothelial growth factor (VEGF) [143]. Recentin vitro investigations have confirmed that both cellular prolifera-tion as well as mRNA levels of Runx-2, Sox9 and aggrecan were sig-nificantly higher in PRP-treated cells compared to fetal calf serum(FCS)-treated MSC cultures [144]. Furthermore, the level of chon-drogenic potential of MSCs is dependent on the tissue source thecells are isolated from. Studies showed that bone-marrow-derivedMSCs demonstrated higher levels of in vitro chondrogenic poten-tial compared to adipose-derived MSCs [145–148]. Sakaguchiet al. even demonstrated that MSCs isolated from synoviumshowed superior chondrogenic potential than MSCs isolated fromother mesenchymal tissues [149].

Tables 3 and 4 give an overview about representative protocolsused for in vitro chondrogenic differentiation of MSCs includingdex, asc, TGF-b, BMPs and IGF as well as the respective commer-cially available media.

Page 6: Culture Media for the Differentiation of Mesenchymal Stromal Cells

Table 3Representative protocols used for in vitro chondrogenic differentiation of MSCs (asc, ascorbic acid, BSA, bovine serum albumin; BMP, bone morphogenetic protein; dex,dexamethasone; FCS, fetal calf serum; IGF, insulin-like growth factor; ITS, insulin-transferrin-selenious acid; TGF-b, transforming growth factor-beta; BM, bone marrow; UCWJ,umbilical cord Wharton’s jelly; AT, adipose tissue). In all protocols cells were cultured under three-dimensional conditions (pellet, scaffold).

Reference FCS dex asc TGF-b Other Cell source

Matsuda et al. [141] 10% 39 ng ml�1 50 lg ml�1 100 ng ml�1 IGF (100 ng ml�1) Human BMSekiya et al. [131] – 100 nM 50 lg ml�1 100 ng ml�1 BSA (1.25 mg ml�1)

ITS (50 mg ml�1)linoleic acid (5.35 mg ml�1)BMP-2/4/6 (500 ng ml�1)

Human BM

Vogel et al. [264] – 100 nM 170 lM 10 ng ml�1 BSA (1.25 mg ml�1)insulin (5 lg ml�1)transferring (5 lg ml�1)selenous acid (5 lg ml�1)

Human BM

Shen et al. [265] – 100 nM 50 lg ml�1 10 ng ml�1 BSA (1.25 mg ml�1)ITS (1%) linoleic acid (5.35 lg ml�1)BMP-7 (100 ng ml�1)

Human BM

Chen et al. [66] – 100 nM 50 lg ml�1 10 ng ml�1 BSA (1.25 mg ml�1)ITS (1%)

Human BM/UCWJ

Fernandes et al. [72] – 100 nM 200 lM 10 ng ml�1 ITS (50 lg ml�1)BMP-6 (500 ng ml�1)

Human BM

Diekman et al. [266] – 100 nM 37.5 lg ml�1 10 ng ml�1 ITS (1%)BMP-6 (10/500 ng ml�1)

Human BM/AT

Table 4Media commercially available for chondrogenic differentiation of MSCs.

Company Product

Invitrogen (Carlsbad, USA) StemPro� Chondrogenesis Differentiation KitR&D Systems (Minneapolis, USA) Human/Mouse StemXVivo Chondrogenic Base Media

Human/Mouse StemXVivo Chondrogenic SupplementMiltenyi Biotec (Bergisch Gladbach, Germany) NH ChondroDiff MediumCellular Engineering Technologies Inc. (Coralville, USA) Chondrogenic Differentiation MediaProvitro GmbH (Berlin, Germany) hMSC chondrogenesis induction mediumPromoCell GmbH (Heidelberg, Germany) Mesenchymal Stem Cell Chondrogenic Differentiation Medium

468 C. Vater et al. / Acta Biomaterialia 7 (2011) 463–477

4. Adipogenic differentiation

4.1. Background

The formation of adipocytes and their aggregation in order toform adipose tissue denotes an important step in the evolutionof vertebrates, as it guarantees independence from food intakeover longer periods of time [150]. In plastic and reconstructive sur-gical procedures, adipose tissue is frequently needed to repair softtissue defects that result from traumatic injury (i.e. significantburns), tumour resections (i.e. mastectomy and carcinoma re-moval), and congenital defects [151]. The American Society of Plas-tic Surgeons reported that over 5 million reconstructive procedureswere performed in 2004, approximately 4 million of which weredue to tumour removal [152]. Strategies to repair soft tissue de-fects, e.g. breast reconstruction procedures, include the use of im-plants and fillers [153,154]. However, there exists no void-fillingmaterial that will satisfy all clinical needs. The use of autologousfat tissue has not been consistently successful in patients[155,156] due to the fact that fat transplants lack sufficient vascu-larization, i.e. central graft blood flow is not adequate for long-termsurvival of the tissue, and often leads to tissue resorption [157]. Inthis context, tissue engineering using MSCs is an appealing strat-egy to provide adipose tissue grafts.

4.2. Morphology and differentiation markers

Adipose cell differentiation is a multistep process characterizedby a sequence of events during which preadipocytes divide untilconfluence [158]. When being differentiated into adipocytes, fibro-blastic MSCs are converted to a spherical shape expressing severaltypes of ECM proteins, including fibronectin, laminin, and types 1,

3, 4, 5 and 6 collagen. Initially a fibronectin network develops,followed by the formation of a type I collagen network [159–161]. Finally, differentiation of MSCs into adipocytes leads to accu-mulation of intracellular lipid-rich vacuoles that can be stainedpositively by oil red O (Fig. 2C). Quantitative analyses can be per-formed either by staining the cells with oil red O and extractingthe dye from the cells with isopropanol [103] or by determiningthe activity of glycerol-3-phosphate dehydrogenase (GPDH)[162–164].

The adipocyte-specific peroxisome proliferation-activatedreceptor c2 (PPARc2) belongs to the PPARs family, and is, togetherwith CCAAT-enhancer-binding proteins (C/EBP), one of the tran-scription factors that regulate expression of genes responsible forinduction and progression of adipogenesis [165,166]. Lipoproteinlipase (LPL) and the a chain 2 of type 6 collagen (a2Col6) areknown to be early markers of adipogenic differentiation, whereasleptin, fatty acid-binding protein-4 (FABP4/aP2) and adiponectinare expressed during later stages of differentiation [158,167–170].

4.3. Differentiation protocols

A routinely used method to stimulate adipogenic differentiationof preadipocytes and MSCs is the cultivation of these cells untilconfluency, followed by incubation with dex, 3-isobutyl-1-methyl-xanthine (IBMX), insulin and indomethacin, or combinations ofthese. In addition to these agents, triiodothyonine (T3), Asc-2-Pand basic FGF (bFGF-2) are also used for adipogenic differentiationas will be described in detail below.

The glucocorticoid dex induces the accumulation of transcrip-tion factors including C/EBP and PPARc which are crucial for adi-pose conversion of precursor cells [171]. In a study conducted byCui and coworkers, adipogenesis (as quantified by the presence

Page 7: Culture Media for the Differentiation of Mesenchymal Stromal Cells

C. Vater et al. / Acta Biomaterialia 7 (2011) 463–477 469

of triglyceride-containing vesicles and by expression of FABP4) wasinduced in a murine MSC cell line by treating the cells with dex[172].

The positive effect of glucocorticoids on adipogenic differentia-tion can be amplified by cAMP-elevating agents such as IBMX[173]. While inhibiting the activity of phosphodiesterase and tu-mour necrosis factor-a (TNF-a), IBMX enhances the expression ofPPARc2 and LPL and down-regulates the expression of osteogenicmarker genes such as Runx-2 and OPN by activation of protein ki-nase A [170,174–176].

Among the commonly used adipogenesis-inducing drugs, insu-lin is known to be the most potent [177]. It stimulates adipogenesisthrough the Akt-TSC2-mTORC1 pathway by increasing and acceler-ating triglyceride accumulation [178,179]. To maintain in vitro adi-pogenic differentiation of MSCs, the effect of insulin can beenhanced by insulin sensitizers, e.g. rosiglitazone or troglitazone,which act by stimulating PPARc [180–182].

The nonsteroidal anti-inflammatory drug indomethacin is acyclooxygenase inhibitor and blocks osteoclast and osteoblast dif-ferentiation, while promoting hMSC commitment to the adipogeniclineage [183,184]. Furthermore, indomethacin was found to bind di-rectly to the PPARc receptors and to act as a PPARc agonist, leadingin confluent MSC monolayers to the expression of adipogenic mark-ers such as adipose differentiation-related protein [185,186].

Several other growth factors, including thyroid hormones, e.g.T3, Asc-2-P and FGF-2, have been shown to positively influenceadipogenesis [187–189]. While T3 multiplies the effects of insulin,media supplementation with 500 lM Asc-2-P results in the accu-mulation of the highest number of oil droplets compared to controlmedium without Asc-2-P [103]. FGF-2, which belongs to the familyof heparin-binding growth factors, increases cellular proliferation

Table 5Representative protocols used for in vitro adipogenic differentiation of MSCs (asc, ascorbicT3, triiodothyonine; AT, adipose tissue; BM, bone marrow; UCWJ, umbilical cord Wharton

Reference FCS dex Insulin IBMX

van Harmelen et al. [267] – – 66 nM 500 lM

Hemmrich et al. [163] – 100 nM 66 nM 500 lM

Bunnell et al. [268] 20% 500 nM – 500 lMChoi et al. [103] 10% 1 mM 10 mg ml 500 lMTsuji et al. [164] – 100 nM 50 lM –

Vashi et al. [269] – 1 mM 10 mM 500 lMChen et al. [66] 10% 1 mM 5 mg ml 500 lMPytlik et al. [73] 10% 100 nM 10 lg ml 500 lMVallee et al. [161] 3% 1 lM 100 nM 250 lM

Table 6Media commercially available for adipogenic differentiation of MSCs.

Company

Invitrogen (Carlsbad, USA)Trevigen (Gaithersburg, USA)Thermo Scientific (Waltham, USA)R&D Systems (Minneapolis, USA)

Miltenyi Biotec (Bergisch Gladbach, Germany)Cellular Engineering Technologies Inc. (Coralville, USA)Provitro GmbH (Berlin, Germany)PromoCell GmbH (Heidelberg, Germany)

and exerts a direct effect on adipogenic differentiation by inducingthe expression of PPARc [190–193]. Matrigel functionalized withFGF-2 has been found to be proadipogenic when injected subcuta-neously alone [188] or in combination with gelatin microspheresinto mice [194]. Within the implantation period, a visible fat padwas formed at the injection site, which is probably due to prea-dipocyte and endothelial cell migration to the injection site[188,195].

The process of adipogenic differentiation of MSCs starts uponcellular monolayer confluence and is induced by the factors men-tioned above. To reach confluence MSCs are often cultured in med-ium containing FCS. However, it has been shown that extendedmaintenance of MSCs in FCS-supplemented medium markedlyand irreversibly reduces the capability for an adipogenic conver-sion to mature adipocytes, most likely due to the effect of serumfactors present in the FCS, e.g. TNF-a or TGF-b [196]. ThereforeHemmrich et al. recommended the use of human serum which,in contrast to FCS, allows substantial proliferation and differentia-tion even after long cell expansion periods [163].

Representative protocols used for in vitro adipogenic differenti-ation of MSCs, including dex, IBMX, insulin, indomethacin, T3 andasc, as well as commercially available culture media, are summa-rized in Tables 5 and 6.

5. Endothelial differentiation

5.1. Background

A major aim in tissue engineering is to ensure adequate vascu-larization of artificial tissue-like constructs. Previous studies haveshowed that vascularization within in vitro engineered tissues

acid; FCS, fetal calf serum; dex, dexamethasone; IBMX, 3-isobutyl-1-methylxanthine;’s jelly).

Indo-methacin Other Cell source

– Transferrin (10 lg ml�1)T3 (200 pM)

cortisol (100 nM)

Human AT

– Transferrin (10 lg ml�1)pioglitazone (100 lg ml�1)T3 (1 nM)

Human AT

50 lM – Rat AT10 mM asc (50 lM) Human BM– T3 (200 pM)

transferring (100 nM)calcium pantotenate (17 mM)biotin (33 mM)

Human AT

200 mM – Rat BM60 mM – Human BM/UCWJ200 lM – Human BM– asc (250 lM)

T3 (200 pM)rosiglitazone (1 lM)

Human AT

Product

STEMPRO� Adipogenesis Differentiation KitMesenchymal Stem Cell Adipogenic Differentiation Kit,HyClone AdvanceSTEM Adipogenic Differentiation KitHuman/Mouse StemXVivo Osteogenic/Adipogenic Base MediaHuman/Mouse StemXVivo Adipogenic SupplementNH AdipoDiff MediumAdipogenic Differentiation MediahMSC adipogenesis induction mediumMesenchymal Stem Cell Adipogenic Differentiation Medium

Page 8: Culture Media for the Differentiation of Mesenchymal Stromal Cells

Table 7Representative protocols used for in vitro endothelial differentiation of MSCs (EGF,epidermal growth factor; FCS, fetal calf serum; VEGF, vascular endothelial growthfactor; BM, bone marrow; UCWJ, umbilical cord Wharton’s jelly).

Reference FCS VEGF Other Cell Source

Jiang et al. [270] – 10 ng ml�1 – Human BMOswald et al. [5] 2% 50 ng ml�1 – Human BMChen et al. [66] 5% 100 ng ml�1 EGF (50 ng ml�1)

hydrocortisone(1 lg ml�1)

Human BM/UCWJ

Table 8Media commercially available for endothelial differentiation of MSCs.

Company Product

PromoCell GmbH(Heidelberg, Germany)

Endothelial Progenitor Medium

Cellular Engineering Technologies Inc.(Coralville, USA)

Endothelial Progenitor CellDifferentiation Media

470 C. Vater et al. / Acta Biomaterialia 7 (2011) 463–477

using mature endothelial cells (ECs) improved blood perfusion andcell viability during and after transplantation [197,198]. Blood ves-sels primarily consist of three cell types. While ECs can be found inthe innermost layer of a vessel, vascular smooth muscle cells(VSMCs) are typically located in the intermediate layer and fibro-blast-like cells in the adventitial outermost layer. However, matureECs and SMCs have limited proliferation potential and their use forgenerating vascular conduits would require laborious autologouscell isolation. In contrast, the fact that MSCs can be easily isolatedand differentiated into ECs makes them a favourable alternativecell source. In addition, the use of allogeneic or even xenogenicMSCs may be feasible without the aid of extended medical immu-nosuppression, thereby further alleviating the availability of MSCsas cellular component for tissue-engineering applications[199,200].

5.2. Morphology and differentiation markers

When being differentiated in semi-solid medium (see below)into endothelial cells, MSCs form three-dimensional capillary-liketubular structures (Fig. 2D) and have the ability to take up acety-lated low-density lipoproteins (LDLs) via special receptors. Theincorporation of dye-conjugated LDLs, e.g. 1,10-dioctadecyl-1-3,3,30,30-tetramethyl-indo-carbocyanine perchlorate conjugatedto acetylated-LDL (e.g. DiI-Ac-LDL, Invitrogen, Carlsbad, USA), canbe determined by fluorescence microscopy and fluorescence-acti-vated cell sorting [66,200]. Vascular endothelial growth factor(VEGF)-receptor 2 is one of the two major VEGF receptors and isexpressed during the early stage of endothelial differentiation ofMSCs [201,202]. Markers that characterize later stages of endothe-lial differentiation include tissue-type plasminogen activator, vonWillebrand factor, platelet-endothelial cell adhesion molecule 1(CD31) and vascular endothelial-cadherin [200,202].

5.3. Differentiation protocols

A commonly used method to stimulate the differentiation ofMSCs into endothelial cells is the cultivation of these cells in so-called semi-solid medium (e.g. Matrigel™, BD Biosiences, FranklinLakes, USA). The presence of VEGF – the most common supplemen-tary agent to date – markedly enhances endothelial differentiationof MSCs [5]. The VEGF family comprises seven members (i.e. VEGF-A/B/C/D/E/F and placental growth factor) that all have a commonVEGF homology domain. Alternative splicing of the VEGF-A genegenerates six isoforms of VEGF-A composed of 121, 145, 165,183, 189 and 206 amino acids, respectively. Of these, VEGF165 isthe most commonly expressed isoform [203,204]. VEGF-A is typi-cally synthesized by vascular and tumour cells, and has beenshown to be expressed by primary human MSCs as well [200]. Totest whether the MSC-secreted VEGF is able to contribute to angi-ogenesis, Beckermann et al. added supernatant from cultured MSCsto human umbilical vein endothelial cells [205]. This resulted instrong induction of sprouting in similar intensity as observed withthe addition of recombinant VEGF alone. In addition, VEGF inducesvascular hyperpermeability needed, for example, during woundhealing [206] and is known to be a chemoattractant for MSCs[207], for ECs [208,209] and for osteoclasts [210,211]. In the corneaand in bone grafts, VEGF induces growth of capillary sprouts frompreexisting blood vessels [212], whereas osteogenic commitmentis directly prevented by VEGF by inhibition of BMP-2 expression[213].

Other factors that can be used for endothelial differentiation ofMSCs include FGF and platelet-derived growth factor (PDGF). FGFstimulates EC proliferation [190] and migration [214] as well asproduction of plasminogen activator and collagenase, which playa role in the clotting system [215]. After implantation on the mes-

enteric membrane in rat peritoneum, poly-(lactic-co-glycolic acid)-based microspheres releasing FGF induced the formation of largeand mature blood vessels [216]. However, these angiogenic effectsof FGF contrast with the fact that vascular development is nothampered in FGF-deficient mice, suggesting highly restricted rolesfor FGF under normal developmental and physiological conditions[217].

Within the PDGF family four members have been identified: theclassical PDGFs, PDGF-A and PDGF-B, which have been studiedintensively for more than 20 years; and the novel PDGFs, PDGF-Cand PDGF-D, which were only recently discovered [218]. PDGF-Aand PDGF-B bind as disulphide-linked homo- or heterodimers totheir tyrosine kinase receptors (PDGF receptor a and PDGF recep-tor b) and are physiologically synthesized by platelets, monocytes,macrophages and endothelial cells [219,220]. Capillary endothelialcells stimulated by PDGF-BB not only increase DNA synthesis[221], but also form angiogenic sprouts in vitro [222,223].

Representative protocols used for in vitro endothelial differenti-ation of MSCs including VEGF as well as commercially availablemedia are shown in Tables 7 and 8.

6. Vascular smooth muscle differentiation

6.1. Background

VSMCs play an important role in angiogenesis, mechanical sup-port of vessels and blood pressure control [224]. Thus, generating afunctional VSMC layer is a prerequisite for successful constructionof tissue-engineered blood vessels. The replicative ability of autol-ogous VSMCs derived from older donors, representing the majorityof potential recipients of vascular grafts for treatment of cardiovas-cular diseases, is limited [225,226]. Thus, MSCs represent anappealing source of smooth muscle progenitor cells for vascularengineering approaches.

6.2. Morphology and differentiation markers

VSMCs demonstrate both a synthetic and a contractile pheno-type. The synthetic phenotype demonstrates fibroblast-likemorphology, high proliferation rate and production of ECMcomprised of collagen, elastin and proteoglycans, which allowsfor elasticity and improved compliance [227]. In contrast, contrac-tile VSMCs are spindle-shaped with a prominent, centrally locatednucleus, proliferate at an extremely low rate but are capable ofcontraction [228]. Contraction is predominantly mediated by

Page 9: Culture Media for the Differentiation of Mesenchymal Stromal Cells

C. Vater et al. / Acta Biomaterialia 7 (2011) 463–477 471

Ca2+-sensitive ion channels [229,230]. Microscopically, contractil-ity can be measured by a collagen gel lattice contraction assay incombination with stimulative agents, e.g. potassium chloride orcarbachol [227,231].

On the molecular level there are several markers commonlyused to assess VSMC phenotype, including a-smooth muscle actin(a-SMA), transgelin (SM22-a), calponin (Fig. 2E), caldesmon,smooth muscle myosin heavy chain (SMMHC) and smoothelin[232–234]. a-SMA and SM22-a are early markers of smooth mus-cle differentiation, whereas calponin, caldesmon and SMMHC aremarkers of late-stage VSMC differentiation. Smoothelin, a cytoskel-etal protein that is only found in contractile VSMCs, is known to bea marker of mature VSMCs [233]. Transcriptional activation of theVSMC marker genes described above is mediated by myocardin,GATA-binding protein 6 (Gata-6) and serum response factor (SRF)[230].

6.3. Differentiation protocols

The most potent inducer of VSMC differentiation of MSCs isTGF-b1. During the differentiation process it plays a role in bothinitial commitment and further differentiation of VSMCs [235].Through activation of the transcription factors Gata-6 and SRF,TGF-b1 mediates the upregulation of the VSMC marker genesa-SMA, SM22-a, calponin and SMMHC in MSCs [236,237].SMC-specific transcription is regulated by SRF, which binds toCArG (CC(A=T)6GG) cis elements that are found in the promotersof almost all SMC marker genes [238–241]. Gong et al. showed thata concentration of 0.1–10 ng ml�1 TGF-b1 inhibited human MSCproliferation, but increased calponin expression in a dose-depen-dent manner [234], correlating well with expression levels foundin human coronary arterial smooth muscle cells. TGF-b1 deficiencyis correlated with atherosclerosis and stenosis of the majorcoronary vessels [242]. In contrast to TGF-b1, disruption of theTGF-b2 gene results in 100% mortality just prior to or just afterbirth, and affects developmental processes that are involved in epi-thelial–mesenchymal transitions, cell growth, ECM production andtissue remodeling [243]. These studies indicate that the presenceof all TGF-b isoforms is important for proper maturation of smoothmuscle cells [244].

PDGF is known to be a growth factor influencing VSMC differen-tiation of MSCs. However, the role of PDGF-BB in SMC differentia-

Table 9Representative protocols used for in vitro vascular smooth muscle differentiation of MSCsprotein-4; FCS, fetal calf serum; HGF, hepatocyte growth factor; PDGF, platelet-derived growtissue). Vascular smooth muscle cells demonstrate a synthetic and a contractile phenotypemarker proteins. However, most of the protocols listed below mainly focus on the expressihave also addressed the contraction ability of differentiated MSCs (e.g. Wang et al. [231])

Reference FCS TGF-b1

Kanematsu et al. [271] 0.5% 5 ng ml�1

Ross et al. [230] - 2.5 ng ml�1

Shukla et al. [272] 0.5/5% 5 ng ml�1

Narita et al. [249] 20% 0.1/1/10 ng ml�1

Gong et al. [234] 10% 0.01/0.1/1/10 ng ml�1

Kurpinski et al. [273] 10% 5 ng ml�1

Gong et al. [6] 10% 1 ng ml�1

Harris et al. [227] 13% 2 ng ml�1

Wang et al. [231] 1% 5 ng ml�1

tion is controversial. During embryonic development, PDGF-BB isinvolved in the recruitment of pericytes and SMC precursors to-wards endothelial cells [245]. Postnatally, PDGF-BB is associatedwith SMC proliferation, e.g. in response to vascular injury and ath-erosclerosis [224]. Thus, PDGF-BB appears to be responsible forphenotypic modulation and dedifferentiation of SMCs in vivo.There is also evidence that PDGF-BB suppresses expression ofmarkers consistent with terminal differentiation in vitro, includingSM22-a and SMMHC [246], and induces a proliferative and syn-thetic VSMC phenotype [224]. However, studies by Dandre et al.show that PDGF-BB-induced inhibition of SMC marker expressionis only observed in low-density cultures but not when cells aregrowth-arrested by culture at subconfluent density in serum-freeconditions or when maintained at high cell density in serum-richconditions [246].

In addition to TGF-b1 and PDGF-BB, factors such as angiotensinII [247], sphingosylphosphorylcholine [248] and ascorbic acid[249] have also been applied for VSMC differentiation. Further-more, close cell-to-cell contact is required for VSMC differentiation[250].

Several protocols are available for the differentiation of MSCsinto VSMCs. Representative protocols for this, including TGF-b1and PDGF-BB, are shown in Table 9. Selected commercially avail-able media for VSMC differentiation of MSCs are listed in Table 10.

7. General aspects

7.1. Cell source

MSCs can be isolated from various tissues, including bonemarrow, adipose tissue, umbilical cord blood, placental tissue[251], liver, spleen [20], testes [19], menstrual blood, amniotic fluid[17], pancreas [16], dermis [252], dental pulp [253], periosteum[254] and even lung [255]. Within these, bone marrow is believedto be the enriched reservoir of MSCs and the major source for theseprecursor cells, which populate other adult tissues and organs[256]. Because of their abundant availability and easy accessibility,MSCs derived from adipose tissue, in particular, are considered tobe an attractive alternative to MSCs from bone marrow. However,in terms of their osteogenic potential, adipose tissue-derived MSCsseem to be inferior to bone-marrow-derived MSCs [199]. For MSCsfrom umbilical cord blood (UCB) conflicting results in terms of

(asc, ascorbic acid; bFGF, basic fibroblast growth factor; BMP-4, bone morphogeneticth factor; TGF-b1, transforming growth factor-beta 1; BM, bone marrow; AT, adiposethat is characterized by morphology, proliferation rate, and the expression of special

on of smooth muscle-specific genes and proteins, respectively. Only a few researchers.

Other Cell source

HGF (40 ng ml�1) Rat BMPDGF-BB (0–100 ng ml�1) Murine/rat/human/porcine BMPDGF-BB (25 ng ml�1) Porcine BMasc (30/300 lM) Human BMPDGF-BB (10 ng ml�1)PDGF-CC (10 ng ml�1)bFGF (10 ng ml�1)asc (50 lg ml�1)

Human BM

– Human MSCs (Cambrex)Copper sulfate (3 ng ml�1)proline (50 mg ml�1)glycine (50 mg ml�1)ascorbic acid (50 mg ml�1)alanine (20 mg ml�1)

Human BM

Heparin (7.5 U ml�1)angiotensin II (2 lM)sphingosylphosphorylcholine (2 lM)

Human AT

BMP-4 (2.5 ng ml�1) Human AT

Page 10: Culture Media for the Differentiation of Mesenchymal Stromal Cells

Table 10Media commercially available for vascular smooth muscle differentiation of MSCs.

Company Product

PromoCell GmbH(Heidelberg, Germany)

Smooth Muscle Cell Growth Medium 2

Invitrogen (Carlsbad, USA) Medium 231 Smooth MuscleDifferentiation Supplement (SMDS)

BD Biosciences (Bedford, USA) SMC Differentiation Medium

472 C. Vater et al. / Acta Biomaterialia 7 (2011) 463–477

success rate of MSC isolation have been initially reported, probablybecause of their low frequency in UCB [257,258].

In general, MSCs are a minor fraction in bone marrow and othertissues; the exact frequency is difficult to calculate because of thedifferent methods of harvest and separation. However, the fre-quency in human bone marrow has been estimated to be of the or-der of 0.001–0.01% of the total nucleated cells, and therefore about10-fold less abundant than haematopoietic stem cells. Further-more, the frequency of MSCs declines with age, from 1/10,000nucleated marrow cells in a newborn to about 1/10,00,000 nucle-ated marrow cells in a 80 year old person [259].

7.2. Use of FCS

Most of the protocols used today recommend the use of FCS-supplemented medium for the differentiation of MSCs because ofthe nutrients and growth factors present in the FCS. While MSCsserum concentrations in the range of 10–20% are used forexpansion, differentiation of MSCs occurs mainly in medium sup-plemented with 2–10% FCS. However, there is a considerablebatch-to-batch variation within the FCS, which may result in a sig-nificant variation in MSC properties and differentiation behavior,respectively. Therefore it is important to perform a FCS batch testprior to a study to find the batch that is most suitable for therespective experiment. In addition, it must be kept in mind thatFCS is an undesirable additive to cells that are expanded and differ-entiated for therapeutic purposes in humans because the use ofFCS carries the risk of transmitting viral and prion diseases andproteins that may initiate xenogeneic immune responses. Becauseof this, various alternatives have been considered, including, forexample, (autologous) human platelet lysates or serum [260,261]as well as formulated defined media.

8. Summary

MSCs are a promising source of precursor cells which may be ap-plied in various tissue-engineering strategies. By using differentia-tion-specific protocols, MSCs can be induced to differentiatetowards a variety of mature target cells. In this context, the compo-sition of the cell culture media used is crucial. Currently, differentcommercial culture media with or without FCS are available forMSC differentiation. More importantly, however, particular supple-mentary agents need to be added to the media for lineage-specificdifferentiation of MSCs. In general, culture supplements requiredfor osteogenic differentiation include dex, b-GP, asc, vitD3, TGF-band BMPs. Chondrogenic differentiation of MSCs is achieved bygrowing cells in pellet with media containing dex, asc, TGF-b, BMPsand IGF. Supplements including dex, IBMX, insulin, indomethacinand T3 have been shown to be effective in adipogenic differentiation.In order to induce MSC differentiation towards endothelial cell line-age VEGF is routinely used as supplementary agent. MSC differenti-ation towards the VSMC phenotype is achieved by stimulating thecells with TGF-b1 and PDGF-BB.

In practice, the combination of FCS and these supplementaryagents such as growth factors and cytokines and their concentra-tion should be optimized to meet each individual need.

Disclosure of interests

The authors indicate no potential conflict of interests.

Acknowledgements

We thank Peter Bernstein, MD, Juliane Rauh, PhD, Angela Jacobi,PhD, and Mike Tipsword for proof-reading of this manuscript.Thanks a lot to Cornelia Liebers and Suzanne Manthey for assistingwith cell culture and histology. M.S. and C.V. are financially sup-ported by the German Academic Exchange Service/German FederalMinistry of Education and Research (Grant No. D/09/04774) and bythe Center for Regenerative Therapies Dresden, Germany (SeedGrant No. 09-08).

Appendix A. Figures with essential colour discrimination

Certain figures in this article, particularly Figures 1 and 2, are dif-ficult to interpret in black and white. The full colour images can befound in the on-line version, at doi:10.1016/j.actbio.2010.07.037.

References

[1] Owen M, Friedenstein AJ. Stromal stem cells: marrow-derived osteogenicprecursors. Ciba Found Symp 1988;136:42–60.

[2] Mackay AM, Beck SC, Murphy JM, Barry FP, Chichester CO, Pittenger MF.Chondrogenic differentiation of cultured human mesenchymal stem cellsfrom marrow. Tissue Eng 1998;4:415–28.

[3] Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al.Multilineage potential of adult human mesenchymal stem cells. Science1999;284:143–7.

[4] Drost AC, Weng S, Feil G, Schäfer J, Baumann S, Kanz L, et al. In vitro myogenicdifferentiation of human bone marrow-derived mesenchymal stem cells as apotential treatment for urethral sphincter muscle repair. Ann NY Acad Sci2009;1176:135–43.

[5] Oswald J, Boxberger S, Jørgensen B, Feldmann S, Ehninger G, Bornhäuser M,et al. Mesenchymal stem cells can be differentiated into endothelial cellsin vitro. Stem Cells 2004;22:377–84.

[6] Gong Z, Calkins G, Cheng E, Krause D, Niklason LE. Influence of culturemedium on smooth muscle cell differentiation from human bone marrow-derived mesenchymal stem cells. Tissue Eng Part A 2009;15:319–30.

[7] Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D,et al. Minimal criteria for defining multipotent mesenchymal stromal cells.The International Society for Cellular Therapy position statement.Cytotherapy 2006;8:315–7.

[8] Kokai LE, Rubin JP, Marra KG. The potential of adipose-derived adult stemcells as a source of neuronal progenitor cells. Plast Reconstr Surg2005;116:1453–60.

[9] Safford KM, Rice HE. Stem cell therapy for neurologic disorders: therapeuticpotential of adipose-derived stem cells. Curr Drug Targets 2005;6:57–62.

[10] Phinney DG, Isakova I. Plasticity and therapeutic potential of mesenchymalstem cells in the nervous system. Curr Pharm Des 2005;11:1255–65.

[11] Kassem M. Mesenchymal stem cells: biological characteristics and potentialclinical applications. Cloning Stem Cells 2004;6:369–74.

[12] Di Gioacchino G, Di Campli C, Zocco MA, Piscaglia AC, Novi M, Santoro M,et al. Transdifferentiation of stem cells in pancreatic cells: state of the art.Transplant Proc 2005;37:2662–3.

[13] Timper K, Seboek D, Eberhardt M, Linscheid P, Christ-Crain M, Keller U, et al.Human adipose tissue-derived mesenchymal stem cells differentiate intoinsulin, somatostatin, and glucagon expressing cells. Biochem Biophys ResCommun 2006;341:1135–40.

[14] Moriscot C, de Fraipont F, Richard M, Marchand M, Savatier P, Bosco D, et al.Human bone marrow mesenchymal stem cells can express insulin and keytranscription factors of the endocrine pancreas developmental pathway upongenetic and/or microenvironmental manipulation in vitro. Stem Cells2005;23:594–603.

[15] Chen L, Jiang X, Yang L. Differentiation of rat marrow mesenchymal stem cellsinto pancreatic islet beta-cells. World J Gastroenterol 2004;10:3016–20.

[16] Kruse C, Kajahn J, Petschnik AE, Maass A, Klink E, Rapoport DH, et al. Adultpancreatic stem/progenitor cells spontaneously differentiate in vitro intomultiple cell lineages and form teratoma-like structures. Ann Anat2006;188:503–17.

[17] De Coppi P, Bartsch GJ, Siddiqui MM, Xu T, Santos CC, Perin L, et al. Isolation ofamniotic stem cell lines with potential for therapy. Nat Biotechnol2007;25:100–6.

[18] Meng X, Ichim TE, Zhong J, Rogers A, Yin Z, Jackson J, et al. Endometrialregenerative cells: a novel stem cell population. J Transl Med 2007;5:57.

Page 11: Culture Media for the Differentiation of Mesenchymal Stromal Cells

C. Vater et al. / Acta Biomaterialia 7 (2011) 463–477 473

[19] Guan K, Nayernia K, Maier LS, Wagner S, Dressel R, Lee JH, et al. Pluripotencyof spermatogonial stem cells from adult mouse testis. Nature2006;440:1199–203.

[20] in’t Anker P, Noort W, Scherjon S, Kleijburg-van der Keur C, Kruisselbrink A,van Bezooijen R, et al. Mesenchymal stem cells in human second-trimesterbone marrow, liver, lung, and spleen exhibit a similar immunophenotype buta heterogeneous multilineage differentiation potential. Haematologica2003;88:845–52.

[21] Javazon EH, Beggs KJ, Flake AW. Mesenchymal stem cells: paradoxes ofpassaging. Exp Hematol 2004;32:414–25.

[22] Chamberlain G, Fox J, Ashton B, Middleton J. Concise review: mesenchymalstem cells: their phenotype, differentiation capacity, immunological features,and potential for homing. Stem Cells 2007;25:2739–49.

[23] Haynesworth S, Baber M, Caplan A. Cell surface antigens on human marrow-derived mensenchymal cells are detected by monoclonal antibodies. Bone1992;13(1):69–80.

[24] Lodie TA, Blickarz CE, Devarakonda TJ, He C, Dash AB, Clarke J, et al.Systematic analysis of reportedly distinct populations of multipotent bonemarrow-derived stem cells reveals a lack of distinction. Tissue Eng2002;8:739–51.

[25] Suva D, Garavaglia G, Menetrey J, Chapuis B, Hoffmeyer P, Bernheim L, et al.Non-hematopoietic human bone marrow contains long-lasting,pluripotential mesenchymal stem cells. J Cell Physiol 2004;198:110–8.

[26] Niemeyer P, Seckinger A, Simank HG, Kasten P, Südkamp N, Krause U.Allogenic transplantation of human mesenchymal stem cells for tissueengineering purposes: an in vitro study. Orthopade 2004;33:1346–53.

[27] Di Nicola M, Carlo-Stella C, Magni M, Milanesi M, Longoni PD, Matteucci P, et al.Human bone marrow stromal cells suppress T-lymphocyte proliferationinduced by cellular or nonspecific mitogenic stimuli. Blood 2002;99:3838–43.

[28] Bartholomew A, Sturgeon C, Siatskas M, Ferrer K, McIntosh K, Patil S, et al.Mesenchymal stem cells suppress lymphocyte proliferation in vitro andprolong skin graft survival in vivo. Exp Hematol 2002;30:42–8.

[29] Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulateallogeneic immune cell responses. Blood 2005;105:1815–22.

[30] Jiang X, Zhang Y, Liu B, Zhang S, Wu Y, Yu X, et al. Human mesenchymal stemcells inhibit differentiation and function of monocyte-derived dendritic cells.Blood 2005;105:4120–6.

[31] Beyth S, Borovsky Z, Mevorach D, Liebergall M, Gazit Z, Aslan H, et al. Humanmesenchymal stem cells alter antigen-presenting cell maturation and induceT-cell unresponsiveness. Blood 2005;105:2214–9.

[32] Corcione A, Benvenuto F, Ferretti E, Giunti D, Cappiello V, Cazzanti F, et al. Humanmesenchymal stem cells modulate B-cell functions. Blood 2006;107:367–72.

[33] Bernstein P, Bornhäuser M, Günther K, Stiehler M. Bone tissue engineering inclinical application: assessment of the current situation. Orthopäde2009;38(11):1029–37.

[34] Orlic D, Kajstura J, Chimenti S, Limana F, Jakoniuk I, Quaini F, et al. Mobilizedbone marrow cells repair the infarcted heart, improving function andsurvival. Proc Natl Acad Sci USA 2001;98:10344–9.

[35] Lee ST, Jang JH, Cheong J, Kim JS, Maemg H, Hahn JS, et al. Treatment of high-risk acute myelogenous leukaemia by myeloablative chemoradiotherapyfollowed by co-infusion of T cell-depleted haematopoietic stem cells andculture-expanded marrow mesenchymal stem cells from a related donorwith one fully mismatched human leucocyte antigen haplotype. Br JHaematol 2002;118:1128–31.

[36] Urbán VS, Kiss J, Kovács J, Gócza E, Vas V, Monostori E, et al. Mesenchymalstem cells cooperate with bone marrow cells in therapy of diabetes. StemCells 2008;26:244–53.

[37] Guhathakurta S, Subramanyan UR, Balasundari R, Das CK, Madhusankar N,Cherian KM. Stem cell experiments and initial clinical trial of cellularcardiomyoplasty. Asian Cardiovasc Thorac Ann 2009;17:581–6.

[38] Horwitz EM, Gordon PL, Koo WKK, Marx JC, Neel MD, McNall RY, et al.Isolated allogeneic bone marrow-derived mesenchymal cells engraft andstimulate growth in children with osteogenesis imperfecta: implications forcell therapy of bone. Proc Natl Acad Sci USA 2002;99:8932–7.

[39] Constantin G, Marconi S, Rossi B, Angiari S, Calderan L, Anghileri E, et al.Adipose-derived mesenchymal stem cells ameliorate chronic experimentalautoimmune encephalomyelitis. Stem Cells 2009;27:2624–35.

[40] Newman RE, Yoo D, LeRoux MA, Danilkovitch-Miagkova A. Treatment ofinflammatory diseases with mesenchymal stem cells. Inflamm Allergy DrugTargets 2009;8:110–23.

[41] Sorger J, Hornicek F, Zavatta M, Menzner J, Gebhardt M, Tomford W, et al.Allograft fractures revisited. Clin Orthop Relat Res 2001;Jan(382):66–74.

[42] Song I, Caplan AI, Dennis JE. In vitro dexamethasone pretreatment enhancesbone formation of human mesenchymal stem cells in vivo. J Orthop Res2009;27:916–21.

[43] Ducy P, Zhang R, Geoffroy V, Ridall AL, Karsenty G. Osf2/Cbfa1: atranscriptional activator of osteoblast differentiation. Cell 1997;89:747–54.

[44] Ducy P, Starbuck M, Priemel M, Shen J, Pinero G, Geoffroy V, et al. A Cbfa1-dependent genetic pathway controls bone formation beyond embryonicdevelopment. Genes Dev 1999;13:1025–36.

[45] Kern B, Shen J, Starbuck M, Karsenty G. Cbfa1 contributes to theosteoblast-specific expression of type I collagen genes. J Biol Chem2001;276:7101–7.

[46] Nakashima K, Zhou X, Kunkel G, Zhang Z, Deng JM, Behringer RR, et al. Thenovel zinc finger-containing transcription factor osterix is required forosteoblast differentiation and bone formation. Cell 2002;108:17–29.

[47] Lian JB, Stein GS, Javed A, van Wijnen AJ, Stein JL, Montecino M, et al.Networks and hubs for the transcriptional control of osteoblastogenesis. RevEndocr Metab Disord 2006;7:1–16.

[48] Romero-Prado M, Blázquez C, Rodríguez-Navas C, Muñoz J, Guerrero I,Delgado-Baeza E, et al. Functional characterization of human mesenchymalstem cells that maintain osteochondral fates. J Cell Biochem2006;98:1457–70.

[49] Spector JA, Luchs JS, Mehrara BJ, Greenwald JA, Smith LP, Longaker MT.Expression of bone morphogenetic proteins during membranous bonehealing. Plast Reconstr Surg 2001;107:124–34.

[50] Zhu JX, Sasano Y, Takahashi I, Mizoguchi I, Kagayama M. Temporal and spatialgene expression of major bone extracellular matrix molecules duringembryonic mandibular osteogenesis in rats. Histochem J 2001;33:25–35.

[51] Long MW. Osteogenesis and bone-marrow-derived cells. Blood CellsMolecules Dis 2001;27:677–90.

[52] Young MF. Bone matrix proteins: their function, regulation, and relationshipto osteoporosis. Osteoporos Int 2003;14(Suppl. 3):S35–42.

[53] Leboy PS, Beresford JN, Devlin C, Owen ME. Dexamethasone induction ofosteoblast mRNAs in rat marrow stromal cell cultures. J Cell Physiol1991;146:370–8.

[54] Herbertson A, Aubin JE. Dexamethasone alters the subpopulation make-up ofrat bone marrow stromal cell cultures. J Bone Miner Res 1995;10:285–94.

[55] Hildebrandt C, Büth H, Thielecke H. Influence of cell culture media conditionson the osteogenic differentiation of cord blood-derived mesenchymal stemcells. Ann Anat 2009;191:23–32.

[56] Ogata Y, Yamauchi M, Kim R, Li J, Freedman L, Sodek J. Glucocorticoidregulation of bone sialoprotein (BSP) gene expression. Identification of aglucocorticoid response element in the bone sialoprotein gene promoter. EurJ Biochem 1995;230(1):183–92.

[57] Hong D, Chen H, Xue Y, Li D, Wan X, Ge R, et al. Osteoblastogenic effects ofdexamethasone through upregulation of TAZ expression in rat mesenchymalstem cells. J Steroid Biochem Mol Biol 2009;116:86–92.

[58] Hamidouche Z, Fromigué O, Ringe J, Häupl T, Vaudin P, Pagès J, et al. Primingintegrin alpha5 promotes human mesenchymal stromal cell osteoblastdifferentiation and osteogenesis. Proc Natl Acad Sci USA 2009;106(44):18587–91.

[59] Jørgensen NR, Henriksen Z, Sørensen OH, Civitelli R. Dexamethasone, BMP-2,and 1, 25-dihydroxyvitamin D enhance a more differentiated osteoblastphenotype: validation of an in vitro model for human bone marrow-derivedprimary osteoblasts. Steroids 2004;69:219–26.

[60] Walsh S, Jordan GR, Jefferiss C, Stewart K, Beresford JN. High concentrations ofdexamethasone suppress the proliferation but not the differentiation or furthermaturation of human osteoblast precursors in vitro: relevance toglucocorticoid-induced osteoporosis. Rheumatology (Oxford) 2001;40:74–83.

[61] Lukert BP. Clinical and basic aspects of glucocorticoids action in bone. In:Bilezikian JP, Raisz LG, Rodan GA, editors. Principles of bone biology. SanDiego: Academic Press; 1996. p. 533–48.

[62] Gundle R. Microscopical and biochemical studies of mineralized matrixproduction by human bone derived cells. Ph.D. Thesis, University of Oxford;1995.

[63] Song I, Caplan AI, Dennis JE. Dexamethasone inhibition of confluence-inducedapoptosis in human mesenchymal stem cells. J Orthop Res 2009;27:216–21.

[64] Chang YL, Stanford CM, Keller JC. Calcium and phosphate supplementationpromotes bone cell mineralization: implications for hydroxyapatite (HA)-enhanced bone formation. J Biomed Mater Res 2000;52:270–8.

[65] Chung C, Golub E, Forbes E, Tokuoka T, Shapiro I. Mechanism of action ofbeta-glycerophosphate on bone cell mineralization. Calcif Tissue Int1992;51(4):305–11.

[66] Chen M, Lie P, Li Z, Wei X. Endothelial differentiation of Wharton’s jelly-derived mesenchymal stem cells in comparison with bone marrow-derivedmesenchymal stem cells. Exp Hematol 2009;37:629–40.

[67] Kielty C, Hopkinson I, Grant M. Collagen: the collagen family: structure,assembly, and organization in the extracellular matrix. In: Royce SB,Steinmann B, editors. Connective tissue and its heritable disorders.Molecular, genetic, and medical aspects. New York: Wiley-Liss; 1993. p.103–47.

[68] Prockop DJ, Kivirikko KI. Collagens: molecular biology, diseases, andpotentials for therapy. Annu Rev Biochem 1995;64:403–34.

[69] Takamizawa S, Maehata Y, Imai K, Senoo H, Sato S, Hata R. Effects of ascorbicacid and ascorbic acid 2-phosphate, a long-acting vitamin C derivative, on theproliferation and differentiation of human osteoblast-like cells. Cell Biol Int2004;28:255–65.

[70] Maehata Y, Takamizawa S, Ozawa S, Kato Y, Sato S, Kubota E, et al. Both directand collagen-mediated signals are required for active vitamin D3-eliciteddifferentiation of human osteoblastic cells: roles of osterix, an osteoblast-related transcription factor. Matrix Biol 2006;25:47–58.

[71] Hata R, Senoo H. L-Ascorbic acid 2-phosphate stimulates collagenaccumulation, cell proliferation, and formation of a three-dimensionaltissue-like substance by skin fibroblasts. J Cell Physiol 1989;138:8–16.

[72] Fernandes H, Mentink A, Bank RA, Stoop R, van Blitterswijk C, De Boer J.Endogenous collagen influences differentiation of human multipotentmesenchymal stromal cells. Tissue Eng Part A 2009;16(5):1693–702.

[73] Pytlík R, Stehlík D, Soukup T, Kalbácová M, Rypácek F, Trc T, et al. Thecultivation of human multipotent mesenchymal stromal cells in clinicalgrade medium for bone tissue engineering. Biomaterials 2009;30:3415–27.

Page 12: Culture Media for the Differentiation of Mesenchymal Stromal Cells

474 C. Vater et al. / Acta Biomaterialia 7 (2011) 463–477

[74] Wang QM, Jones JB, Studzinski GP. Cyclin-dependent kinase inhibitor p27 as amediator of the G1-S phase block induced by 1,25-dihydroxyvitamin D3 inHL60 cells. Cancer Res 1996;56:264–7.

[75] Verlinden L, Verstuyf A, Convents R, Marcelis S, Van Camp M, Bouillon R.Action of 1,25(OH)2D3 on the cell cycle genes, cyclin D1, p21 and p27 in MCF-7 cells. Mol Cell Endocrinol 1998;142:57–65.

[76] Nakagawa K, Tsugawa N, Okamoto T, Kishi T, Ono T, Kubodera N, et al. Rapidcontrol of transmembrane calcium influx by 1alpha, 25-dihydroxyvitamin D3and its analogues in rat osteoblast-like cells. Biol Pharm Bull1999;22:1058–63.

[77] Fromigué O, Marie PJ, Lomri A. Differential effects of transforming growthfactor beta2, dexamethasone and 1, 25-dihydroxyvitamin D on human bonemarrow stromal cells. Cytokine 1997;9:613–23.

[78] Jaiswal N, Haynesworth SE, Caplan AI, Bruder SP. Osteogenic differentiation ofpurified, culture-expanded human mesenchymal stem cells in vitro. J CellBiochem 1997;64:295–312.

[79] Bonewald LF, Wakefield L, Oreffo RO, Escobedo A, Twardzik DR, Mundy GR.Latent forms of transforming growth factor-beta (TGF beta) derived frombone cultures: identification of a naturally occurring 100-kDa complex withsimilarity to recombinant latent TGF beta. Mol Endocrinol 1991;5:741–51.

[80] Dallas SL, Park-Snyder S, Miyazono K, Twardzik D, Mundy GR, Bonewald LF.Characterization and autoregulation of latent transforming growth factorbeta (TGF beta) complexes in osteoblast-like cell lines. Production of a latentcomplex lacking the latent TGF beta-binding protein. J Biol Chem1994;269:6815–21.

[81] Centrella M, McCarthy TL, Canalis E. Transforming growth factor beta is abifunctional regulator of replication and collagen synthesis in osteoblast-enriched cell cultures from fetal rat bone. J Biol Chem 1987;262:2869–74.

[82] Centrella M, McCarthy TL, Canalis E. Transforming growth factor-beta andremodeling of bone. J Bone Joint Surg Am 1991;73:1418–28.

[83] Jäger M, Sager M, Knipper A, Degistirici O, Fischer J, Kögler G, et al. In vivo andin vitro bone regeneration from cord blood derived mesenchymal stem cells.Orthopäde 2004;33:1361–72.

[84] Hogan BL. Bone morphogenetic proteins in development. Curr Opin GenetDev 1996;6:432–8.

[85] Holleville N, Quilhac A, Bontoux M, Monsoro-Burq AH. BMP signals regulateDlx5 during early avian skull development. Dev Biol 2003;257:177–89.

[86] Hanada K, Dennis JE, Caplan AI. Stimulatory effects of basic fibroblast growthfactor and bone morphogenetic protein-2 on osteogenic differentiation of ratbone marrow-derived mesenchymal stem cells. J Bone Miner Res1997;12:1606–14.

[87] Diefenderfer DL, Osyczka AM, Reilly GC, Leboy PS. BMP responsiveness inhuman mesenchymal stem cells. Connect Tissue Res 2003;44(Suppl.1):305–11.

[88] Knippenberg M, Helder MN, Zandieh Doulabi B, Wuisman PIJM, Klein-NulendJ. Osteogenesis versus chondrogenesis by BMP-2 and BMP-7 in adipose stemcells. Biochem Biophys Res Commun 2006;342:902–8.

[89] Abe E, Yamamoto M, Taguchi Y, Lecka-Czernik B, O’Brien CA, Economides AN,et al. Essential requirement of BMPs-2/4 for both osteoblast and osteoclastformation in murine bone marrow cultures from adult mice: antagonism bynoggin. J Bone Miner Res 2000;15:663–73.

[90] Lou J, Tu Y, Li S, Manske PR. Involvement of ERK in BMP-2 inducedosteoblastic differentiation of mesenchymal progenitor cell line C3H10T1/2.Biochem Biophys Res Commun 2000;268:757–62.

[91] Cheng H, Jiang W, Phillips FM, Haydon RC, Peng Y, Zhou L, et al. Osteogenicactivity of the fourteen types of human bone morphogenetic proteins (BMPs).J Bone Joint Surg Am 2003;85-A:1544–52.

[92] Osyczka AM, Diefenderfer DL, Bhargave G, Leboy PS. Different effects of BMP-2 on marrow stromal cells from human and rat bone. Cells Tissues Organs2004;176:109–19.

[93] Cheifetz S, Li IW, McCulloch CA, Sampath K, Sodek J. Influence of osteogenicprotein-1 (OP-1;BMP-7) and transforming growth factor-beta 1 on boneformation in vitro. Connect Tissue Res 1996;35:71–8.

[94] Li IW, Cheifetz S, McCulloch CA, Sampath KT, Sodek J. Effects of osteogenicprotein-1 (OP-1, BMP-7) on bone matrix protein expression by fetal ratcalvarial cells are differentiation stage specific. J Cell Physiol1996;169:115–25.

[95] Lecanda F, Avioli LV, Cheng SL. Regulation of bone matrix protein expressionand induction of differentiation of human osteoblasts and human bonemarrow stromal cells by bone morphogenetic protein-2. J Cell Biochem1997;67:386–96.

[96] Locklin RM, Riggs BL, Hicok KC, Horton HF, Byrne MC, Khosla S.Assessment of gene regulation by bone morphogenetic protein 2 inhuman marrow stromal cells using gene array technology. J Bone MinerRes 2001;16:2192–204.

[97] Fuller JA, Ghadially FN. Ultrastructural observations on surgically producedpartial-thickness defects in articular cartilage. Clin Orthop Relat Res1972;86:193–205.

[98] Nesic D, Whiteside R, Brittberg M, Wendt D, Martin I, Mainil-Varlet P.Cartilage tissue engineering for degenerative joint disease. Adv Drug DelivRev 2006;58:300–22.

[99] Koga H, Engebretsen L, Brinchmann JE, Muneta T, Sekiya I. Mesenchymal stemcell-based therapy for cartilage repair: a review. Knee Surg Sports TraumatolArthrosc 2009;17:1289–97.

[100] Hardingham TE, Oldershaw RA, Tew SR. Cartilage, SOX9 and Notch signals inchondrogenesis. J Anat 2006;209:469–80.

[101] Chang Y, Ueng SWN, Lin-Chao S, Chao CC. Involvement of Gas7 along theERK1/2 MAP kinase and SOX9 pathway in chondrogenesis of human marrow-derived mesenchymal stem cells. Osteoarthritis Cartilage 2008;16:1403–12.

[102] An C, Cheng Y, Yuan Q, Li J. IGF-1 and BMP-2 induces differentiation ofadipose-derived mesenchymal stem cells into chondrocytes-like cells. AnnBiomed Eng 2010;38(4):1647–54.

[103] Choi K, Seo Y, Yoon H, Song K, Kwon S, Lee H, et al. Effect of ascorbic acid onbone marrow-derived mesenchymal stem cell proliferation anddifferentiation. J Biosci Bioeng 2008;105:586–94.

[104] Lefebvre V, Huang W, Harley VR, Goodfellow PN, de Crombrugghe B. SOX9 isa potent activator of the chondrocyte-specific enhancer of the pro alpha1(II)collagen gene. Mol Cell Biol 1997;17:2336–46.

[105] Bridgewater LC, Lefebvre V, de Crombrugghe B. Chondrocyte-specificenhancer elements in the Col11a2 gene resemble the Col2a1 tissue-specificenhancer. J Biol Chem 1998;273:14998–5006.

[106] Sekiya I, Tsuji K, Koopman P, Watanabe H, Yamada Y, Shinomiya K, et al.SOX9 enhances aggrecan gene promoter/enhancer activity and is up-regulated by retinoic acid in a cartilage-derived cell line, TC6. J Biol Chem2000;275:10738–44.

[107] Zhang P, Jimenez SA, Stokes DG. Regulation of human COL9A1 geneexpression. Activation of the proximal promoter region by SOX9. J BiolChem 2003;278:117–23.

[108] Kou I, Ikegawa S. SOX9-dependent and -independent transcriptionalregulation of human cartilage link protein. J Biol Chem 2004;279:50942–8.

[109] Huang W, Lu N, Eberspaecher H, De Crombrugghe B. A new long form of c-Maf cooperates with Sox9 to activate the type II collagen gene. J Biol Chem2002;277:50668–75.

[110] Tsuda M, Takahashi S, Takahashi Y, Asahara H. Transcriptional co-activatorsCREB-binding protein and p300 regulate chondrocyte-specific geneexpression via association with Sox9. J Biol Chem 2003;278:27224–9.

[111] Schmid TM, Linsenmayer TF. Immunohistochemical localization of short chaincartilage collagen (type X) in avian tissues. J Cell Biol 1985;100:598–605.

[112] Ornitz DM, Marie PJ. FGF signaling pathways in endochondral andintramembranous bone development and human genetic disease. GenesDev 2002;16:1446–65.

[113] Shen G. The role of type X collagen in facilitating and regulating endochondralossification of articular cartilage. Orthod Craniofac Res 2005;8:11–7.

[114] Mwale F, Stachura D, Roughley P, Antoniou J. Limitations of using aggrecanand type X collagen as markers of chondrogenesis in mesenchymal stem celldifferentiation. J Orthop Res 2006;24:1791–8.

[115] Johnstone B, Hering TM, Caplan AI, Goldberg VM, Yoo JU. In vitrochondrogenesis of bone marrow-derived mesenchymal progenitor cells.Exp Cell Res 1998;238:265–72.

[116] Jones EA, Kinsey SE, English A, Jones RA, Straszynski L, Meredith DM, et al.Isolation and characterization of bone marrow multipotential mesenchymalprogenitor cells. Arthritis Rheum 2002;46:3349–60.

[117] Jingushi S, Heydemann A, Kana SK, Macey LR, Bolander ME. Acidic fibroblastgrowth factor (aFGF) injection stimulates cartilage enlargement and inhibitscartilage gene expression in rat fracture healing. J Orthop Res 1990;8:364–71.

[118] Yoo JU, Barthel TS, Nishimura K, Solchaga L, Caplan AI, Goldberg VM, et al. Thechondrogenic potential of human bone-marrow-derived mesenchymalprogenitor cells. J Bone Joint Surg Am 1998;80:1745–57.

[119] Mastrogiacomo M, Cancedda R, Quarto R. Effect of different growth factors onthe chondrogenic potential of human bone marrow stromal cells.Osteoarthritis Cartilage 2001;9(Suppl. A):S36–40.

[120] Na K, Choi S, Kim S, Sun BK, Woo DG, Chung H, et al. Enhancement of cellproliferation and differentiation by combination of ascorbate anddexamethasone in thermo-reversible hydrogel constructs embedded withrabbit chondrocytes. Biotechnol Lett 2007;29:1453–7.

[121] Derfoul A, Perkins GL, Hall DJ, Tuan RS. Glucocorticoids promotechondrogenic differentiation of adult human mesenchymal stem cells byenhancing expression of cartilage extracellular matrix genes. Stem Cells2006;24:1487–95.

[122] Richter W. Mesenchymal stem cells and cartilage in situ regeneration. J InternMed 2009;266:390–405.

[123] Miyazono K. Positive and negative regulation of TGF-beta signaling. J Cell Sci2000;113(Pt. 7):1101–9.

[124] Lee JW, Kim YH, Kim S, Han SH, Hahn SB. Chondrogenic differentiation ofmesenchymal stem cells and its clinical applications. Yonsei Med J2004;45(Suppl.):41–7.

[125] Barry F, Boynton RE, Liu B, Murphy JM. Chondrogenic differentiation ofmesenchymal stem cells from bone marrow: differentiation-dependent geneexpression of matrix components. Exp Cell Res 2001;268:189–200.

[126] Bosnakovski D, Mizuno M, Kim G, Ishiguro T, Okumura M, Iwanaga T, et al.Chondrogenic differentiation of bovine bone marrow mesenchymal stemcells in pellet cultural system. Exp Hematol 2004;32:502–9.

[127] Zhang X, Ziran N, Goater JJ, Schwarz EM, Puzas JE, Rosier RN, et al. Primarymurine limb bud mesenchymal cells in long-term culture completechondrocyte differentiation: TGF-beta delays hypertrophy and PGE2inhibits terminal differentiation. Bone 2004;34:809–17.

[128] Pogue R, Lyons K. BMP signaling in the cartilage growth plate. Curr Top DevBiol 2006;76:1–48.

[129] Xu D, Gechtman Z, Hughes A, Collins A, Dodds R, Cui X, et al. Potentialinvolvement of BMP receptor type IB activation in a synergistic effect ofchondrogenic promotion between rhTGFbeta3 and rhGDF5 or rhBMP7 inhuman mesenchymal stem cells. Growth Factors 2006;24:268–78.

Page 13: Culture Media for the Differentiation of Mesenchymal Stromal Cells

C. Vater et al. / Acta Biomaterialia 7 (2011) 463–477 475

[130] Hennig T, Lorenz H, Thiel A, Goetzke K, Dickhut A, Geiger F, et al. Reducedchondrogenic potential of adipose tissue derived stromal cells correlates withan altered TGFbeta receptor and BMP profile and is overcome by BMP-6. J CellPhysiol 2007;211:682–91.

[131] Sekiya I, Larson BL, Vuoristo JT, Reger RL, Prockop DJ. Comparison of effect ofBMP-2, -4, and -6 on in vitro cartilage formation of human adult stem cellsfrom bone marrow stroma. Cell Tissue Res 2005;320:269–76.

[132] Solchaga LA, Penick K, Porter JD, Goldberg VM, Caplan AI, Welter JF. FGF-2enhances the mitotic and chondrogenic potentials of human adult bonemarrow-derived mesenchymal stem cells. J Cell Physiol 2005;203:398–409.

[133] Stewart AA, Byron CR, Pondenis HC, Stewart MC. Effect of dexamethasonesupplementation on chondrogenesis of equine mesenchymal stem cells. Am JVet Res 2008;69:1013–21.

[134] Luyten FP, Hascall VC, Nissley SP, Morales TI, Reddi AH. Insulin-like growthfactors maintain steady-state metabolism of proteoglycans in bovinearticular cartilage explants. Arch Biochem Biophys 1988;267:416–25.

[135] Longobardi L, O’Rear L, Aakula S, Johnstone B, Shimer K, Chytil A, et al. Effectof IGF-I in the chondrogenesis of bone marrow mesenchymal stem cells inthe presence or absence of TGF-beta signaling. J Bone Miner Res2006;21:626–36.

[136] Nixon AJ, Goodrich LR, Scimeca MS, Witte TH, Schnabel LV, Watts AE, et al.Gene therapy in musculoskeletal repair. Ann NY Acad Sci 2007;1117:310–27.

[137] Nixon AJ, Fortier LA, Williams J, Mohammed H. Enhanced repair of extensivearticular defects by insulin-like growth factor-I-laden fibrin composites. JOrthop Res 1999;17:475–87.

[138] Weiss S, Hennig T, Bock R, Steck E, Richter W. Impact of growth factors andPTHrP on early and late chondrogenic differentiation of human mesenchymalstem cells. J Cell Physiol 2010;223:84–93.

[139] Milne M, Quail JM, Rosen CJ, Baran DT. Insulin-like growth factor bindingproteins in femoral and vertebral bone marrow stromal cells: expression andregulation by thyroid hormone and dexamethasone. J Cell Biochem2001;81:229–40.

[140] Indrawattana N, Chen G, Tadokoro M, Shann LH, Ohgushi H, Tateishi T, et al.Growth factor combination for chondrogenic induction from humanmesenchymal stem cell. Biochem Biophys Res Commun 2004;320:914–9.

[141] Matsuda C, Takagi M, Hattori T, Wakitani S, Yoshida T. Differentiation ofhuman bone marrow mesenchymal stem cells to chondrocytes forconstruction of three-dimensional cartilage tissue. Cytotechnology2005;47:11–7.

[142] Wang C, Chen L, Kuo P, Chang J, Wang Y, Hung S. Apoptosis in chondrogenesisof human mesenchymal stem cells: effect of serum and mediumsupplements. Apoptosis 2010;15(4):439–49.

[143] Eppley BL, Woodell JE, Higgins J. Platelet quantification and growth factoranalysis from platelet-rich plasma: implications for wound healing. PlastReconstr Surg 2004;114:1502–8.

[144] Mishra A, Tummala P, King A, Lee B, Kraus M, Tse V, et al. Buffered platelet-rich plasma enhances mesenchymal stem cell proliferation and chondrogenicdifferentiation. Tissue Eng Part C Methods 2009;15:431–5.

[145] Colter DC, Sekiya I, Prockop DJ. Identification of a subpopulation of rapidlyself-renewing and multipotential adult stem cells in colonies of humanmarrow stromal cells. Proc Natl Acad Sci USA 2001;98:7841–5.

[146] Huang JI, Kazmi N, Durbhakula MM, Hering TM, Yoo JU, Johnstone B.Chondrogenic potential of progenitor cells derived from human bone marrowand adipose tissue: a patient-matched comparison. J Orthop Res2005;23:1383–9.

[147] Liu TM, Martina M, Hutmacher DW, Hui JHP, Lee EH, Lim B. Identification ofcommon pathways mediating differentiation of bone marrow- and adiposetissue-derived human mesenchymal stem cells into three mesenchymallineages. Stem Cells 2007;25:750–60.

[148] Prockop DJ, Gregory CA, Spees JL. One strategy for cell and gene therapy:harnessing the power of adult stem cells to repair tissues. Proc Natl Acad SciUSA 2003;100(Suppl. 1):11917–23.

[149] Sakaguchi Y, Sekiya I, Yagishita K, Muneta T. Comparison of human stem cellsderived from various mesenchymal tissues: superiority of synovium as a cellsource. Arthritis Rheum 2005;52:2521–9.

[150] Hauner H, Löffler G. Adipose tissue development: the role of precursor cellsand adipogenic factors. Part I: Adipose tissue development and the role ofprecursor cells. Klin Wochenschr 1987;65:803–11.

[151] Langstein HN, Robb GL. Reconstructive approaches in soft tissue sarcoma.Semin Surg Oncol 1999;17:52–65.

[152] Gomillion CT, Burg KJ. Stem cells and adipose tissue engineering.Biomaterials 2006;27:6052–63.

[153] Ashinoff R. Overview: soft tissue augmentation. Clin Plast Surg2000;27:479–87.

[154] Klein AW, Elson ML. The history of substances for soft tissue augmentation.Dermatol Surg 2000;26:1096–105.

[155] Patrick CWJ. Adipose tissue engineering: the future of breast and soft tissuereconstruction following tumor resection. Semin Surg Oncol2000;19:302–11.

[156] Patrick CWJ. Tissue engineering strategies for adipose tissue repair. Anat Rec2001;263:361–6.

[157] Patrick CW, Mikos AG, McIntire LV. Frontiers in tissue engineering. 1sted. Oxford and New York: Pergamon Press; 1998.

[158] Dani C. Embryonic stem cell-derived adipogenesis. Cells Tissues Organs1999;165:173–80.

[159] Kubo Y, Kaidzu S, Nakajima I, Takenouchi K, Nakamura F. Organization ofextracellular matrix components during differentiation of adipocytes in long-term culture. In Vitro Cell Dev Biol Anim 2000;36:38–44.

[160] Rosen ED, MacDougald OA. Adipocyte differentiation from the inside out. NatRev Mol Cell Biol 2006;7:885–96.

[161] Vallée M, Côté J, Fradette J. Adipose-tissue engineering: taking advantage ofthe properties of human adipose-derived stem/stromal cells. Pathol Biol2009;57:309–17.

[162] Pairault J, Green H. A study of the adipose conversion of suspended 3T3 cellsby using glycerophosphate dehydrogenase as differentiation marker. ProcNatl Acad Sci USA 1979;76:5138–42.

[163] Hemmrich K, von Heimburg D, Cierpka K, Haydarlioglu S, Pallua N.Optimization of the differentiation of human preadipocytes in vitro.Differentiation 2005;73:28–35.

[164] Tsuji W, Inamoto T, Yamashiro H, Ueno T, Kato H, Kimura Y, et al.Adipogenesis induced by human adipose tissue derived stem cells. TissueEng Part A 2008;15:83–93.

[165] Tontonoz P, Hu E, Spiegelman BM. Stimulation of adipogenesis in fibroblastsby PPAR gamma 2, a lipid-activated transcription factor. Cell1994;79:1147–56.

[166] Rosen ED, Sarraf P, Troy AE, Bradwin G, Moore K, Milstone DS, et al. PPARgamma is required for the differentiation of adipose tissue in vivo andin vitro. Mol Cell 1999;4:611–7.

[167] Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positionalcloning of the mouse obese gene and its human homologue. Nature1994;372:425–32.

[168] Gregoire FM, Smas CM, Sul HS. Understanding adipocyte differentiation.Physiol Rev 1998;78:783–809.

[169] Zechner R, Strauss J, Frank S, Wagner E, Hofmann W, Kratky D, et al. The roleof lipoprotein lipase in adipose tissue development and metabolism. Int JObes Relat Metab Disord 2000;24(Suppl. 4):S53–6.

[170] Obregon M. Thyroid hormone and adipocyte differentiation. Thyroid2008;18:185–95.

[171] Shugart EC, Umek RM. Dexamethasone signaling is required to establish thepostmitotic state of adipocyte development. Cell Growth Differ1997;8:1091–8.

[172] Cui Q, Wang GJ, Balian G. Steroid-induced adipogenesis in a pluripotentialcell line from bone marrow. J Bone Joint Surg Am 1997;79:1054–63.

[173] Hauner H, Schmid P, Pfeiffer EF. Glucocorticoids and insulin promote thedifferentiation of human adipocyte precursor cells into fat cells. J ClinEndocrinol Metab 1987;64:832–5.

[174] Prawitt J, Niemeier A, Kassem M, Beisiegel U, Heeren J. Characterization oflipid metabolism in insulin-sensitive adipocytes differentiated fromimmortalized human mesenchymal stem cells. Exp Cell Res2008;314:814–24.

[175] Deree J, Martins JO, Melbostad H, Loomis WH, Coimbra R. Insights into theregulation of TNF-alpha production in human mononuclear cells: the effectsof non-specific phosphodiesterase inhibition. Clinics (Sao Paulo)2008;63:321–8.

[176] Yang D, Tsay H, Lin S, Chiou S, Li M, Chang T, et al. CAMP/PKA regulatesosteogenesis, adipogenesis and ratio of RANKL/OPG mRNA expression inmesenchymal stem cells by suppressing leptin. PLoS One 2008;3:e1540.

[177] Klemm DJ, Leitner JW, Watson P, Nesterova A, Reusch JE, Goalstone ML, et al.Insulin-induced adipocyte differentiation. Activation of CREB rescuesadipogenesis from the arrest caused by inhibition of prenylation. J BiolChem 2001;276:28430–5.

[178] Löffler G, Hauner H. Adipose tissue development: the role of precursor cellsand adipogenic factors. Part II: The regulation of the adipogenic conversionby hormones and serum factors. Klin Wochenschr 1987;65:812–7.

[179] Zhang HH, Huang J, Düvel K, Boback B, Wu S, Squillace RM, et al. Insulinstimulates adipogenesis through the Akt-TSC2-mTORC1 pathway. PLoS One2009;4:e6189.

[180] Rydén M, Dicker A, Götherström C, Aström G, Tammik C, Arner P, et al.Functional characterization of human mesenchymal stem cell-derivedadipocytes. Biochem Biophys Res Commun 2003;311:391–7.

[181] Skurk T, Birgel M, Lee Y, Hauner H. Effect of troglitazone on tumor necrosisfactor alpha and transforming growth factor beta expression and action inhuman adipocyte precursor cells in primary culture. Metabolism2006;55:309–16.

[182] Leyvraz C, Suter M, Verdumo C, Calmes J, Paroz A, Darimont C, et al. Selectiveeffects of PPARgamma agonists and antagonists on human pre-adipocytedifferentiation. Diabetes Obes Metab 2010;12(3):195–203.

[183] Ringold GM, Chapman AB, Knight DM, Torti FM. Hormonal control ofadipogenesis. Ann NY Acad Sci 1986;478:109–19.

[184] Kellinsalmi M, Parikka V, Risteli J, Hentunen T, Leskelä H, Lehtonen S, et al.Inhibition of cyclooxygenase-2 down-regulates osteoclast and osteoblastdifferentiation and favours adipocyte formation in vitro. Eur J Pharmacol2007;572:102–10.

[185] Lehmann JM, Lenhard JM, Oliver BB, Ringold GM, Kliewer SA. Peroxisomeproliferator-activated receptors alpha and gamma are activated byindomethacin and other non-steroidal anti-inflammatory drugs. J BiolChem 1997;272:3406–10.

[186] Ye H, Serrero G. Stimulation of adipose differentiation related protein (ADRP)expression by ibuprofen and indomethacin in adipocyte precursors and inadipocytes. Biochem J 1998;330(Pt. 2):803–9.

Page 14: Culture Media for the Differentiation of Mesenchymal Stromal Cells

476 C. Vater et al. / Acta Biomaterialia 7 (2011) 463–477

[187] Mandrup S, Lane MD. Regulating adipogenesis. J Biol Chem1997;272:5367–70.

[188] Kawaguchi N, Toriyama K, Nicodemou-Lena E, Inou K, Torii S, Kitagawa Y. Denovo adipogenesis in mice at the site of injection of basement membrane andbasic fibroblast growth factor. Proc Natl Acad Sci USA 1998;95:1062–6.

[189] Katz AJ, Llull R, Hedrick MH, Futrell JW. Emerging approaches to the tissueengineering of fat. Clin Plast Surg 1999;26:587–603 [viii].

[190] Gospodarowicz D, Ferrara N, Schweigerer L, Neufeld G. Structuralcharacterization and biological functions of fibroblast growth factor. EndocrRev 1987;8:95–114.

[191] Neubauer M, Fischbach C, Bauer-Kreisel P, Lieb E, Hacker M, Tessmar J, et al.Basic fibroblast growth factor enhances PPARgamma ligand-inducedadipogenesis of mesenchymal stem cells. FEBS Lett 2004;577:277–83.

[192] Inoue S, Hori Y, Hirano Y, Inamoto T, Tabata Y. Effect of culture substrate andfibroblast growth factor addition on the proliferation and differentiation ofhuman adipo-stromal cells. J Biomater Sci Polym Ed 2005;16:57–77.

[193] Kakudo N, Shimotsuma A, Kusumoto K. Fibroblast growth factor-2 stimulatesadipogenic differentiation of human adipose-derived stem cells. BiochemBiophys Res Commun 2007;359:239–44.

[194] Kimura Y, Ozeki M, Inamoto T, Tabata Y. Time course of de novo adipogenesisin matrigel by gelatin microspheres incorporating basic fibroblast growthfactor. Tissue Eng 2002;8:603–13.

[195] Tabata Y, Miyao M, Inamoto T, Ishii T, Hirano Y, Yamaoki Y, et al. De novoformation of adipose tissue by controlled release of basic fibroblast growthfactor. Tissue Eng 2000;6:279–89.

[196] Hauner H, Entenmann G, Wabitsch M, Gaillard D, Ailhaud G, Negrel R, et al.Promoting effect of glucocorticoids on the differentiation of human adipocyteprecursor cells cultured in a chemically defined medium. J Clin Invest1989;84:1663–70.

[197] Levenberg S, Rouwkema J, Macdonald M, Garfein ES, Kohane DS, Darland DC,et al. Engineering vascularized skeletal muscle tissue. Nat Biotechnol2005;23:879–84.

[198] Tsigkou O, Pomerantseva I, Spencer JA, Redondo PA, Hart AR, O’Doherty E,et al. Regenerative medicine special feature: engineered vascularized bonegrafts. Proc Natl Acad Sci USA 2010;107(8):3311–6.

[199] Niemeyer P, Fechner K, Milz S, Richter W, Suedkamp NP, Mehlhorn AT, et al.Comparison of mesenchymal stem cells from bone marrow and adiposetissue for bone regeneration in a critical size defect of the sheep tibia and theinfluence of platelet-rich plasma. Biomaterials 2010;31(13):3572–9.

[200] Huang NF, Li S. Mesenchymal stem cells for vascular regeneration. Regen Med2008;3:877–92.

[201] Ferrara N. Vascular endothelial growth factor: basic science and clinicalprogress. Endocr Rev 2004;25:581–611.

[202] Bai K, Huang Y, Jia X, Fan Y, Wang W. Endothelium oriented differentiation ofbone marrow mesenchymal stem cells under chemical and mechanicalstimulations. J Biomech 2010;43(6):1176–81.

[203] Dvorak HF, Brown LF, Detmar M, Dvorak AM. Vascular permeability factor/vascular endothelial growth factor, microvascular hyperpermeability, andangiogenesis. Am J Pathol 1995;146:1029–39.

[204] Robinson CJ, Stringer SE. The splice variants of vascular endothelial growthfactor (VEGF) and their receptors. J Cell Sci 2001;114:853–65.

[205] Beckermann BM, Kallifatidis G, Groth A, Frommhold D, Apel A, Mattern J,et al. VEGF expression by mesenchymal stem cells contributes toangiogenesis in pancreatic carcinoma. Br J Cancer 2008;99:622–31.

[206] Nagy JA, Benjamin L, Zeng H, Dvorak AM, Dvorak HF. Vascular permeability,vascular hyperpermeability and angiogenesis. Angiogenesis 2008;11:109–19.

[207] Ball SG, Shuttleworth CA, Kielty CM. Mesenchymal stem cells andneovascularization: role of platelet-derived growth factor receptors. J CellMol Med 2007;11:1012–30.

[208] Streeten EA, Brandi ML. Biology of bone endothelial cells. Bone Miner1990;10:85–94.

[209] Gerber HP, Vu TH, Ryan AM, Kowalski J, Werb Z, Ferrara N. VEGF coupleshypertrophic cartilage remodeling, ossification and angiogenesis duringendochondral bone formation. Nat Med 1999;5:623–8.

[210] Engsig MT, Chen QJ, Vu TH, Pedersen AC, Therkidsen B, Lund LR, et al. Matrixmetalloproteinase 9 and vascular endothelial growth factor are essential forosteoclast recruitment into developing long bones. J Cell Biol2000;151:879–89.

[211] Nakagawa M, Kaneda T, Arakawa T, Morita S, Sato T, Yomada T, et al. Vascularendothelial growth factor (VEGF) directly enhances osteoclastic boneresorption and survival of mature osteoclasts. FEBS Lett 2000;473:161–4.

[212] Connolly DT, Heuvelman DM, Nelson R, Olander JV, Eppley BL, Delfino JJ, et al.Tumor vascular permeability factor stimulates endothelial cell growth andangiogenesis. J Clin Invest 1989;84:1470–8.

[213] Schönmeyr BH, Soares M, Avraham T, Clavin NW, Gewalli F, Mehrara BJ. VEGFinhibits BMP2 expression in rat mesenchymal stem cells. Tissue Eng Part A2010;16(2):653–62.

[214] Terranova VP, DiFlorio R, Lyall RM, Hic S, Friesel R, Maciag T. Humanendothelial cells are chemotactic to endothelial cell growth factor andheparin. J Cell Biol 1985;101:2330–4.

[215] Millauer B, Wizigmann-Voos S, Schnürch H, Martinez R, Møller NP, Risau W,et al. High affinity VEGF binding and developmental expression suggest Flk-1as a major regulator of vasculogenesis and angiogenesis. Cell1993;72:835–46.

[216] Perets A, Baruch Y, Weisbuch F, Shoshany G, Neufeld G, Cohen S. Enhancingthe vascularization of three-dimensional porous alginate scaffolds by

incorporating controlled release basic fibroblast growth factormicrospheres. J Biomed Mater Res A 2003;65:489–97.

[217] Miller DL, Ortega S, Bashayan O, Basch R, Basilico C. Compensation byfibroblast growth factor 1 (FGF1) does not account for the mild phenotypicdefects observed in FGF2 null mice. Mol Cell Biol 2000;20:2260–8.

[218] Li X, Eriksson U. Novel PDGF family members: PDGF-C and PDGF-D. CytokineGrowth Factor Rev 2003;14:91–8.

[219] Solheim E. Growth factors in bone. Int Orthop 1998;22:410–6.[220] Papetti M, Herman IM. Mechanisms of normal and tumor-derived

angiogenesis. Am J Physiol Cell Physiol 2002;282:C947–70.[221] Bar RS, Boes M, Booth BA, Dake BL, Henley S, Hart MN. The effects of platelet-

derived growth factor in cultured microvessel endothelial cells.Endocrinology 1989;124:1841–8.

[222] Battegay EJ, Rupp J, Iruela-Arispe L, Sage EH, Pech M. PDGF-BB modulatesendothelial proliferation and angiogenesis in vitro via PDGF beta-receptors. JCell Biol 1994;125:917–28.

[223] Nicosia RF, Nicosia SV, Smith M. Vascular endothelial growth factor, platelet-derived growth factor, and insulin-like growth factor-1 promote rat aorticangiogenesis in vitro. Am J Pathol 1994;145:1023–9.

[224] Owens GK, Kumar MS, Wamhoff BR. Molecular regulation of vascular smoothmuscle cell differentiation in development and disease. Physiol Rev2004;84:767–801.

[225] McKee JA, Banik SSR, Boyer MJ, Hamad NM, Lawson JH, Niklason LE, et al.Human arteries engineered in vitro. EMBO Rep 2003;4:633–8.

[226] Poh M, Boyer M, Solan A, Dahl SLM, Pedrotty D, Banik SSR, et al. Blood vesselsengineered from human cells. Lancet 2005;365:2122–4.

[227] Harris LJ, Abdollahi H, Zhang P, McIlhenny S, Tulenko TN, DiMuzio PJ.Differentiation of adult stem cells into smooth muscle for vascular tissueengineering. J Surg Res 2009 [Epub ahead of print].

[228] Chan-Park MB, Shen JY, Cao Y, Xiong Y, Liu Y, Rayatpisheh S, et al. Biomimeticcontrol of vascular smooth muscle cell morphology and phenotype forfunctional tissue-engineered small-diameter blood vessels. J Biomed MaterRes A 2009;88:1104–21.

[229] Moosmang S, Lenhardt P, Haider N, Hofmann F, Wegener JW. Mouse modelsto study L-type calcium channel function. Pharmacol Ther 2005;106:347–55.

[230] Ross JJ, Hong Z, Willenbring B, Zeng L, Isenberg B, Lee EH, et al. Cytokine-induced differentiation of multipotent adult progenitor cells into functionalsmooth muscle cells. J Clin Invest 2006;116:3139–49.

[231] Wang C, Cen L, Yin S, Liu Q, Liu W, Cao Y, et al. A small diameter elastic bloodvessel wall prepared under pulsatile conditions from polyglycolic acid meshand smooth muscle cells differentiated from adipose-derived stem cells.Biomaterials 2010;31:621–30.

[232] Solway J, Seltzer J, Samaha FF, Kim S, Alger LE, Niu Q, et al. Structure andexpression of a smooth muscle cell-specific gene, SM22 alpha. J Biol Chem1995;270:13460–9.

[233] van der Loop FT, Schaart G, Timmer ED, Ramaekers FC, van Eys GJ.Smoothelin, a novel cytoskeletal protein specific for smooth muscle cells. JCell Biol 1996;134:401–11.

[234] Gong Z, Niklason LE. Small-diameter human vessel wall engineered frombone marrow-derived mesenchymal stem cells (hMSCs). FASEB J2008;22:1635–48.

[235] Sinha S, Hoofnagle MH, Kingston PA, McCanna ME, Owens GK. Transforminggrowth factor-beta1 signaling contributes to development of smooth musclecells from embryonic stem cells. Am J Physiol Cell Physiol2004;287:C1560–8.

[236] Hirschi KK, Rohovsky SA, D’Amore PA. PDGF, TGF-beta, and heterotypic cell–cell interactions mediate endothelial cell-induced recruitment of 10T1/2 cellsand their differentiation to a smooth muscle fate. J Cell Biol1998;141:805–14.

[237] Deaton RA, Su C, Valencia TG, Grant SR. Transforming growth factor-beta1-induced expression of smooth muscle marker genes involves activation ofPKN and p38 MAPK. J Biol Chem 2005;280:31172–81.

[238] Madsen CS, Hershey JC, Hautmann MB, White SL, Owens GK. Expression ofthe smooth muscle myosin heavy chain gene is regulated by a negative-acting GC-rich element located between two positive-acting serum responsefactor-binding elements. J Biol Chem 1997;272:6332–40.

[239] Li L, Liu Z, Mercer B, Overbeek P, Olson EN. Evidence for serum responsefactor-mediated regulatory networks governing SM22alpha transcription insmooth, skeletal, and cardiac muscle cells. Dev Biol 1997;187:311–21.

[240] Herring BP, Smith AF. Telokin expression in A10 smooth muscle cells requiresserum response factor. Am J Physiol 1997;272:C1394–404.

[241] Miano JM, Carlson MJ, Spencer JA, Misra RP. Serum response factor-dependent regulation of the smooth muscle calponin gene. J Biol Chem2000;275:9814–22.

[242] Böttinger EP, Letterio JJ, Roberts AB. Biology of TGF-beta in knockout andtransgenic mouse models. Kidney Int 1997;51:1355–60.

[243] Sanford LP, Ormsby I, Gittenberger-de Groot AC, Sariola H, Friedman R, BoivinGP, et al. TGFbeta2 knockout mice have multiple developmental defects thatare non-overlapping with other TGFbeta knockout phenotypes. Development1997;124:2659–70.

[244] Hungerford JE, Little CD. Developmental biology of the vascular smoothmuscle cell: building a multilayered vessel wall. J Vasc Res 1999;36:2–27.

[245] Hellström M, Kalén M, Lindahl P, Abramsson A, Betsholtz C. Role of PDGF-Band PDGFR-beta in recruitment of vascular smooth muscle cells and pericytesduring embryonic blood vessel formation in the mouse. Development1999;126:3047–55.

Page 15: Culture Media for the Differentiation of Mesenchymal Stromal Cells

C. Vater et al. / Acta Biomaterialia 7 (2011) 463–477 477

[246] Dandré F, Owens GK. Platelet-derived growth factor-BB and Ets-1transcription factor negatively regulate transcription of multiple smoothmuscle cell differentiation marker genes. Am J Physiol Heart Circ Physiol2004;286:H2042–51.

[247] Kim YM, Jeon ES, Kim MR, Jho SK, Ryu SW, Kim JH. Angiotensin II-induceddifferentiation of adipose tissue-derived mesenchymal stem cells to smoothmuscle-like cells. Int J Biochem Cell Biol 2008;40:2482–91.

[248] Jeon ES, Moon HJ, Lee MJ, Song HY, Kim YM, Bae YC, et al.Sphingosylphosphorylcholine induces differentiation of humanmesenchymal stem cells into smooth-muscle-like cells through a TGF-beta-dependent mechanism. J Cell Sci 2006;119:4994–5005.

[249] Narita Y, Yamawaki A, Kagami H, Ueda M, Ueda Y. Effects of transforminggrowth factor-beta 1 and ascorbic acid on differentiation of human bone-marrow-derived mesenchymal stem cells into smooth muscle cell lineage.Cell Tissue Res 2008;333:449–59.

[250] Galmiche MC, Koteliansky VE, Brière J, Hervé P, Charbord P. Stromal cellsfrom human long-term marrow cultures are mesenchymal cells thatdifferentiate following a vascular smooth muscle differentiation pathway.Blood 1993;82:66–76.

[251] Chang Y, Hwang S, Tseng C, Cheng F, Huang S, Hsu L, et al. Isolation ofmesenchymal stem cells with neurogenic potential from the mesoderm ofthe amniotic membrane. Cells Tissues Organs 2010;192:93–105.

[252] Sellheyer K, Krahl D. Cutaneous mesenchymal stem cells: status of currentknowledge, implications for dermatopathology. J Cutan Pathol2010;37:624–34.

[253] Pierdomenico L, Bonsi L, Calvitti M, Rondelli D, Arpinati M, Chirumbolo G,et al. Multipotent mesenchymal stem cells with immunosuppressive activitycan be easily isolated from dental pulp. Transplantation 2005;80:836–42.

[254] Ringe J, Leinhase I, Stich S, Loch A, Neumann K, Haisch A, et al. Humanmastoid periosteum-derived stem cells: promising candidates for skeletaltissue engineering. J Tissue Eng Regen Med 2008;2:136–46.

[255] Sabatini F, Petecchia L, Tavian M, Jodon de Villeroché V, Rossi GA, Brouty-Boyé D. Human bronchial fibroblasts exhibit a mesenchymal stem cellphenotype and multilineage differentiating potentialities. Lab Invest2005;85:962–71.

[256] Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues.Science 1997;276:71–4.

[257] Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells in humanumbilical cord blood. Br J Haematol 2000;109:235–42.

[258] Bieback K, Kern S, Klüter H, Eichler H. Critical parameters for the isolation ofmesenchymal stem cells from umbilical cord blood. Stem Cells2004;22:625–34.

[259] Caplan AI. The mesengenic process. Clin Plast Surg 1994;21:429–35.[260] Prins H, Rozemuller H, Vonk-Griffioen S, Verweij VGM, Dhert WJA, Slaper-

Cortenbach ICM, et al. Bone-forming capacity of mesenchymal stromal cells

when cultured in the presence of human platelet lysate as substitute for fetalbovine serum. Tissue Eng Part A 2009;15:3741–51.

[261] Shahdadfar A, Frønsdal K, Haug T, Reinholt FP, Brinchmann JE. In vitroexpansion of human mesenchymal stem cells: choice of serum is adeterminant of cell proliferation, differentiation, gene expression, andtranscriptome stability. Stem Cells 2005;23:1357–66.

[262] Friedman MS, Long MW, Hankenson KD. Osteogenic differentiation of humanmesenchymal stem cells is regulated by bone morphogenetic protein-6. J CellBiochem 2006;98:538–54.

[263] Stiehler M, Bünger C, Baatrup A, Lind M, Kassem M, Mygind T. Effect ofdynamic 3-D culture on proliferation, distribution, and osteogenicdifferentiation of human mesenchymal stem cells. J Biomed Mater Res A2009;89:96–107.

[264] Vogel JP, Szalay K, Geiger F, Kramer M, Richter W, Kasten P. Platelet-richplasma improves expansion of human mesenchymal stem cells and retainsdifferentiation capacity and in vivo bone formation in calcium phosphateceramics. Platelets 2006;17:462–9.

[265] Shen B, Wei A, Whittaker S, Williams LA, Tao H, Ma DDF, et al. The role ofBMP-7 in chondrogenic and osteogenic differentiation of human bonemarrow multipotent mesenchymal stromal cells in vitro. J Cell Biochem2010;109(2):406–16.

[266] Diekman BO, Rowland CR, Caplan AI, Lennon D, Guilak F. Chondrogenesis ofadult stem cells from adipose tissue and bone marrow: induction by growthfactors and cartilage derived matrix. Tissue Eng Part A 2010;16(2):523–33.

[267] van Harmelen V, Röhrig K, Hauner H. Comparison of proliferation anddifferentiation capacity of human adipocyte precursor cells from the omentaland subcutaneous adipose tissue depot of obese subjects. Metabolism2004;53:632–7.

[268] Bunnell BA, Flaat M, Gagliardi C, Patel B, Ripoll C. Adipose-derived stem cells:isolation, expansion and differentiation. Methods 2008;45:115–20.

[269] Vashi AV, Keramidaris E, Abberton KM, Morrison WA, Wilson JL, O’Connor AJ,et al. Adipose differentiation of bone marrow-derived mesenchymal stemcells using Pluronic F-127 hydrogel in vitro. Biomaterials 2008;29:573–9.

[270] Jiang Y, Jahagirdar BN, Reinhardt RL, Schwartz RE, Keene CD, Ortiz-GonzalezXR, et al. Pluripotency of mesenchymal stem cells derived from adultmarrow. Nature 2002;418:41–9.

[271] Kanematsu A, Yamamoto S, Iwai-Kanai E, Kanatani I, Imamura M, Adam RM,et al. Induction of smooth muscle cell-like phenotype in marrow-derivedcells among regenerating urinary bladder smooth muscle cells. Am J Pathol2005;166:565–73.

[272] Shukla D, Box GN, Edwards RA, Tyson DR. Bone marrow stem cells forurologic tissue engineering. World J Urol 2008;26:341–9.

[273] Kurpinski K, Chu J, Wang D, Li S. Proteomic profiling of mesenchymal stemcell responses to mechanical strain and TGF-beta1. Cell Mol Bioeng2009;2:606–14.