schwann-like cell differentiation of rat adipose-derived stem cells by indirect co-culture with...

11
Schwann-like cell differentiation of rat adipose-derived stem cells by indirect co-culture with Schwann cells in vitro Y. Wei*, K. Gong*, Z. Zheng*, L. Liu*, A. Wang†, L. Zhang‡, Q. Ao§, Y. Gong* and X. Zhang* *School of Life Sciences, State Key Laboratory of Biomembrane and Membrane Biotechnology, Tsinghua University, Beijing, China, Department of Bioengineering, University of California, Berkeley, CA, USA, Chinese PLA General Hospital, Beijing, China, and §Department of Neurosurgery, Yuquan Hospital, Tsinghua University, Beijing, China Received 15 January 2010; revision accepted 11 June 2010 Abstract Objectives: Schwann cell (SC) transplantation is a promising therapy for peripheral nerve transaction, however, clinical use of SCs is limited due to their very limited availability. Adipose-derived stem cells (ADSCs) have been identified as an alternative source of adult stem cells in recent years. The aim of this study was to evaluate the feasibility of using ADSCs as a source of stem cells for differentiation into Schwann-like cells by an indirect co-culture approach, in vitro. Materials and methods: Multilineage differentiation potential of the obtained ADSCs was assayed by test- ing their ability to differentiate into osteoblasts and adipocytes. The ADSCs were co-cultured with SCs to be induced into Schwann-like cells through prox- imity, using a Millicell system. Expression of typical SC markers S-100, GFAP and P75NTR of the treated ADSCs was determined by immunocytochemical staining, western blotting and RT-PCR. Myelination capacity of the differentiated ADSCs (dADSCs) was evaluated in dADSC dorsal root ganglia neuron (DRGN) co-cultures. Results: The treated ADSCs adopted a spindle shaped-like morphology after co-cultured with SCs for 6 days. All results of immunocytochemical stain- ing, western blotting and RT-PCR showed that the treated cells expressed S-100, GFAP and P75NTR, indications of differentiation. dADSCs could form Schwann-like cell myelin in co-culture with DRGNs. Undifferentiated ADSCs (uADSCs) did not form myelin compared to DRGNs cultured alone, but could produce neurite extension. Conclusions: These results demonstrate that this indirect co-culture microenvironment could induce ADSCs to differentiate into Schwann-like cells in vitro, which may be beneficial for treatment of peripheral nerve injuries in the near future. Introduction Schwann cells (SCs) play a critical role in regeneration of peripheral nerves and are thus, the most promising candi- date cell type as healing treatment for transected periph- eral nerves (1). In animal models, SCs transplanted into nerve bridges in injured sciatic nerves have been shown to promote re-growth of injured axons and of functional recovery (2). However, it is difficult to obtain sufficiently large numbers of functional SCs for in vitro work within a short span of time, which greatly limits their clinical application. In the past few decades, bone marrow stromal cells (BMSCs) have been considered as an alternative cell source for tissue regeneration after peripheral nerve injury as they are capable of selectively differentiating into SCs and enhancing axonal regeneration (3). However, BMSC procurement procedures are painful for the donor and frequently require general or spinal anaesthesia and yet may yield low numbers of BMSCs upon harvest (4). For these reasons, alternative stem cell types need to be investigated. The ideal transplantable source cell should be easily accessible, proliferate rapidly in vitro, possess multipoten- tal differentiation properties and successfully integrate into host tissue, with non- or hypo-immunogenicity (5). Cells isolated from adipose tissue, termed adipose-derived stem cells (ADSCs), can satisfy the above requirements. Both ADSCs and BMSCs are originally derived from embryonic mesoderm, they have similar phenotype and Correspondence: X. Zhang, School of Life Sciences, State Key Laboratory of Biomembrane and Membrane Biotechnology, Tsinghua University, Beijing 100084, China. Tel.: 0086-10-6278 3261; Fax: 0086-10-6279 4214; E-mail: [email protected] 606 ȑ 2010 Blackwell Publishing Ltd. Cell Prolif., 2010, 43, 606–616 doi: 10.1111/j.1365-2184.2010.00710.x

Upload: y-wei

Post on 15-Jul-2016

217 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: Schwann-like cell differentiation of rat adipose-derived stem cells by indirect co-culture with Schwann cells in vitro

Schwann-like cell differentiation of rat adipose-derived stem cells by indirect

co-culture with Schwann cells in vitro

Y. Wei*, K. Gong*, Z. Zheng*, L. Liu*, A. Wang†, L. Zhang‡, Q. Ao§, Y. Gong* and X. Zhang*

*School of Life Sciences, State Key Laboratory of Biomembrane and Membrane Biotechnology, Tsinghua University, Beijing, China, †Department ofBioengineering, University of California, Berkeley, CA, USA, ‡Chinese PLA General Hospital, Beijing, China, and §Department of Neurosurgery,Yuquan Hospital, Tsinghua University, Beijing, China

Received 15 January 2010; revision accepted 11 June 2010

AbstractObjectives: Schwann cell (SC) transplantation is apromising therapy for peripheral nerve transaction,however, clinical use of SCs is limited due to theirvery limited availability. Adipose-derived stem cells(ADSCs) have been identified as an alternativesource of adult stem cells in recent years. The aim ofthis study was to evaluate the feasibility of usingADSCs as a source of stem cells for differentiationinto Schwann-like cells by an indirect co-cultureapproach, in vitro.Materials and methods: Multilineage differentiationpotential of the obtained ADSCs was assayed by test-ing their ability to differentiate into osteoblasts andadipocytes. The ADSCs were co-cultured with SCsto be induced into Schwann-like cells through prox-imity, using a Millicell system. Expression of typicalSC markers S-100, GFAP and P75NTR of the treatedADSCs was determined by immunocytochemicalstaining, western blotting and RT-PCR. Myelinationcapacity of the differentiated ADSCs (dADSCs) wasevaluated in dADSC ⁄dorsal root ganglia neuron(DRGN) co-cultures.Results: The treated ADSCs adopted a spindleshaped-like morphology after co-cultured with SCsfor 6 days. All results of immunocytochemical stain-ing, western blotting and RT-PCR showed that thetreated cells expressed S-100, GFAP and P75NTR,indications of differentiation. dADSCs could formSchwann-like cell myelin in co-culture with DRGNs.Undifferentiated ADSCs (uADSCs) did not form

myelin compared to DRGNs cultured alone, butcould produce neurite extension.Conclusions: These results demonstrate that thisindirect co-culture microenvironment could induceADSCs to differentiate into Schwann-like cellsin vitro, which may be beneficial for treatment ofperipheral nerve injuries in the near future.

Introduction

Schwann cells (SCs) play a critical role in regeneration ofperipheral nerves and are thus, the most promising candi-date cell type as healing treatment for transected periph-eral nerves (1). In animal models, SCs transplanted intonerve bridges in injured sciatic nerves have been shown topromote re-growth of injured axons and of functionalrecovery (2). However, it is difficult to obtain sufficientlylarge numbers of functional SCs for in vitro work withina short span of time, which greatly limits their clinicalapplication.

In the past few decades, bone marrow stromal cells(BMSCs) have been considered as an alternative cell sourcefor tissue regeneration after peripheral nerve injury asthey are capable of selectively differentiating into SCsand enhancing axonal regeneration (3). However, BMSCprocurement procedures are painful for the donor andfrequently require general or spinal anaesthesia and yetmayyield low numbers of BMSCs upon harvest (4). For thesereasons, alternative stem cell types need to be investigated.

The ideal transplantable source cell should be easilyaccessible, proliferate rapidly in vitro, possess multipoten-tal differentiation properties and successfully integrateinto host tissue, with non- or hypo-immunogenicity (5).Cells isolated from adipose tissue, termed adipose-derivedstem cells (ADSCs), can satisfy the above requirements.Both ADSCs and BMSCs are originally derived fromembryonic mesoderm, they have similar phenotype and

Correspondence: X. Zhang, School of Life Sciences, State Key Laboratoryof Biomembrane and Membrane Biotechnology, Tsinghua University,Beijing 100084, China. Tel.: 0086-10-6278 3261; Fax: 0086-10-62794214; E-mail: [email protected]

606 � 2010 Blackwell Publishing Ltd.

Cell Prolif., 2010, 43, 606–616 doi: 10.1111/j.1365-2184.2010.00710.x

Page 2: Schwann-like cell differentiation of rat adipose-derived stem cells by indirect co-culture with Schwann cells in vitro

gene expression profiles (6,7); both can differentiate intoseveral mesenchymal tissue lineages, including adipocytes(8), osteoblasts (9), myocytes (10), chondrocytes (11),endothelial cells (12) and cardiomyocytes (13). They canalso be induced into neural lineages in vitro (14–16). Sub-cutaneous adipose tissue is abundant and possesses moremesenchymal stem cells (MSCs; 2 in 100 cells) than doesthe bone marrow (1 in 25 000 or 100 000 cells) (17–20).ADSCs can be easily isolated by conventional liposuctionprocedures, which overcome tissue morbidity comparedto bone marrow aspiration, and proliferate rapidly in cul-ture. Thus, ADSCs may be an ideal alternative cell sourcefor SC transplantation in regeneration of peripheral nerve.

Previous studies have shown that ADSCs can beinduced into SC-like cells using a protocol similar to themethod used to induce BMSCs to differentiate into SC-like cells (21,22). Reports have documented that SC-likecells from ADSCs are spindle-like in shape and expressSC markers such as S-100. In our study, we have aimed toinvestigate whether ADSCs could be induced into Schw-ann-like cells by indirect co-culture with SCs in vitro, andwhether the differentiated ADSCs (dADSCs) wouldexhibit Schwann-like cell behaviour when co-culturedwith dorsal root ganglia neurons (DRGNs) in vitro, whichwould provide a foundation for transplantation of dADS-Cs in the vicinity of transected peripheral nerves.

Materials and methods

Materials

Trypsin and Dulbecco’s modified Eagle’s medium ⁄F-12Ham nutrient mixture (DMEM ⁄F-12) were purchasedfrom Invitrogen, Carlsbad, CA, USA. Collagenase type awas purchased from Merck. Recombinant human beta-NGF was from Peprotech (Rocky Hill, Connecticut,USA). Cytosine-B-arabinoside hydrochloride (Ara-C),3-isobutyl-1-methylxanthine, ascorbate, dexamethasone,b-glycerophosphate, insulin, penicillin and streptomycinwere purchased from Sigma, St. Louis, MO, USA. Humanheregulin1-b1 extracellular domain (HRG1-b1 ECD) wasfrom R&D (Minneapolis, USA). Foetal bovine serum(FBS) was obtained from Hyclone, Logan, UT, USA. 12-well Millicell cell culture dishes with inserted hanginggeometry (1.0 lm PET transparent) were purchased fromMillipore (Boston, MA, USA). All culture plates werefrom Costar (IL, USA). All other reagents were localproducts of analytical grade.

Primary antibody employed for immunocytochemis-try, rabbit anti-S-100 protein monoclonal antibody, wasfrom Sigma. Mouse anti-tubulin, beta isoform monoclonalantibody, rabbit anti-P75 neurotrophin receptor polyclonalantibody and rabbit anti-glial fibrillary acidic protein

(GFAP) polyclonal antibody were from Millipore.Secondary antibodies including FITC goat anti-rabbit IgG(H+L), FITC goat anti-mouse IgG (H+L), TRITC goatanti-rabbit IgG (H+L) and goat anti-rabbit IgG ⁄HRP werepurchased from Invitrogen.

Neonatal Sprague–Dawley rats (1–3 days old) andadult Sprague–Dawley rats (male, 6–8 weeks old) wereobtained from Beijing Haidian Experimental Animal Cen-ter. All animal experiments were carried out in accordancewith the US National Institute of Health Guide for theCare and Use of Laboratory Animals (NIH PublicationsNo. 80-23) revised 1996 and approved by the BeijingAdministration Committee for Experimental Animals.

Isolation and culture of ADSCs

Adult Sprague–Dawley rats were deeply anaesthetizedusing chloral hydrate, and adipose tissue was obtainedfrom testicular fat pads. Isolation of ADSCs was accom-plished using a modification of a previously publishedmethod (22). Adipose tissue samples were washed in coldDMEM ⁄F-12 medium (containing 1% penicillin ⁄ strepto-mycin) and were mechanically dissociated carefully afterdigestion in collagenase type II for 40 min at 37 �C. Sus-pensions were filtered through 80-lm metal mesh andcentrifuged to separate floating adipocytes from the stro-mal vascular fraction. Supernatant was discarded and cellsin the stromal vascular fraction were resuspended inDMEM ⁄F-12 medium (containing 1% penicillin ⁄ strepto-mycin and 10% FBS) and seeded on culture plates.Twenty-four hours later, culture medium was changed toeliminate non-adherent cells. Cells used in our experi-ments were passaged 3–5 times.

Isolation and culture of SC

Schwann cells were enriched from sciatic nerves of neona-tal Sprague–Dawley rats as described in our previousstudy (23). The protocol is based on different rates of cellproliferation and attachment properties between culturedfibroblasts and SCs in vitro. In brief, cells were first trea-ted with Ara-C for 24 h. Then medium was replaced withgrowth medium containing 20 ng ⁄ml human heregulin1-b1 extracellular domain (HRG1-b1 ECD). After 48 h fur-ther incubation, cells were treated with 0.05% trypsin,which resulted in detachment of SCs from the dishes butnot fibroblasts. In each category, suspended cells were col-lected into conical tubes and centrifuged. After removal ofsupernatant, pellets were resuspended in DMEM ⁄F-12medium (containing 1% penicillin ⁄ streptomycin, 20 ng ⁄ml HRG1-b1 ECD and 10% FBS) and plated on cultureplates at density of 5 · 104 cells ⁄ml. Cells were grown toconfluence, and then passaged by splitting them 1:3.

� 2010 Blackwell Publishing Ltd, Cell Proliferation, 43, 606–616.

Schwann-like cell differentiation of rat ADSCs 607

Page 3: Schwann-like cell differentiation of rat adipose-derived stem cells by indirect co-culture with Schwann cells in vitro

Proliferation assay of ADSCs

ADSCs at passages 3, 5 and 10, were seeded1 · 104 cells ⁄ml on 24-well plates to determine prolifera-tion activity of the cells. After growing ADSCs for 1, 3, or5 days, cell proliferation was determined by MTT assay,which detects mitochondrial dehydrogenase activity ofviable cell, s spectrophotometrically. Briefly, cells werecultured for 1, 3, and 5 days, and 100 ll of MTT (3-[4, 5-dimethylthiazol-2-yl]-2, 5-diphenyl tetrasodium bromide,5 mg ⁄ml), was added to each well and incubated at 37 �Cfor 4 h. Then, MTT solution was carefully removed andinsoluble blue formazan crystals were dissolved in 1 mldimethyl sulphoxide. One hundred microlitres of solutionwas aspirated from each well and poured into 96-wellplates. Absorbance of each well was measured by ELISAat 570 nm. In addition, MTT assay was performed ondirectly counted SCs (0.5 · 105, 1 · 105, 2 · 105,4 · 105, 8 · 105, 1.6 · 106, 3.2 · 106), and absorbancevalues were plotted against counted cell numbers to estab-lish a standard calibration curve. Viable cell numbers werethen determined from the standard curve based on theirMTT absorbance values. Cell numbers are represented asmean ± standard deviation (SD).

Adipogenic and osteogenic differentiation of rat ADSCs

Multilineage differentiation potential was assayed by test-ing ability of ADSCs to differentiate into osteoblasts andadipocytes, as described previously (24). Briefly, osteo-blast differentiation was achieved after cells were treatedwith 50 lg ⁄ml ascorbate, 0.1 lM dexamethasone and10 mM b-glycerophosphate for 3 weeks. For adipocytedifferentiation, cultures were treated with 1 lM dexameth-asone, 10 lg ⁄ml insulin and 100 lg ⁄ml 3-isobutyl-1-methylxanthine for 3 weeks. von Kossa and oil red Ostaining methods were employed to identify osteoblastsand adipocytes respectively.

Differentiation of ADSCs by co-culture with SCs

For co-culture experiments, 5 · 103 ADSCs ⁄well wereseeded on 12-well plates with DMEM ⁄F-12 medium

containing 10% FBS overnight and replaced with freshmedium to eliminate non-adherent cells. As shown inFig. 1 (III), SCs were seeded on Millicell and co-culturedwith ADSCs in 12-well plates with replacement of 1 ⁄2volume co-culture medium every 3 days. Millicell plateshad porous polystyrene membranes with pore size of1 lm, allowing free exchange of proteins in the mediumwhile preventing cell migration between the two compart-ments. ADSCs without co-culturing with SCs under iden-tical conditions were negative controls and SCs alonewere positive controls.

Immunofluorescence staining

For immunophenotype characterization of dADSCs, SCmarkers, including S-100, GFAP and P75NTR, were used.Immunostaining was performed according to protocolspublished previously (25). Briefly, cells cultured on cover-slips were fixed in 4% paraformaldehyde (PFA) in phos-phate-buffered saline (PBS) for 10 min, and thenpermeabilized with 0.05% Triton X-100 for 10 min. Non-specific binding sites were blocked using 5% goat serumfor 1 h. Cells were then incubated with primary monoclo-nal antibody (1:200) at room temperature for 1 h. Cellswere washed three times in PBS and incubated with corre-sponding secondary antibodies (1:200) for 2 h at roomtemperature. After second rinsing in PBS, cells were incu-bated in 4¢, 6¢-diamidino-2-phenylindole dihydrochloride(DAPI, 1:1000) to counterstain cell nuclei, for 5 min atroom temperature. Finally, specimens were washed threetimes in PBS, each for 5 min, and were then mounted withantifade solution. Labelled cells were examined by fluo-rescence microscopy. Images were digitally recorded andprocessed using Image-Pro Plus (Media Cybernetics,USA). Cultures of SCs and uADSCs were similarlystained as positive and negative controls accordingly, withantibodies above.

Cultures to be stained for b-tubulin were fixed in 4%PFA for 30 min at room temperature. Cultures were per-meabilized with 0.1% Triton X-100 for 10 min. Cultureswere then treated with ice-cold 50% acetone for 2 min,ice-cold 100% acetone for 2 min and 50% acetonefor 2 min. They were then washed twice in PBS for

Figure 1. Model of ADSCs co-cultured with SCs. (I) ADSCs cultured alone; (II) Schwann cells; (III) ADSCs co-cultured with Schwann cells.A: DMEM ⁄ F-12 medium; B: ADSCs; C: SCs; D: Millicell.

� 2010 Blackwell Publishing Ltd, Cell Proliferation, 43, 606–616.

608 Y. Wei et al.

Page 4: Schwann-like cell differentiation of rat adipose-derived stem cells by indirect co-culture with Schwann cells in vitro

3 min each and incubated with mouse anti-tubulin beta bisoform, monoclonal antibody for 2 h at room tempera-ture. Rinsing three times in PBS was followed by incuba-tion with corresponding secondary antibodies (1:100) for1 h at room temperature. After the second rinsing stepin PBS, cells were incubated in DAPI (1:1000) for5 min at room temperature to counterstain cell nuclei.Finally, specimens were washed three times in PBS,each for 5 min, and then were mounted with antifade solu-tion. Labelled cells were examined by fluorescencemicroscopy.

Western blot analysis

Cells were washed three times in cold PBS, scraped intoice-cold lysis buffer (20 mM Tris–HCl, pH 7.4, 2 mM

EDTA, 25 mM NaF, 1% Triton X-100 plus proteaseinhibitors) and incubated at 4 �C for 30 min. After incu-bation, cell lysates were sonicated for 15 s and then cen-trifuged at 12 520 g at 4 �C for 15 min. Supernatantswere collected and their total protein concentration wasdetermined using BCA Protein Assay Kit (Pierce, Rock-ford, USA). Protein samples were completely denaturedby boiling in bromophenol blue sample buffer for 5 min,separated by SDS–PAGE (10%), and then transferredelectrophoretically on to polyvinylidenedifluoride(PVDF) membranes. Non-specific antibody binding wasblocked with 5% non-fat dry milk in Tris-buffered salinecontaining 0.1% Tween 20 (TBST) for 2 h at room tem-perature. Immunoblotting was performed with specificprimary antibody and secondary antibodies conjugated tohorseradish peroxidase. Protein blots were developed byenhanced chemiluminescence method. b-actin was usedas loading control, and results were analysed as detectedprotein ⁄ b-actin ratio.

RT-PCR analysis

RT-PCR was used to evaluate expression of genes encod-ing S-100, GFAP and P75NTR in ADSCs co-culturedwith SCs. Total RNA was isolated using Trizol reagent(Invitrogen) and genomic DNA was removed usingDNase (Takara, Japan). Two micrograms of RNA wasreverse transcribed at 42 �C for 45 min in 20 ll reaction

mixture using Reverse Transcription System (Promega,Madison, USA). Total RNA concentration was deter-mined spectrophotometrically at 260 nm. Primers ofdetected genes are listed in Table 1. The reaction systemand procession were employed as described previously(26). GAPDH was used to normalize the amount of tem-plate for each sample. ADSCs not co-cultured with SCsunder identical conditions were negative controls and SCsalone were positive controls.

Functional assay in dADSC ⁄DRGN co-cultures

DRGN culture was prepared according to a previous studywith minor modification (27). In brief, dorsal root ganglia(DRG) neurons were removed from neonatal Sprague–Dawley rats, cleaned of connective tissue and chemicallydissociated in trypsin. Trypsin activity was stopped byaddition of DMEM ⁄F-12 medium containing foetalbovine serum. Ganglia were then washed in DMEM ⁄F-12, before mechanical dissociation by gentle triturationusing a pipette. Dissociated neurons were passed though a70 lm mesh to remove non-dissociated tissues and myelindebris. Suspensions were centrifuged, and pellets wereresuspended in NLA negative (neurobasal medium plusB27, supplemented with nerve growth factor at10 ng ⁄ml). Approximately 5000–7000 DRG cells wereplated per well (24-well plate). After seeding and over-night incubation, cultures were flooded and subsequentlytreated with NLA negative medium containing 5¢-fluoro-2¢-deoxyuridine (FUDR) at 20 lg ⁄ml and uridine at10 lg ⁄ml 2 days to kill any remaining non-neuronal cells.After this antimitotic treatment, resulting DRGN cultureswere maintained on NLA) medium for at least 1 week toensure that residual FUDR was removed when SCs ordADSCs were added.

For myelination experiments, co-cultures were pre-pared by addition of 50 000 dADSCs, SCs and uADSCs,to purified DRGN cultures. DRGN cultures and co-cultures were fed every 2–3 days with NLA negativemedium for 2 weeks. For myelination induction, eachculture of DRGN, SC ⁄DRGN, dADSC ⁄DRGN anduADSC ⁄DRGN was switched to NLA+ medium. NLA+

medium was the same as NLA) medium but with additionof 50 lg ⁄ml ascorbic acid. All cultures were then

Table 1. Primers for RT-PCR

Gene Forward primer (5¢–3¢) Reverse primer (5¢–3¢) PCR product (bp)

S-100 GAGAGAGGGTGACAAGCACAA GGCCATAAACTCCTGGAAGTC 169P75NTR AGGGCACATACTCAGACGAA CAAGATGGAGCAATAGAGAG 327GFAP CTTCCCGCAACGCAGAG GAGCCGTGGGCACTAAA 631GAPDH GCCACCCAGAAGACTGTGGAT TGGTCCAGGGTTTCTTACTCC 500

� 2010 Blackwell Publishing Ltd, Cell Proliferation, 43, 606–616.

Schwann-like cell differentiation of rat ADSCs 609

Page 5: Schwann-like cell differentiation of rat adipose-derived stem cells by indirect co-culture with Schwann cells in vitro

maintained for additional 2 weeks, with replacement ofappropriate medium every 2–3 days.

Sudan black staining and cell counting

Cultures were fixed for 10 min in 4% PFA, rinsed threetimes in 0.1 M PO4 and further fixed in 0.1% OsO4 for 1 hat 4 �C. Cultures were then washed four times in 0.1 M

PO4 followed by dehydration in 25%, 50% and 70% etha-nol, each for 5 min; 0.5% Sudan black ⁄70% ethanol solu-tion was filtered before staining for 1 h. Cultures wererinsed in 0.1 M PO4 and mounted on glycerine jelly aftertrimming the dish. Images of myelinated axons were cap-tured using a CCD video camera (CCD colour camera;Hitachi, Japan). All experiments were performed in tripli-cate and repeated at least twice.

Statistical analysis

Data are expressed as mean ± SD. For comparison ofquantitative measures, the values were subjected to statis-tical analysis using Student’s t-test, considered significantat P < 0.05.

Results

Characterization of rat ADSCs and SCs

Rat testicular adipose tissue was enzymatically digestedand then centrifuged to isolate the stromal cell fractionfrom mature adipocytes. After two passages in culture,

cells from the stromal fraction morphologically exhibiteda homogenous population of fibroblast-like cells (Fig. 2a).Cells were then trypsinized and plated for MTT pro-liferation assay (Fig. 3). There was an apparent stationaryphase in population growth of cells during the first 24 h.Rate of cell proliferation then expanded more rapidly.Although overall cell population growth rate of ADSCs atpassages 5 and 10 decreased, they still proliferated veryrapidly. These rat ADSCs could be passaged at least 10times, and did not spontaneously differentiate duringculture expansion (data not shown).

As Fig. 2d shows, SCs were typically spindle-shapedand were either bipolar or occasionally multipolar, oftenaligning in fascicles; purity of SCs was more than 99%.

To determine whether cells isolated from adipose tis-sue exhibited multipotential properties of mesenchymalstem cells, they were treated with agents known to inducedifferentiation in cells originating from the mesoderm.Adipocyte differentiation was confirmed by lipid dropletswith oil red O (Fig. 4a) and osteoblast differentiation wasassessed by production of calcium deposits detected bystaining with the von Kossa reagent (Fig. 4b).

Morphology of dADSCs

At co-culture days 6 and 21, ADSCs were visualizedusing a phase-contrast microscope. As Fig. 2 shows,ADSCs co-cultured with SCs exhibited striking morpho-logical changes compared to controls. ADSCs culturedalone appeared as a monolayer of large, fibroblast-likeflattened cells. When ADSCs were co-cultured with SCs

(a) (b)

(d)(c)

Figure 2. Morphology of ADSCs, dADSCsand SCs by phase-contrast microscopy. (a)ADSCs cultured alone showed flattened fibro-blast-like morphology; (b) ADSCs co-culturedwith Schwann cells for 6 days differentiated to aSchwann cell phenotype; (c) Morphology oftreated cells was exactly the same as SCs, whenco-cultured for 21 days; (d) Images of purifiedSchwann cells (magnification: ·100).

� 2010 Blackwell Publishing Ltd, Cell Proliferation, 43, 606–616.

610 Y. Wei et al.

Page 6: Schwann-like cell differentiation of rat adipose-derived stem cells by indirect co-culture with Schwann cells in vitro

for 21 days, cells showed spindle shaped morphology andwere bi- or tripolar, resembling the appearance of SCs(Fig. 2c). Unfortunately, almost all control ADSCsdetached from the plates and died after 21 days of culturewithout passage.

Expression of SC markers

dADSCs were assessed for expression of SC markers:S-100, GFAP and P75NTR, to determine evidence ofphenotypic progression along an SC lineage. Control SCcultures were immunoreactive for all three makers, S-100,GFAP and P75NTR. The cells demonstrated typical bipo-lar or tripolar morphology, had oval nuclei, and brightlystained for the above three protein markers, as shown inFig. 5a–c respectively. Positive expression of S-100,GFAP and P75NTR was observed in treated groups ondays 6 and 21 (Fig. 5d–i). ADSCs cultured alone werenegative for S-100, GFAP and P75NTR (Fig. 5j–l); how-ever, if ADSCs were cultured up to passage 8–10, therewas occasional, faint expression of S-100, but not ofGFAP or P75NTR (Fig. 6).

To confirm that results obtained by immunocytochem-ical staining were not due to artefacts of cell shrinkage,western blotting and RT-PCR were performed. Lysates ofthe treated ADSCs but not ADSCs cultured alone showedS-100, GFAP and P75NTR-immunoreactive bands corre-sponding to a molecular weight of 12, 52 and 75 kDarespectively. This was present in SC lysates as well(Fig. 7). Results of RT-PCR of the treated ADSCs mRNAprobed with S-100, GFAP and P75NTR primers also sup-ported the results found on immunocytochemical staining.As shown in Fig. 8, S-100, GFAP and P75NTR geneexpressions were detected in dADSCs and SCs, but not inuADSCs cultures. All the above results supported theresults found on immunocytochemical staining.

To investigate whether the cells could maintain theirdifferentiated state after removing SCs from the co-culturesystem, dADSCs were transferred to normal medium.Absence of SCs induced morphological changes indADSCs during the first 2 weeks, which though main-tained expression of SC markers, thus suggesting an indi-cation of long-lasting differentiation (data not shown).After that, cells gradually regained basal morphology andphenotype. However, if ADSCs was kept in co-culturewith SCs, their differentiated state could last for 21 days(Fig. 5g–i).

Functional assay of dADSC ⁄DRGN co-cultures

To evaluate ability of dADSCs to produce myelin underconditions that enabled SCs to produce myelin,dADSC ⁄DRGN, SC ⁄DRGN and uADSC ⁄DRGN co-cul-tures were prepared as described previously (27). Afteraddition of dADSCs, SCs and uADSCs, cultures weremaintained for a 2-week period in NLA) medium to allowfor cell proliferation. Cultures were then switched toNLA+ medium and incubated for an additional 2-weekperiod to allow myelination to occur. In bothdADSC ⁄DRGN and SC ⁄DRGN co-cultures, extensivemyelin was formed during the final weeks (Fig. 9a,b). Nomyelin was formed in uADSC ⁄DRGN co-cultures overthe same period (Fig. 9c); identical results were obtained

Figure 3. Cell proliferation of ADSCs isolated from adipose tissue.MTT assay was used to determine growth rate of cell populationsobtained from adipose tissue at passage 3( ), 5( ) and 10( ). Valuesrepresent mean ± SEM, corresponding to five replicates.

(a) (b)

Figure 4. ADSCs exhibit properties of stemcells. Cultures were treated with agents to inducedifferentiation to adipogenic and osteogeniclineages. Cells were then stained with oil redO (a) and von Kossa reagent (b) respectively(magnification: ·100).

� 2010 Blackwell Publishing Ltd, Cell Proliferation, 43, 606–616.

Schwann-like cell differentiation of rat ADSCs 611

Page 7: Schwann-like cell differentiation of rat adipose-derived stem cells by indirect co-culture with Schwann cells in vitro

(a) (b) (c)

(d) (e) (f)

(g) (h) (i)

(j) (k) (l)

Figure 5. Immunofluorescence staining in Schwann cells, dADSCs and uADSCs. (a, d, g, j) S-100; (b, e, h, k) GFAP and (c, f, i, l) P75NTR. Nucleilabelled blue with DAPI. Morphology of dADSCs: purity of isolated Schwann cells was more than 99% (magnification: ·100).

(a) (b) (c)

Figure 6. S-100 immunostaining in ADSCs.(a) DAPI; (b) S-100 and (c) merged. Therewas occasional, faint expression of S-100 whenADSCs were cultured up to passage 8–10(magnification: ·200).

� 2010 Blackwell Publishing Ltd, Cell Proliferation, 43, 606–616.

612 Y. Wei et al.

Page 8: Schwann-like cell differentiation of rat adipose-derived stem cells by indirect co-culture with Schwann cells in vitro

in repeat experiments. Myelin was not formed in controlDRGN cultures to which no exogenous cells were added(Fig. 9d).

One week after addition of dADSCs, SC and uADSC,cultures were further studied by immunocytochemicalstaining to better characterize axon–glial relationships inSC ⁄DRGN, dADSC ⁄DRGN and uADSC ⁄DRGN co-cul-tures. At termination of cultures for analysis, dADSCswere demonstrated to have greatly enhanced neurite out-growth (Fig. 10b), which was similar to the level achievedby SCs (Fig. 10a). uADSCs produced neurite extensionssuperior to those seen with DRGNs cultured alone(Fig. 10c,d), but inferior to those seen with SCs anddADSCs.

Discussion

After transaction of peripheral nerves, SCs play a crucialpart in functional recovery of injured peripheral nerves by

Figure 7. Western blotting analysis of S-100,GFAP and P75NTR in Schwann cells, dADSCsand uADSCs. SCs, dADSCs and uADSCslysates were blotted for S-100, GFAP andP75NTR proteins. b-actin was used as a loadingcontrol.

Figure 8. Detection of S-100, P75NTR and GFAP expression inSchwann cells, dADSCs and uADSCs. Lane 1: DNA markers; Lane 2:Schwann cells; Lane 3: ADSCs cultured alone; Lane 4: ADSCs co-cul-tured with Schwann cells for 6 days; Lane 5: ADSCs co-cultured withSchwann cells for 21 days.

(a) (b)

(c) (d)

Figure 9. dADSCs could form myelin underconditions that enabled SCs to produce myelin.(a) SC ⁄DRGN co-cultures; (b) dADSCs ⁄DRGNco-cultures; (c) uADSCs ⁄DRGN co-cultures; (d)DRGN cultures. Myelin sheaths (arrows in a andb) are found in both SC ⁄DRGN and dADSCs ⁄DRGN co-cultures (magnification: ·100).

� 2010 Blackwell Publishing Ltd, Cell Proliferation, 43, 606–616.

Schwann-like cell differentiation of rat ADSCs 613

Page 9: Schwann-like cell differentiation of rat adipose-derived stem cells by indirect co-culture with Schwann cells in vitro

forming Bungner bands, to help regrowing axons elon-gate, secrete surface cell adhesion molecules, cell matrixprotein and neurotrophic factors, and remyelinating regen-erative axons (28,29). Therefore, reduced time periodsbetween injury and SC transplantation would facilitate tis-sue integration in clinical processes. This would enhanceprobability of functional recovery after reconstruction sur-gery, which depends definitely on a crucial time windowto obtain enough population of SCs, naıve or differenti-ated, from stem cells.

Schwann-like cell differentiation of ADSCs, inducedeither with chemical factors or by co-culture with SCs –by cell–cell contact or SC-conditioned medium, has beenobtained by several groups (21,30). In our study, weaimed to evaluate the potential of ADSCs to differentiateinto Schwann-like cells by an indirect co-culture approachthrough Millicell porous membranes. Millicell membraneswere placed in such a way that allowed molecules secretedby SCs to permeate through the pores and reach the ADS-Cs beneath. There was no direct contact between SCs andADSCs; ADSCs and SCs could communicate with eachother only via secreted proteins in the medium during theco-culture process. Our purpose was to assess whethersuch communication could induce Schwann-like celldifferentiation of ADSCs in vitro. Results showed that

morphological changes were induced in the treated ADS-Cs co-cultured with SCs, which subsequently successfullyexpressed SC markers, S-100, GFAP and P75NTR, bothat protein and mRNA levels, thus suggesting that somemolecules secreted by SCs might be responsible for theSchwann-like cell differentiation of ADSCs.

We focused our attention on ADSCs as they can beharvested by less invasive procedures than can BMSCs,and cultured with higher proliferation rates. Similar toBMSCs, ADSCs are non-haematopoietic progenitor cellsand possess multilineage differentiation potential intotissues of mesenchymal origin, as well as tissues of endo-dermal and ectodermal origin. Previous studies havereported that a mixture of glial growth factors (containingGGF ⁄heregulin, FSK, PDGF and bFGF) can induceADSCs to differentiate into cells that show spindle shapeswith bi- or tripolar orientation, morphology resemblingthe appearance of SCs, and express S-100, GFAP andP75NTR, similar to natural SCs (30). However, a furtherstudy has reported that such chemical factors may be apotent stimulus inducing initial, although poorly specific,changes in neural differentiation of MSCs, and that SCco-culture may provide additional, necessary conditionsto complete such a process (21). They also showed thatSC-conditioned medium could not induce morphological

(a) (b)

(c) (d)

Figure 10. DRGNs stained for b-tubulin to showneurite outgrowth. (a) SC ⁄DRGN co-cultures;(b) dADSC ⁄DRGN co-cultures; (c) uADSC ⁄DRGN co-cultures; (d) DRGN cultures. Differ-entiated ADSCs enhanced neurite outgrowth inco-culture with neurons. Schwann cells evoked asimilar response, while undifferentiated ADSCshad a definite effect compared to the neuronscultured alone (magnification: ·200).

� 2010 Blackwell Publishing Ltd, Cell Proliferation, 43, 606–616.

614 Y. Wei et al.

Page 10: Schwann-like cell differentiation of rat adipose-derived stem cells by indirect co-culture with Schwann cells in vitro

changes in MSCs, and their treated MSCs failed toexpress any myelin protein. In our co-culture system,ADSCs were used for culture with SCs in indirectco-culture. After 6 days co-culture, the majority ofADSCs changed shape from a fibroblast-like appearanceto bipolar and tripolar spindle shapes, a characteristic phe-notype of SCs. Most excitingly, morphology of the treatedcells was exactly the same as SCs, when co-cultured for21 days. Immunocytochemical staining revealed thatbipolar and tripolar cells were positive for S-100, GFAPand P75NTR. To confirm that the results obtained byimmunocytochemical staining were not due to any arte-fact of cell shrinkage, western blotting and RT-PCR wereperformed. Lysates of treated ADSCs but not ADSCscultured alone, showed S-100, GFAP and P75NTR-immunoreactive bands corresponding to molecularweights of 12, 52 and 75 kDa respectively. Results of RT-PCR of treated ADSC mRNA probed with S-100, GFAPand P75NTR primers also supported the results foundby immunocytochemical staining. S-100, GFAP andP75NTR gene expressions were detected in ADSCs co-cultured with SCs and SCs cultured alone, but not inADSCs cultured alone, arguing against the notion thatstaining was merely caused by disruption of the cytoskel-eton (31). To investigate whether the cells could maintaintheir differentiated state after removing SCs from theco-culture system, dADSCs were transferred to normalmedium. Absence of SCs induced morphological changesin dADSCs during the first 2 weeks, which retained theability to express SC markers, thus suggesting an indica-tion for long-lasting differentiation. In our experiments,we also found that SCs proliferated rapidly during theco-culture process without adding any additional neuro-trophic factors (data not shown), suggesting cell–cellcross-talk between the two co-cultured cell types by solu-ble secreted proteins, which has also been observed inSC ⁄BMSC co-culture systems in recent reports (32).

One defining property of SCs is their unique affinityfor axons. The manner in which SCs are associated withaxons is an easily recognizable feature of SC ⁄DRGN cul-tures. Thus, it is a well-established method to evaluatemyelinating capacity of dADSCs. When presented withDRGN axons in vitro, extensive myelin was formed inboth dADSC ⁄DRGN and SC ⁄DRGN co-cultures; also, nomyelin was formed in uADSC ⁄DRGN co-cultures duringthe same period. Myelin was not formed in control DRGNcultures. These results demonstrate that dADSCs demon-strate the same property as SCs. Immunocytochemicalstaining demonstrated that dADSCs produced neuriteextensions superior to those seen with uADSCs, and mod-erate neurite growth caused with the latter would suggestthat some benefit can be conferred by stem cells, evenprior to their differentiation.

Cell transplantation needs large numbers of cells, butin vitro expansion potential of committed SCs is limited.In vitro expansion of ADSCs is considered to be a power-ful approach to overcome some practical and ethical con-cerns of cell transplantation. In this study, ADSCs wereobtained from rat testicular adipose tissue. Primary cul-tures of adipose tissue are heterogeneous, containing hae-matopoietic cells, endothelial cells, smooth muscle cellsand pericytes (4). Numbers of these contaminating cells issmall though, and frequency of these cells diminishesquickly through serial passaging (33). Purified rat ADSCsexhibit typical mesenchymal stem cell characteristics:fibroblast morphology and multipotential capability. Weused rat ADSCs at passages 3–5 for our experiments andobserved that they could undergo osteogenic and adipo-genic differentiation. They can be expanded through atleast 10 passages and maintained in their undifferentiatedstate, except for occasional, faint expression of S-100.Compared to previous methods, our protocol is relativelysimple and easy to perform. It is similar to regular cell cul-ture procedures, and does not require any special growthfactors. Furthermore, this method is of low cost and isreproducible, which might become an alternative culturesystem for Schwann-like cell differentiation of ADSCs.

In summary, our results demonstrate that ADSCscan be easily obtained from a small amount of fat tissueand expanded rapidly in culture. These ADSCs have theability to differentiate into osteoblasts and adipocytes.They can be selectively induced to undergo morphologi-cal and phenotypic changes consistent with Schwann-like cell differentiation in an in vitro co-cultureapproach. Furthermore, the dADSCs could form myelinand enhance neurite outgrowth in a co-culture bioassay,which clearly demonstrated functional potential of dif-ferentiated stem cells. Although a broad spectrum ofphenotypic changes consistent with Schwann-like celldifferentiation was observed in treated ADSCs in vitro,further studies are needed to detect which growth fac-tors secreted by SCs play a key role in Schwann-likecell differentiation of ADSCs and to confirm whetherdADSCs can enhance regeneration of injured peripheralnerves after they are transplanted into experimentalnerve bridges.

Acknowledgements

This study was supported by the National Natural ScienceFoundation of China (contract grant number: 30670528,30700848, 30772443), National Basic Research Programof China (also called the 973 Program, contract grantnumber: 2005CB623905) and the National NaturalScience Foundation of Beijing (contract grant number:7082090).

� 2010 Blackwell Publishing Ltd, Cell Proliferation, 43, 606–616.

Schwann-like cell differentiation of rat ADSCs 615

Page 11: Schwann-like cell differentiation of rat adipose-derived stem cells by indirect co-culture with Schwann cells in vitro

References

1 Bunge RP (1993) Expanding roles for the Schwann cell: ensheath-ment, myelination, trophism and regeneration. Curr. Opin. Neurobiol.3, 805–809.

2 Mosahebi A, Woodward B, Wiberg M, Martin R, Terenghi G (2001)Retrovitral labeling of Schwann cells: in vitro characterization and invivo transplantation to improve peripheral nerve regeneration. Glia34, 8–17.

3 Hou SY, Zhang HY, Quan DP, Liu XL, Zhu JK (2006) Tissue-engi-neered peripheral nerve grafting by differentiated bone marrow stro-mal cells. Neuroscience 140, 101–110.

4 Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ et al.(2001) Multilineage cells from human adipose tissue: implicationsfor cell-based therapies. Tissue Eng. 7, 211–228.

5 Baksh D, Song L, Tuan RS (2004) Adult mesenchymal stem cells:characterization, differentiation and application in cell and gene ther-apy. J. Cell Mol. Med. 8, 301–316.

6 De Ugarte DA, Alfonso Z, Zuk PA, Elbarbary A, Zhu M, Ashijian Pet al. (2003) Different expression of stem cell mobilization-associ-ated molecules on multi-lineage cells from adipose tissue and bonemarrow. Immunol. Lett. 89, 267–270.

7 De Ugarte DA, Morizono K, Elbarbary A, Alfonso Z, Zuk PA, ZhuM et al. (2003) Comparison of multi-lineage cells from human adi-pose tissue and bone marrow. Cells Tissues Organs 174, 101–109.

8 Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, MoscaJD et al. (1999) Multilineage potential of adult human mesenchymalstem cells. Science 288, 143–147.

9 Erickson GR, Gimble JM, Franklin DM, Rice HE, Awad H, Guilak F(2006) Chondrogenic potential of adipose tissue-derived stromalcells in vitro and in vivo. Biochem. Biophys. Res. Commun. 290,763–769.

10 Lee JH, Kemp DM (2006) Human adipose-derived stem cells displaymyogenic potential and perturbed function in hypoxic conditions.Biochem. Biophys. Res. Commun. 341, 882–888.

11 Mehlhorn AT, Zwingmann J, Finkenzeller G, Niemeyer P, Dauner M,Stark B et al. (2009) Chondrogenesis of adipose-derived adult stemcells in a poly-lactide-co-glycolide scaffold. Tissue Eng. Part A 15,1159–1167.

12 Altman AM, Yan YS, Matthias N, Bai XW, Rios C, Mathur AB et al.(2009) IFATS collection: human adipose-derived stem cells seededon a silk fibroin-chitosan scaffold enhance wound repair in a murinesoft tissue injury model. Stem Cells 27, 250–258.

13 Zhu Y, Liu T, Song K, Ning R, Ma X, Cui Z (2009) ADSCs differen-tiated into cardiomyocytes in cardiac microenvironment. Mol. Cell.Biochem. 324, 117–129.

14 Wang B, Han J, Gao Y, Xiao ZF, Chen B, Wang X et al. (2007) Thedifferentiation of rat adipose-derived stem cells into OEC-like cellson collagen scaffolds by co-culturing with OECs. Neurosci. Lett.421, 191–196.

15 Franco Lambert AP, Fraga Zandonai A, Bonatto D, CantarelliMachado D, Pegas Henriques JA (2009) Differentiation of humanadipose-derived adult stem cells into neuronal tissue: does it work?Differentiation 77, 221–228.

16 Caddick J, Kingham PJ, Gardiner NJ, Wiberg M, Terenghi G (2006)Phenotypic and functional characteristics of mesenchymal stem cellsdifferentiated along a Schwann cell lineage. Glia 54, 840–849.

17 Strem BM, Hicok KC, Zhu M, Wulur I, Alfonso Z, Schreiber REet al. (2005) Multipotential differentiation of adipose tissue-derivedstem cells. Keio J. Med. 54, 132–141.

18 D’Ippolito G, Schilller PC, Ricordi C, Roos BA, Howard GA (1999)Age-related osteogenic potential of mesenchymal stromal stem cellsfrom human vertebral bone marrow. J. Bone Miner. Res. 14, 1115–1122.

19 Banfi A, Bianchi G, Galotto M, Cancedda R, Quarto R (2001) Bonemarrow stromal damage after chemo ⁄ radiotherapy: occurrence, con-sequences and possibilities of treatment. Leuk. Lymphoma 42, 863–870.

20 Muschler GF, Nitto H, Boehm CA, Easley KA (2001) Age- and gen-der-related changes in the cellularity of human bone marrow and theprevalence of osteoblastic progenitors. J. Orthop. Res. 19, 117–125.

21 Kingham PJ, Kalbermatten DF, Mahay D, Armstrong SJ, Wiberg M,Terenghi G (2007) Adipose-derived stem cells differentiate into aSchwann cell phenotype and promote neurite outgrowth in vitro.Exp. Neurol. 207, 267–274.

22 Xu YF, Liu L, Li Y, Zhou C, Xiong F, Liu ZS et al. (2008) Myelin-forming ability of Schwann cell-like cells induced from rat adipose-derived stem cells in vitro. Brain Res. 1239, 49–55.

23 Wei YJ, Zhou JL, Zheng ZH, Wang AJ, Ao Q, Gong YD et al.(2009) An improved method for isolating Schwann cells from postna-tal rat sciatic nerves. Cell Tissue Res. 337, 361–369.

24 Bunnell BA, Flaat M, Gagliardi C, Patel B, Ripoll C (2008) Adipose-derived stem cells: isolation, expansion and differentiation. Methods45, 115–120.

25 Komiyama T, Nakao Y, Toyama Y, Asou H, Vacanti CA, Vacanti MP(2003) A novel technique to isolate adult Schwann cells for an artifi-cial nerve conduit. J. Neurosci. Methods 122, 195–200.

26 Keilhoff G, Goihl A, Langnase K, Fansa H, Wlof G (2006) Transdif-ferentiation of mesenchymal stem cells into Schwann cell-like myeli-nating cells. Eur. J. Cell Biol. 85, 11–24.

27 Plant GW, Currier PF, Cuervo EP, Bates ML, Pressman Y, BungeMB et al. (2002) Purified adult ensheathing glia fail to myelinateaxons under culture conditions that enable Schwann cells to formmyelin. J. Neurosci. 22, 6083–6091.

28 Dezawa M, Takahashi I, Esaki M, Takano M, Sawada H (2001) Sci-atic nerve regeneration in rats induced by transplantation of in vitrodifferentiated bone-marrow stromal cells. Eur. J. Neurosci. 14, 1771–1776.

29 Ribeiro-Resende VT, Koenig B, Nichterwitz S, Oberhoffner S, Sch-losshauer B (2009) Strategies for inducing the formation of bands ofBungner in peripheral nerve regeneration. Biomaterials 30, 5251–5259.

30 Xu YF, Liu ZS, Liu L, Zhao CP, Xiong F, Zhou C et al. (2008) Neur-ospheres from rat adipose-derived stem cells could be induced intofunctional Schwann cell-like cells in vitro. BMC Neurosci. 9, 21.

31 Lu P, Blesch A, Tuszynski MH (2004) Induction of bone marrowstromal cells to neurons: differentiation, transdifferentiation, or arti-fact? J. Neurosci. Res. 77, 174–191.

32 Wang J, Ding F, Gu Y, Liu J, Gu XS (2009) Bone marrow mesenchy-mal stem cells promote cell proliferation and neurotrophic functionof Schwann cells in vitro and in vivo. Brain Res. 1262, 7–15.

33 Gronthos S, Franklin DM, Leddy HA, Robey PG, Storms RW, Gim-ble JM (2001) Surface protein characterization of human adipose tis-sue-derived stromal cells. J. Cell. Physiol. 189, 54–63.

� 2010 Blackwell Publishing Ltd, Cell Proliferation, 43, 606–616.

616 Y. Wei et al.