edinburgh research explorer · bone chips with sodium hyaluronate, and it was chosen for its proven...

11
Edinburgh Research Explorer Perivascular stem cells Citation for published version: James, AW, Zara, JN, Zhang, X, Askarinam, A, Goyal, R, Chiang, M, Yuan, W, Chang, L, Corselli, M, Shen, J, Pang, S, Stoker, D, Wu, B, Ting, K, Péault, B & Soo, C 2012, 'Perivascular stem cells: a prospectively purified mesenchymal stem cell population for bone tissue engineering', Stem cells translational medicine, vol. 1, no. 6, pp. 510-9. https://doi.org/10.5966/sctm.2012-0002 Digital Object Identifier (DOI): 10.5966/sctm.2012-0002 Link: Link to publication record in Edinburgh Research Explorer Document Version: Publisher's PDF, also known as Version of record Published In: Stem cells translational medicine Publisher Rights Statement: ©AlphaMed Press 2012 General rights Copyright for the publications made accessible via the Edinburgh Research Explorer is retained by the author(s) and / or other copyright owners and it is a condition of accessing these publications that users recognise and abide by the legal requirements associated with these rights. Take down policy The University of Edinburgh has made every reasonable effort to ensure that Edinburgh Research Explorer content complies with UK legislation. If you believe that the public display of this file breaches copyright please contact [email protected] providing details, and we will remove access to the work immediately and investigate your claim. Download date: 19. Apr. 2020

Upload: others

Post on 17-Apr-2020

10 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Edinburgh Research Explorer · bone chips with sodium hyaluronate, and it was chosen for its proven osteoinductive characteristics and clinical translation (human demineralized bone

Edinburgh Research Explorer

Perivascular stem cells

Citation for published version:James, AW, Zara, JN, Zhang, X, Askarinam, A, Goyal, R, Chiang, M, Yuan, W, Chang, L, Corselli, M, Shen,J, Pang, S, Stoker, D, Wu, B, Ting, K, Péault, B & Soo, C 2012, 'Perivascular stem cells: a prospectivelypurified mesenchymal stem cell population for bone tissue engineering', Stem cells translational medicine,vol. 1, no. 6, pp. 510-9. https://doi.org/10.5966/sctm.2012-0002

Digital Object Identifier (DOI):10.5966/sctm.2012-0002

Link:Link to publication record in Edinburgh Research Explorer

Document Version:Publisher's PDF, also known as Version of record

Published In:Stem cells translational medicine

Publisher Rights Statement:©AlphaMed Press 2012

General rightsCopyright for the publications made accessible via the Edinburgh Research Explorer is retained by the author(s)and / or other copyright owners and it is a condition of accessing these publications that users recognise andabide by the legal requirements associated with these rights.

Take down policyThe University of Edinburgh has made every reasonable effort to ensure that Edinburgh Research Explorercontent complies with UK legislation. If you believe that the public display of this file breaches copyright pleasecontact [email protected] providing details, and we will remove access to the work immediately andinvestigate your claim.

Download date: 19. Apr. 2020

Page 2: Edinburgh Research Explorer · bone chips with sodium hyaluronate, and it was chosen for its proven osteoinductive characteristics and clinical translation (human demineralized bone

®

Perivascular Stem Cells: A Prospectively PurifiedMesenchymal Stem Cell Population for Bone TissueEngineering

AARON W. JAMES,a,b,c,d* JANETTE N. ZARA,c,d* XINLI ZHANG,a,b ASAL ASKARINAM,a,b

RAGHAV GOYAL,a,b MICHAEL CHIANG,a,b WEI YUAN,c,d LE CHANG,a,b MIRKO CORSELLI,c,d JIA SHEN,a,b

SHEN PANG,a,b DAVID STOKER,e,f BEN WU,g KANG TING,a,b,c,d BRUNO PÉAULT,c,d,h CHIA SOOc,d

Key Words. Adult stem cells • Pericytes • Mesenchymal stem cells • Osteoblast

aDental and CraniofacialResearch Institute, bSectionof Orthodontics, School ofDentistry, and gDepartmentof Bioengineering, Universityof California, Los Angeles, LosAngeles, California, USA;cDepartment of OrthopaedicSurgery and dOrthopaedicHospital Research Center,Orthopaedic Hospital/University of California, LosAngeles, Los Angeles,California, USA; ePrivatepractice, Marina del Rey,California, USA; fDivision ofPlastic and ReconstructiveSurgery, University ofSouthern California, LosAngeles, California, USA;hCenter for CardiovascularScience and MRC Center forRegenerative Medicine,University of Edinburgh,Edinburgh, United Kingdom

*Contributed equally as firstauthors.

Correspondence: Chia Soo, M.D.,Department of Orthopaedic Sur-gery, University of California, LosAngeles, 675 Charles E. YoungDrive South, MRL 2641A, LosAngeles, California 90095-1579,USA. Telephone: 310-794-5479;Fax: 310-206-7783; e-mail:[email protected]; or Kang Ting,D.M.D., D.Med.Sc., Section ofOrthodontics, UCLA School ofDentistry, CHS 30-117, 10833 LeConte Avenue, Los Angeles, Cali-fornia 90095, USA. Telephone:310-206-6305; Fax: 310-206-5349; [email protected];or Bruno Peault, Ph.D., Depart-ment of Orthopaedic Surgery,David Geffen School of Medicineat UCLA, Orthopaedic HospitalResearch Center, Box 957358,Los Angeles, California 90095-7358. Telephone: 310-794-1339;Fax: 310-825-5409; e-mail:[email protected]

Received January 4, 2012;accepted for publication May 1,2012; first published online inSCTM EXPRESS June 11, 2012.

©AlphaMed Press1066-5099/2012/$20.00/0

http://dx.doi.org/10.5966/sctm.2012-0002

ABSTRACT

Adipose tissue is an ideal source of mesenchymal stem cells for bone tissue engineering: it islargely dispensable and readily accessible with minimal morbidity. However, the stromal vas-cular fraction (SVF) of adipose tissue is a heterogeneous cell population, which leads to unreli-able bone formation. In the present study, we prospectively purified human perivascular stemcells (PSCs) from adipose tissue and compared their bone-forming capacity with that of tradi-tionally derived SVF. PSCs are a population (sorted by fluorescence-activated cell sorting) ofpericytes (CD146�CD34−CD45−) and adventitial cells (CD146−CD34�CD45−), each of which wehave previously reported to have properties of mesenchymal stem cells. Here, we found thatPSCs underwent osteogenic differentiation in vitro and formed bone after intramuscular im-plantation without the need for predifferentiation. We next sought to optimize PSCs for in vivobone formation, adopting a demineralized bone matrix for osteoinduction and tricalcium phos-phate particle formulation for protein release. Patient-matched, purified PSCs formed signifi-cantly more bone in comparison with traditionally derived SVF by all parameters. Recombinantbone morphogenetic protein 2 increased in vivo bone formation but with a massive adipogenicresponse. In contrast, recombinant Nel-like molecule 1 (NELL-1; a novel osteoinductive growthfactor) selectively enhanced bone formation. These studies suggest that adipose-derived hu-man PSCs are a new cell source for future efforts in skeletal regenerative medicine. Moreover,PSCs are a stem cell-based therapeutic that is readily approvable by the U.S. Food and DrugAdministration, with potentially increased safety, purity, identity, potency, and efficacy. Fi-nally, NELL-1 is a candidate growth factor able to induce human PSC osteogenesis. STEM CELLSTRANSLATIONAL MEDICINE 2012;1:510–519

INTRODUCTION

Since the original description of adipose tissue asa source of adult multilineage stem cells [1], thepreclinical study of adipose-derived cells for re-pair and regeneration of tissues has increaseddramatically (reviewed in [2]). The theoreticaladvantages of adipose-derived cells over othercell types for biologic bone regeneration strate-gies are numerous. For example, although au-tograft bone remains the gold standard for bonerepair, its disadvantages are significant, includingdonor site morbidity, extended operating timecomplications, and limited autogenous supply[3–5]. On the other hand, bone marrow mesen-chymal stem cells (BMSCs) have long been stud-ied for their application to skeletal engineering.However, BMSCs have distinct disadvantagesover adipose-derived cells, including limitationsin autogenous supply of bonemarrowaspirate ascompared with lipoaspirate, and relatively lower

cell yield of BMSCs in comparison with adipose-derived stem cells (ASCs) (estimated at �104

stem cells vs. �106 stem cells per 40 ml) [6–8].Finally, human ASCs have been used successfullyto heal small animal [9, 10] and large animal [11]skeletal defects.

Despite the advantages of fat tissue-derivedcells for bone regeneration, there exist clear hur-dles that must be overcome for their successfulclinical use. For example, adipose-derived stro-mal cells are defined by their adherence to cellculture plates and ex vivo expansion. This ex vivoexpansion increases the risk of immunogenicity,infection, and genetic instability [12, 13]. As analternative to ASCs, the cells of noncultured totalstromal vascular fraction (SVF) from adipose tis-sues have been used. Available studies using SVFshow poor and unreliable bone formation [14] or

lower bone regeneration efficacy relative to cul-

tured ASCs [15]. SVF of adipose tissue is a highly

TISSUE ENGINEERING AND REGENERATIVE MEDICINE

STEM CELLS TRANSLATIONAL MEDICINE 2012;1:510–519 www.StemCellsTM.com

Page 3: Edinburgh Research Explorer · bone chips with sodium hyaluronate, and it was chosen for its proven osteoinductive characteristics and clinical translation (human demineralized bone

heterogeneous cell population, including nonmesenchymalstem cell types, such as inflammatory cells, hematopoietic stemcells, and endothelial cells, among others [16]. Variability in cellcomposition presents clear disadvantages forU.S. Food andDrugAdministration (FDA) approval of a future stem cell-based ther-apeutic, potentially including reduced safety, purity, identity, po-tency, and efficacy. Notably, these are the criteria upon whichthe Center for Biologics Evaluation and Research evaluates stemcell-based product applications [17]. For example, given the het-erogeneity of SVF, precise product characterization is not feasi-ble, leading to lack of human stromal vascular fraction (hSVF)product identity. Batch-to-batch variability and nonuniformity ineffect (again attributable to variable stem cell content) reducesthe efficacy and potency of hSVF compounds and potentiallyreduces product safety. With these regulatory hurdles in mind,our approach was to reduce the inherent heterogeneity withinhSVF to obtain a safer andmore efficacious stem cell-based ther-apeutic.

To address this issue of SVF heterogeneity, previous investi-gators have used fluorescence-activated cell sorting (FACS) topurify fat-derived cell populations, with some success. For exam-ple, cell sorting of adipose tissue has allowed for enrichment ofosteoprogenitor [18] or chondroprogenitor [19] cell types—butnot the isolation ofmultipotentmesenchymal stem cells (MSCs).Our solution has been to identify the native perivascular pheno-type of MSCs and prospectively isolate perivascular MSCs usingmulticolor FACS. We have previously shown two populations ofperivascular stem cells (PSCs) associated with blood vessels[20–24]. These include CD34�CD146�CD45� pericytes sur-rounding microvessels and capillaries [20, 25] and a second dis-tinct adventitial cell (CD34�CD146�CD45�) [26], associatedwith larger blood vessels. Both cell types are isolatable from ad-ipose tissue and resembleMSCs bymorphology, growth, surfacemarkers, and clonal multilineage differentiation potential[20–24].

In this report, we describe the use of a novel, FACS-purifiedcell source for improved bone tissue engineering: humanperivascular stem cells (hPSCs). Using an intramuscular, ectopicbone model, we observed that adipose-derived, FACS-isolatedhPSCs formed bone to a greater degree than patient-matched,unsorted hSVF. Next, two growth factors were examined fortheir osteoinductive effects: bone morphogenetic protein 2(BMP2) and Nel-like molecule-1 (NELL-1). The novel cytokineNELL-1 was identified as an appropriate osteogenic stimulusrather than BMP2, which had pleiotropic effects, including adi-pogenic differentiation and cyst-like bone formation.

MATERIALS AND METHODS

Isolation of SVF from Human LipoaspirateHuman lipoaspirate (n� 70 donors) was obtained from patientsundergoing cosmetic liposuction. No patient identifiers were ob-tained. Lipoaspirate was stored for no more than 128 hours at4°C before processing. The hSVF was obtained by collagenasedigestion as previously described [26]. Briefly, lipoaspirate wasdilutedwith an equal volume of phosphate-buffered saline (PBS)before digestion with Dulbecco’s modified Eagle’s medium(DMEM) containing 3.5% bovine serum albumin (Sigma-Aldrich,St. Louis, http://www.sigmaaldrich.com) and 1mg/ml type II col-lagenase for 70 minutes under agitation at 37°C. Next, adi-

pocyteswere separated and removed by centrifugation. The pel-let was then resuspended in red-cell lysis buffer (155mMNH4Cl,10 mM KHCO3, and 0.1 mM EDTA) and incubated for 10 minutesat room temperature. After centrifugation, pellets were resus-pended in PBS and filtered at 70 �m. The resulting hSVF waseither further processed for cell sorting (to isolate PSCs) or usedimmediately for in vivo application. In order to calculate live cellnumber for implantation, trypan blue staining was performed toassess cell viability. Cells were kept on ice until in vivo implanta-tion. n � 70 patient samples were used for in vitro experiments,and n � 6 patient samples were used for in vivo experiments;demographics were recorded, including age, sex, and anatomiclocation of harvest, and are presented in supplemental onlineTable 1.

Purification of PSCs from Human LipoaspiratePSCs were purified by FACS of the hSVF as previously described[26]. hSVF was incubated with amixture of the following directlyconjugated antibodies: anti-CD34-phycoerythrin (1:100; Dako,Glostrup, Denmark, http://www.dako.com), anti-CD45-allophy-cocyanin (1:100; Santa Cruz Biotechnology Inc., Santa Cruz, CA,http://www.scbt.com), and anti-CD146-fluorescein isothiocya-nate (1:100; AbD Serotec, Raleigh, NC, http://www.ab-direct.com). All incubations were performed at 4°C for 15 minutes inthe dark. Before sorting, 4�,6-diamidino-2-phenylindole (DAPI;1:1,000; Invitrogen, Carlsbad, CA, http://www.invitrogen.com)was added for dead cell exclusion; the solution was then passedthrough a 70-�mcell filter and then run on a FACSAria cell sorter(BD Biosciences, San Diego, CA, http://www.bdbiosciences.com). Sorted cells were used for in vivo application immediatelyor plated for in vitro studies. In this manner, distinct microvesselpericytes (CD34�, CD146�, CD45�) and adventitial cells(CD34�, CD146�, CD45�) were isolated and combined to con-stitute the PSC population. For select experiments, fluorescentlabeling of live cells was performed prior to implantation usingPKH26 fluorescent cell linker (Sigma-Aldrich). Cells were kept onice until in vivo implantation.

In Vitro AssaysThe expansion of cells was performed in DMEM, 20% fetal bo-vine serum (FBS), 1% penicillin/streptomycin. Osteogenic differ-entiation of human PSCs was performed as previously described[9, 27], withminormodifications. Passage 4 PSCs were seeded ata density of 35,000 cells perwell in 12-well plates. Differentiationmedium consisted of DMEM, 10% FBS, 100 �g/ml ascorbic acid,and 10mM�-glycerophosphate. Assessments, including alkalinephosphatase and alizarin red staining, were performed as previ-ously described [10, 28].

Implant PreparationImplants were prepared as per supplemental online Tables 2–5.Two different scaffolds were used for cell delivery. First, an ab-sorbable collagen sponge of defined dimensions (0.5 � 1 � 1.5cm) (component of the INFUSE Bone Graft; Medtronic, Minne-apolis, MN, http://www.medtronic.com) was used (data pre-sented in Fig. 1). This carrier was chosen as a nonosteoinductivecarrier, as intramuscular implantation of absorbable collagensponge alone is known to have no bone forming effects [29].Second, an ovine DBX Putty was used (100 �l; MusculoskeletalTransplant Foundation, Edison, NJ, http://www.mtf.org). DBXPutty combines morselized cortical-cancellous demineralized

511James, Zara, Zhang et al.

www.StemCellsTM.com

Page 4: Edinburgh Research Explorer · bone chips with sodium hyaluronate, and it was chosen for its proven osteoinductive characteristics and clinical translation (human demineralized bone

bone chips with sodium hyaluronate, and it was chosen for itsproven osteoinductive characteristics and clinical translation(human demineralized bone matrix [DBX] is already 510(k)approved) (data presented in Figs. 2–5). Select experimentsadded growth factors (recombinant human BMP2 or NELL-1),which were lyophilized onto tricalcium phosphate (�-TCP)particles (Chronos �-TCP granules, 200–300 �m diameter;Synthes, West Chester, PA, http://www.synthes.com) andstored at 4°C prior to use. This avoids burst-type release ki-netics in favor of sustained growth factor release [30]. Con-centrations of BMP2 were based on the clinical-range dosesneeded for human bone formation (up to 1,500 �g/ml) ratherthan the lower doses more commonly used in mice [31]. Con-centrations of NELL-1 were based on our previous publicationusing an intramuscular model and fetal pancreatic pericytes[32]. The individual components (DBX � �-TCP) were mixedmechanically prior to implantation. Finally, defined numbersof viable cells in PBS suspension (20 �l) from the hSVF or PSCswere applied (either allowed to soak into absorbable collagensponge or mechanically mixed with DBX/�-TCP). The cell �scaffold suspension was kept on ice until implantation.

Surgical ProceduresIntramuscular implantation was performed in 8-week-old maleSCID mice. Animals were anesthetized by isoflurane inhalation(5% induction, 2% maintenance) and premedicated with 0.05mg/kg buprenorphine. Bilateral incisions in the hind limbs weremade, and pockets were cut in the biceps femoris muscles byblunt dissection, parallel to the muscle fiber long axis. Implantswere placed bilaterally; implant compositions are fully describedin supplemental online Tables 2–6. Numbers of animals per ex-periment can be found in the figure legends. The fasciae overly-ing the muscle were sutured with a simple continuous pattern,and the skin was closed in a separate layer using 5-0 Vicryl (Ethi-con, San Angelo, TX, http://www.ecatalog.ethicon.com) suturesin a subcuticular pattern. Animals were postoperatively treated

with buprenorphine for 48 hours and trimethoprim/sulfame-thoxazole for 10 days for pain management and infection pre-vention, respectively. Animals were housed and experimentswere performed in accordance with guidelines of the Chan-cellor’s Animal Research Committee of the Office for Protec-tion of Research Subjects at the University of California, LosAngeles.

Radiographic Imaging: High-Resolution XR andQuantitative Microcomputed Tomography AnalysisLive high-resolution radiographs were taken every 2 weeks forselect experiments using a cabinet radiography system (FaxitronBioptics, Lincolnshire, IL, http://www.faxitron.com). Mice wereanesthetized using isoflurane (5% induction, 2% maintenance).

Samples were harvested en bloc from 4 to 6 weeks postim-plantation, formalin-fixed, and imaged using high-resolution mi-crocomputed tomography (microCT) (Skyscan 1172F; Skyscan,Kontich, Belgium, http://www.skyscan.be) at an image resolu-tion of 19.73 �m and analyzed using DataViewer, Recon, CTAn,and CTVol programs provided by the manufacturer. For com-puted tomography (CT) data analysis, the region of interest in-cludes the entire volume of the scannedmuscle pouch excludingthe native femur to quantify bone volume (BV) and bonemineraldensity (BMD). Threshold values for three-dimensional recon-structions and quantification can be found in the figure legends.All quantitative and structural morphometric data use nomen-clature described by the American Society for Bone and MineralResearch Nomenclature Committee [33].

Histology and Immunohistochemistry AnalysisAfter CT analysis, samples were decalcified in 19% EDTA andembedded in paraffin. Hematoxylin andeosin (H&E) andMovat’spentachrome staining were performed as previously described[32]. For select experiments, bone formation was quantified byblinded histomorphometric analyses using H&E staining (Figs. 2,6) and Photoshop (Adobe Systems Inc., San Jose, CA, http://

Figure 1. Human perivascular stem cells (hPSCs) are an osteocompetent cell population. (A, B): hPSCs were cultured under osteogenicconditions (10% fetal bovine serum, 100�g/ml ascorbic acid, 10mM�-glycerophosphate). (A):Representative image of alkaline phosphatasestaining at 5 days of differentiation. (B): Representative image of alizarin red staining, 10 days of differentiation. (C–F): hPSCs were implantedin the thigh complex of a SCID mouse using a collagen sponge carrier (2.5 � 105 cells, sponge size 2.0 � 1.0 � 0.5 cm). (C): High-resolutionradiography at 2 and 4 weeks postimplantation. (D): Three-dimensional microcomputed tomography reconstructions at threshold 90. (E, F):Pentachrome staining of histological sections. n � 5 implants, from n � 1 patient specimens. See supplemental online Table 2 for treatmentgroups. Abbreviations: cb, cortical bone; eb, ectopic bone; m, muscle.

512 Perivascular Stem Cells in Bone Formation

STEM CELLS TRANSLATIONAL MEDICINE

Page 5: Edinburgh Research Explorer · bone chips with sodium hyaluronate, and it was chosen for its proven osteoinductive characteristics and clinical translation (human demineralized bone

www.adobe.com) quantification as expressed by mean bonearea and bone area/tissue area [34]. For select experiments, ad-ipogenesis was quantified by blinded calculations of lipid dropletnumber per �100 field (Fig. 4). Immunohistochemistry wasperformed with primary antibodies against bone sialoprotein(Chemicon, Temecula, CA, http://www.chemicon.com), osteo-calcin (Santa Cruz Biotechnology), and major histocompatibilitycomplex (MHC) class I surface antigen (Santa Cruz Biotechnol-

ogy) using the ABC (Vector Laboratories, Burlingame, CA, http://www.vectorlabs.com)method. Select quantification of immuno-histochemistry was performed again using the magic wand toolin Photoshop with a tolerance setting of 30 (Fig. 2).

Statistical AnalysisStatistical analysis was performed using an appropriate analysisof variance test when more than two groups were compared,

Figure 2. hPSCs undergomore robust differentiation in comparison with hSVF. Equal numbers of viable hSVF cells or hPSCs (2.5� 105) fromthe same patient samples were implanted intramuscularly in the thigh of a SCID mouse. An osteoinductive DBX Putty was used as scaffold.Assessments were performed at 4 weeks postimplantation. (A, B):Microcomputed tomography (microCT) images of hSVF- and hPSC-treatedsamples. Representative three-dimensional microCT reconstructions at threshold (Th) 90 (left) and corresponding two-dimensional axialslices (right). (C, D): Analysis of BV and BMD among hSVF- and hPSC-treated samples; Th50–120. (E): Representative hematoxylin and eosinstaining. (F): Representative histomorphometric quantification of bone area per �100 field. (G): Representative BSP immunohisto-chemistry. (H): Quantification of relative staining per �400 field. (I): Representative OCN immunohistochemistry. (J): Quantification ofrelative staining per �400 field. Histomorphometric quantification calculated from n � 6 randommicroscopical fields. n � 12 implantsper cell type, n � 3 patient specimens. See supplemental online Table 3 for treatment groups. �, p � .05 in comparison with control asassessed by Student’s t test. Abbreviations: BMD, bone mineral density; BSP, bone sialoprotein; BV, bone volume; eb, ectopic bone; f,femur; hPSC, human perivascular stem cell; hSVF, human stromal vascular fraction; OCN, osteocalcin.

513James, Zara, Zhang et al.

www.StemCellsTM.com

Page 6: Edinburgh Research Explorer · bone chips with sodium hyaluronate, and it was chosen for its proven osteoinductive characteristics and clinical translation (human demineralized bone

followed by Student’s t test to compare two groups. Inequality ofSDs was excluded by using Levene’s test. p� .05 was consideredto be significant.

RESULTS

Stromal Vascular Fraction of Human White AdiposeTissue Contains Two Distinct Perivascular MSCPopulations: Pericytes and Adventitial CellsWe recently described, purified, and characterized two perivas-cular sources of primary MSCs within human adipose tissuebasedon the expression of CD34 andCD146. Pericytes,which aresurrounding microvessels, are isolatable from multiple humanorgans, including adipose tissue, and are CD146�CD34�CD45�[20, 25]. More recently, we have described a second and dis-tinct perivascular MSC population, adventitial cells(CD146�CD34�CD45�) [26], obtainable from human adiposeamong other tissues. Here, we first confirmed our previous re-sults, showing that pericytes (CD146�CD34�) and adventitialcells (CD146�CD34�) are distinct populations isolatable fromhuman lipoaspirate after exclusion of CD45� cells (hematopoi-etic cells) and DAPI� cells (dead cells) (Fig. 6). Both pericytes andadventitial cells have been previously reported to undergo mul-tiple mesodermal lineage differentiation [20, 26]. With n � 70unique samples of lipoaspirate, we observed the mean preva-lence of pericytes and adventitial cells to be 17.1% and 22.5% ofthe total viable cell yield of SVF. From a tissue-engineeringstandpoint, maximal MSC yield per unit of autologous tissue re-moved would improve efficacy for tissue engineering while re-ducing harvest morbidity. For this reason, we chose to combineboth perivascular MSC populations—collectively termedPSCs—to maximize cell yield for our efforts in bone engineering(collectively, PSCs make up, on average, 39.6% of total viableSVF). Importantly, those cells within adipose tissue that areneither pericytes (CD146�CD34�) nor adventitial cells(CD146�CD34�) do not express typical MSC markers or un-dergo mesodermal lineage differentiation [26].

Perivascular Stem Cells Undergo OsteogenicDifferentiation In Vitro and Form Bone In VivohPSCswere first evaluated for their ability to undergo osteogenicdifferentiation in vitro and bone forming potential in vivo (Fig. 1).In standard osteogenic medium, hPSCs showed robust alkalinephosphatase staining after 5 days in differentiation medium and

bone nodule formation after 10 days in differentiation medium(Fig. 1A, 1B). Freshly derived hPSCs were next placed on a colla-gen sponge without predifferentiation or growth factor stimula-tion and inserted intramuscularly (see supplemental online Ta-ble 2 for treatment groups). Over the course of 6 weeks,spontaneous mineralization was observed by radiography(Fig. 1C), three-dimensional CT (Fig. 1D), and histology (Fig. 1E,1F). In contrast, intramuscular implantation of absorbable colla-gen sponge alone is known to have no bone forming effects [29].Collectively, these studies showed that hPSCs (like their individ-ual pericyte and adventitial components) undergo osteogenicdifferentiation in vitro and in vivo and thus may be useful as apurified cell source for bone formation.

Human PSCs Undergo More Robust Bone Formationthan hSVF CellsHaving demonstrated that hPSCs are an osteocompetent cellsource, we next sought to directly compare bone formation inpatient-matched, purified hPSCs to unsorted hSVF. For this head-to-head determination, the same numbers of viable hSVF cells orhPSCs were implanted (2.5 � 105) intramuscularly. An osteoin-ductive carrierwas used: demineralized bonematrix (DBXPutty),which consists of morselized cortical-cancellous demineralizedbone chips with sodium hyaluronate (Musculoskeletal Trans-plant Foundation) (see supplemental online Table 3 for treat-ment groups). At 4 weeks postimplantation, two- and three-di-mensional CT images showed significantly increased boneformation in hPSC in comparison to hSVF samples (Fig. 2A, 2B).CT quantification demonstrated a statistically significant in-crease in BV and BMD in hPSC-treated samples (Fig. 2C, 2D).Histologic analysis was performed by routine H&E (Fig. 2E).Histomorphometric quantification of bone area per randomhigh-powered field showed increased bone among hPSC-treatedsamples (Fig. 2F). Immunohistochemical staining for the bone-specific markers bone sialoprotein and osteocalcin was per-formed (Fig. 2G, 2I). In each instance, photographic quantitationof positive staining intensity showed a significant increase inboth bone markers in hPSC-treated specimens (Fig. 2H, 2J). Im-portantly, we confirmed that those cells surrounding and inter-spersed between bone chips were of predominantly human ori-gin (Fig. 3), using immunohistochemical staining for MHC class Ihuman surface antigen (Fig. 3A) or via fluorescent labeling ofhSVF cells or hPSCs prior to implantation (Fig. 3B). Thus, equiva-lent numbers of purified hPSCs result in increased ectopic boneformation in comparison with patient-matched hSVF cells.

Figure 3. Persistence of hSVF or hPSCsafter implantation. Shown are hSVF- orhPSC-treated samples implanted intra-muscularly on a DBX Putty scaffold andassessed for cell persistence 4 weekspostimplantation. (A): Human majorhistocompatibility complex class I im-munohistochemistry, indicating the pre-sence of human xenografted cells. (B):Fluorescent cell labeling (PKH) per-formed prior to implantation. Red indi-cates persistence of human cells, andHoechst nuclear counterstain appearsblue. Abbreviations: hPSC, human peri-vascular stem cell; hSVF, human stromalvascular fraction.

514 Perivascular Stem Cells in Bone Formation

STEM CELLS TRANSLATIONAL MEDICINE

Page 7: Edinburgh Research Explorer · bone chips with sodium hyaluronate, and it was chosen for its proven osteoinductive characteristics and clinical translation (human demineralized bone

Having demonstrated that equal numbers of hPSCs form in-creased bone as compared with hSVF, we next asked as towhether this difference could be overcome by simply addingmore hSVF cells. For this purpose, two concentrations of cells

were used in the same intramuscular model: either 2.5 � 104 or2.5 � 105 cells per implant. All hSVF or hPSC samples were pa-tient-matched. If reduced MSC numbers were the only differ-ence between hSVF and hPSC, then increasing the hSVF cell num-ber 10-fold would surely mitigate any observed differences.However, we found this not to be the case (supplemental onlineFig. 1). Namely, hPSCs formed greater bone irrespective of cell

Figure 4. Effects of BMP2 on hSVF- or hPSC-mediated ectopic boneformation. hSVF or hPSCswere implanted intramuscularlywith orwith-out BMP2 on a DBX Putty (DBX) scaffold and assessed 4 weeks postim-plantation. See supplemental online Table 5 for treatment groups.(A, B): Colorized representative two-dimensional axial microcomputedtomography images. A scale bar below indicates colorization at variousthresholds. (C):Relative BV. Values are normalized to hSVF�DBXonly.Threshold 100 was used. (D): Relative BMD. Values are normalized tohSVF� DBX only. (E, F): Representative hematoxylin and eosin imagesat low (E) and high (F)magnifications. n� 8 implants per cell type, n�1 patient specimen. (G): Relative lipid droplet number per �100 field.Meanswere calculated from10 randommicroscopic fields.�,p� .05 incomparisonwith hSVF control; #, p� .05 in comparisonwith hPSC con-trol; ��, p� .05 in comparisonwith hSVFwith same dose of BMP2. SeesupplementalonlineTable4 for treatmentgroups.Abbreviations:BMD,bonemineral density; BMP,bonemorphogenetic protein; BV, bonevol-ume; cs, cortical shell; eb, ectopic bone; f, femur; hPSC, human perivas-cular stem cell; hSVF, human stromal vascular fraction.

Figure 5. Effects of NELL-1 on hPSC- or hSVF-mediated ectopic boneformation. hSVF or hPSCs in a SCID mouse muscle pouch with DBXPutty,with orwithoutNELL-1, assessed at 4weeks postimplantation.See supplemental online Table 6 for treatment groups. (A): Repre-sentative three-dimensional microcomputed tomography (microCT)reconstructions. Threshold (Th) 90 was used. (B): Colorized repre-sentative two-dimensional microCT images. A scale bar below indi-cates colorization at various thresholds. (C): Relative BV; Th50–120used. (D): Relative BMD. (E, F): Representative hematoxylin and eo-sin staining at low (E) and high (F)magnifications. (G, H): Histomor-phometric analysis of mean bone area (G) and mean bone area/tissue area (H), as calculated from 30 random high-powered-fields.n � 9 implants per treatment group, n � 1 patient specimen. �, p �.05 in comparison with hSVF control; #, p � .05 in comparison withhPSC control; ��, p � .05 in comparison with hSVF with same doseNELL-1. See supplemental online Table 5 for treatment groups. Ab-breviations: BMD, bone mineral density; BV, bone volume; hPSC,human perivascular stem cell; hSVF, human stromal vascular frac-tion; NELL-1, Nel-like molecule 1.

515James, Zara, Zhang et al.

www.StemCellsTM.com

Page 8: Edinburgh Research Explorer · bone chips with sodium hyaluronate, and it was chosen for its proven osteoinductive characteristics and clinical translation (human demineralized bone

concentration than hSVF. This was observed by microCT recon-structions of representative samples (supplemental onlineFig. 1A), microCT quantifications of BV and BMD (supplementalonline Fig. 1B, 1C), and representative histological fields (supple-mental online Fig. 1D, 1E). Thus, a simple reduction in stem cellnumber likely does not explain the difference in bone formationbetween hSVF and purified hPSC.

BMP2 Promotes hPSC Osteogenic and AdipogenicDifferentiationThemost commonly used growth factor for bone regeneration isBMP2 (INFUSE Bone Graft) [35]. We next sought to determinewhether BMP2 would be an appropriate stimulus for hPSC boneformation (see supplemental online Table 4 for treatmentgroups). Two-dimensional axial CT scans showed a clear increasein bone formation among both hSVF- and hPSC-treated samplesin response to BMP2 (Fig. 4A, 4B). In addition, a striking trendwas observed in which a thin cortical shell (indicated by whitearrows in Fig. 4A) was observed in BMP2-treated specimens thatextended well beyond the usual ectopic bone margins (Fig. 4A,4B). Quantification of BV and BMD were next performed withBMP2-treated specimens. First, we again found that BV andBMDwere increased in hPSC as compared with hSVF-treated samples(Fig. 4C, 4D, far left). However, the effects of BMP2were to someextent unexpected. Although BMP2 produced a significant in-crease in BV (Fig. 4C), it had a negative and opposing effect onBMD as comparedwith hSVF or hPSC alone (Fig. 4D). Histologicalevaluation was more revealing (Fig. 4E, 4F). In hSVF and hPSCsampleswithout BMP2, bone chipswere closer together (Fig. 4E)and were interspersed with cells with fibroblastic morphology(Fig. 4F, left). In BMP2-treated hSVF or hPSC samples, however,bone chips were farther apart and predominantly interspersed

with lipid droplets (Fig. 4F). In order to quantify the adipogenicresponse produced by BMP2, relative lipid droplet number per�100 fieldwas determined.Within both hSVF- and hPSC-treatedsamples, BMP2 induced a statistically significant increase in lipiddroplet number (Fig. 4G). Thus, BMP2 had both pro-osteogenicand also proadipogenic effects within both hSVF- and hPSC-treated implants and may not be the most appropriate cytokinestimulus for induction of hPSC-mediated high-quality bone for-mation.

NELL-1 Promotes hPSC Osteogenic Differentiation OnlyIn clinical practice, the only approved growth factor alternativeto BMP2 is BMP7 (or OP-1; Stryker, Kalamazoo, MI, http://www.stryker.com); however, BMP7 is also known to have proadipo-genic effects [36, 37]. In contrast, NELL-1 is a growth factor inpreclinical development that has potent pro-osteogenic effectsin small and large mammalian models (reviewed in [38]) and hasbeen recently described to have antiadipogenic effects [26].NELL-1 was next added to hSVF- or hPSC-laden intramuscularimplants (see supplemental online Table 5 for treatmentgroups). At 4 weeks postimplantation, hPSC-treated implantsagain showed increased bone formation in comparison withhSVF, as shown by CT reconstructions (Fig. 5A, 5B) and quantifi-cation (Fig. 5C, 5D). The addition of NELL-1 led to a significantincrease in BV and BMD among hPSC-treated implants (Fig. 5C,5D, far right column). Histologic and histomorphometric analysisrevealed similar findings between treatment groups (Fig. 5E–5H). Bone chips were noted to be closer together among hPSC-treated or hPSC � NELL-1-treated implants (Fig. 5E, 5F). Histo-morphometric calculations of bone area and bone area/tissuearea confirmed these observations (Fig. 5G, 5H). A significantincrease in both parameters was observed between hPSC-treated and hSVF-treated implants. Moreover, hPSC � NELL-1 led

Figure 6. Human white adipose tissue houses distinct pericyte and adventitial cell populations, which together represent perivascular stemcells (PSCs). (A, B): After exclusion of DAPI� dead cells (A) and CD45� hematopoietic cells (B), two PSC populations were isolated withdifferential expression of CD34 and CD146. (C): Distinct CD34�CD146� adventitial cells and CD34�CD146� pericytes were obtained andcombined for in vivo implantation. (D): The relative numbers of pericytes and adventitial cells are depicted as a fraction of the total DAPI� SVFcells. Pericytes and adventitial cells make up approximately 17.1% and 22.5%, respectively. PSCs then represent on average 39.6% of totalviable cells. Abbreviations: DAPI, 4�,6-diamidino-2-phenylindole; FSC, forward scatter; SSC, side scatter; SVF, stromal vascular fraction.

516 Perivascular Stem Cells in Bone Formation

STEM CELLS TRANSLATIONAL MEDICINE

Page 9: Edinburgh Research Explorer · bone chips with sodium hyaluronate, and it was chosen for its proven osteoinductive characteristics and clinical translation (human demineralized bone

to a significant increase over hPSC treatment alone. Unlike BMP2,minimal lipid was observed among NELL-1-treated samples.

DISCUSSION

Perivascular Cells as a Purified Stem Cell SourceFor the first time, we have shown that a purified population ofadipose-derived stem cells (hPSCs) is superior to hSVF for bonetissue regeneration. We have shown this in terms of potency(increased bone formation over unsorted hSVF), identity, andpurity (defined cell surface markers yielding an MSC populationof increased homogeneity without extraneous materials andother cell lineages, such as hematopoietic and endothelial). Intheory, hPSCs would also demonstrate improved potency overhSVF, as a defined and homogeneous cell populationwould havemore predictable in vivo outcomes than a nonhomogeneousmixture. Moreover, hPSCs are an attractive cell source, as theyare plentiful within adipose tissue (on average constituting�40% of total viable hSVF). In this regard, even those patientswith minimal excess body fat could donate autologous fat tissuefor hPSC harvest. Moreover, sufficient quantities of hPSCs couldbe harvested without the need for time-consuming and costly invitro expansion. Avoidance of in vitro expansion also reduces therisk of immunogenicity, infection, and genetic instability [12, 13].

Mechanisms of hPSC Improved OsteogenicityAlthough hPSCs showed improved bone formation in compari-son with hSVF cells across all experiments and all parameters,the underlying mechanism by which hPSCs produce higher boneformation is only partially elucidated. One explanation is thathSVF simply contains fewer MSCs than a purified hPSC popula-tion. However, our experiments refute this hypothesis. For ex-ample, adding hSVF cells at a 10:1 ratio in comparisonwith hPSCsstill led to inferior bone formation (supplemental online Fig. 1).Another possible explanation is that within hSVF there exists adifferentiation-inhibiting cell type. In this regard, endothelialcells have been described to negatively regulate the differentia-tion of MSCs, such as ASCs or BMSCs [39, 40]. For instance, hu-man endothelial cells inhibit BMSC differentiation into matureosteoblasts by interfering with Osterix expression [40]. With thisevidence inmind, hPSCs (which are CD31�)may showenhancedbone formation in part by elimination of the endothelial cellcomponent. Finally, the trophic effect of hPSCs should not beoverlooked. Perivascular cells have been observed to secretehigh levels of pro-osteogenic, provasculogenic growth factors,such as vascular endothelial growth factor, fibroblast growth fac-tor 2, and epidermal growth factor [24]. PSCs may induce robustin vivo bone formation largely through trophic effects via para-crine induction of vasculogenesis and osteogenesis.

Choice of Osteoinductive Growth FactorThemarriage of an osteocompetent cell sourcewith osteoinduc-tive growth factors is a logical union for skeletal tissue engineer-ing. We examined both BMP2 and NELL-1 as candidate growthfactors. BMP2 (INFUSE Bone Graft) is the main FDA-approvedosteoinductive factor, but it has significant undesirable side ef-fects, including life-threatening cervical swelling [41], osteoclastactivation [42], adipogenic differentiation [43], and bone cystformation [44, 45]. In particular to our study, we found thatBMP2 induced a massive adipogenic response and showed poor

bone quality (reduced bone mineral density) (Fig. 4). This is in linewith BMP2’s known effects in enhancing the transcription and ac-tivity of peroxisome proliferator-activated receptor �, the maintranscription factor regulating adipocyte lineage commitment [46,47]. In contrast,NELL-1 showedamoreosteospecific response,withnoapparent inductionof adipogenesis (Fig. 5).Our laboratory’s pre-vious publications show that exogenous NELL-1 induces significantbone formation in such models as critical sized rat calvarial defects[45]; rat femoral segmental defects [48]; and spinal fusion in rat [30,49], sheep [50], and nonhuman primate models (manuscript inpreparation). The pro-osteogenic effect of NELL-1was also recentlyreportedusingpericytesderived fromfetalpancreas inan intramus-cular ectopic bone environment [32].

CONCLUSION

In summary, these data show that hPSCs are readily obtainablefrom human lipoaspirate. Moreover, hPSCs are able to formbone in vivo without the need for predifferentiation. Next, un-cultured hPSCswere observed to be superior to patient-matchedhSVF with respect to the quantity and quality of bone formation.Finally, through a series of in vivo experiments, we have opti-mized a combination product for local bone formation by deliv-ering autologous, purified adult hPSCs and an osteoinductiveprotein (NELL-1) on an acellular allograft bone scaffold.

PSCs not only show improved efficacy in bone formation rela-tive to sample-matched hSVF but also show improvements in otherparameters for FDA approval. These include enhanced purity andidentity (PSCs are anMSC population containing no hematopoieticstem cells, endothelial cells, or nonviable cells), and potency (as amore defined cell population, hPSCs will likely have reduced vari-ability in effect as compared with hSVF). In fact, the intramuscularimplantation assay describedmay well serve as an easily replicableassay for hPSCpotency in bone formation. Excitingly, recent studieshave already demonstrated the utility of perivascular stem cells forregeneration of disparate tissue types, including skeletal muscle[51], lung [52], and even myocardium [25]. Further studies will ex-tendour findingsandapply therobustosteogenicpotentialofhPSCsto the healing of bone defects.

ACKNOWLEDGMENTS

This work was supported by California Institute for RegenerativeMedicine (CIRM) Early Translational II Research Award TR2-01821,the NIH/National Institute of Dental and Craniofacial Research(Grants R21-DE0177711 andR01-DE01607), University of CaliforniaDiscoveryGrant 07-10677, T32 training fellowship (5T32DE007296-14; to A.W.J.), and a CIRM Training Grant Research Fellowship (TG-01169; toM.C. and J.N.Z.).

AUTHOR CONTRIBUTIONS

A.W.J. and J.N.Z.: conception and design, collection and/or as-sembly of data, data analysis and interpretation, manuscriptwriting; X.Z., R.G., and J.S.: collection and/or assembly of data,data analysis and interpretation, manuscript writing; A.A., M.Chiang, W.Y., L.C., M. Corselli, and S.P.: collection and/or assem-bly of data, data analysis and interpretation; D.S. and B.W.: pro-vision of studymaterial; K.T.: financial support, administrative sup-port, manuscript writing; B.P. and C.S.: conception and design,

517James, Zara, Zhang et al.

www.StemCellsTM.com

Page 10: Edinburgh Research Explorer · bone chips with sodium hyaluronate, and it was chosen for its proven osteoinductive characteristics and clinical translation (human demineralized bone

financial support, administrative support, manuscript writing, finalapproval of manuscript.

DISCLOSURE OF POTENTIAL CONFLICTS OF INTEREST

X.Z., K.T., B.W., and C.S. are inventors of NELL-1 related patents,and K.T., B.P., and C.S. are inventors of perivascular stem cell-

related patents filed from the University of California, Los Ange-les. X.Z., K.T., B.W., and C.S. are founders of Bone Biologics Inc.,which sublicenses NELL-1 patents from the University of Califor-nia Regents, and K.T. and C.S. are founders of Scarless Laborato-ries Inc., which sublicenses perivascular stem cell-related pat-ents from the University of California Regents. C.S. is also anofficer of Scarless Laboratories, Inc.

REFERENCES1 Zuk PA, Zhu M, Ashjian P et al. Human

adipose tissue is a source of multipotent stemcells. Mol Biol Cell 2002;13:4279–4295.2 Zuk PA. The adipose-derived stem cell:

Looking back and looking ahead. Mol Biol Cell2010;21:1783–1787.3 Laurencin C, Khan Y. BoneGraft Substitute

Materials. emedicine 2004. Available at http://emedicine.medscape.com/article/1230616-overview. Accessed February 16, 2012.4 Laurie SW, Kaban LB, Mulliken JB et al. Do-

nor-site morbidity after harvesting rib and iliacbone. Plast Reconstr Surg 1984;73:933–938.5 Frodel JL, Jr., Marentette LJ, Quatela VC

et al. Calvarial bone graft harvest. Techniques,considerations, and morbidity. Arch Otolaryn-gol Head Neck Surg 1993;119:17–23.6 Meliga E, Strem BM, Duckers HJ et al. Ad-

ipose-derived cells. Cell Transplant 2007;16:963–970.7 De Ugarte DA, Morizono K, Elbarbary A

et al. Comparison of multi-lineage cells fromhuman adipose tissue and bone marrow. CellsTissues Organs 2003;174:101–109.8 Aust L, Devlin B, Foster SJ et al. Yield of hu-

man adipose-derived adult stem cells from lipo-suction aspirates. Cytotherapy 2004;6:7–14.9 Levi B, James AW, Nelson ER et al. Human

adipose derived stromal cells heal critical sizemouse calvarial defects. PLoS One 2010;5:e11177.10 Levi B, James AW, Nelson ER et al. Hu-

man adipose-derived stromal cells stimulateautogenous skeletal repair via paracrineHedgehog signaling with calvarial osteoblasts.Stem Cells Dev 2011;20:243–257.11 Cui L, Liu B, Liu G et al. Repair of cranial

bone defects with adipose derived stem cellsand coral scaffold in a canine model. Biomate-rials 2007;28:5477–5486.12 Gad SC. Pharmaceutical Manufacturing

Handbook: Regulations and Quality. Hoboken,NJ: John Wiley and Sons, 2008.13 Dahl JA, Duggal S, Coulston N et al. Ge-

netic and epigenetic instability of human bonemarrow mesenchymal stem cells expanded inautologous serum or fetal bovine serum. Int JDev Biol 2008;52:1033–1042.14 Muller AM,Mehrkens A, Schafer DJ et al.

Towards an intraoperative engineering of os-teogenic and vasculogenic grafts from the stro-mal vascular fraction of human adipose tissue.Eur Cell Mater 2010;19:127–135.15 Cheung WK, Working DM, Galuppo LD

et al. Osteogenic comparison of expanded anduncultured adipose stromal cells. Cytotherapy2010;12:554–562.16 Paredes B, Santana A, Arribas MI et al.

Phenotypic differences during the osteogenicdifferentiation of single cell-derived clones iso-

lated from human lipoaspirates. J Tissue EngRegen Med 2011;5:589–599.17 Lanza R. Essentials of Stem Cell Biology.

Oxford, U.K.: Elsevier, 2009.18 Levi B,WanDC, Glotzbach JP et al. CD105

protein depletion enhances human adipose-derived stromal cell osteogenesis through re-duction of transforming growth factor beta1(TGF-beta1) signaling. J Biol Chem 2011;286:39497–39509.19 Jiang T, Liu W, Lv X et al. Potent in vitro

chondrogenesis of CD105 enriched human ad-ipose-derived stem cells. Biomaterials 2010;31:3564–3571.20 Crisan M, Yap S, Casteilla L et al. A

perivascular origin for mesenchymal stem cellsin multiple human organs. Cell Stem Cell 2008;3:301–313.21 CrisanM, Huard J, Zheng B et al. Purifica-

tion and culture of human blood vessel-associ-ated progenitor cells. Curr Protoc Stem CellBiol 2008;Chapter 2:Unit 2B.2.1–2B.2.13.22 Crisan M, Deasy B, Gavina M et al. Puri-

fication and long-term culture of multipotentprogenitor cells affiliated with the walls of hu-man blood vessels: Myoendothelial cells andpericytes. Methods Cell Biol 2008;86:295–309.23 Corselli M, Chen CW, Crisan M et al.

Perivascular ancestors of adult multipotentstem cells. Arterioscler Thromb Vasc Biol 2010;30:1104–1109.24 Chen CW, Montelatici E, Crisan M et al.

Perivascular multi-lineage progenitor cells inhuman organs: Regenerative units, cytokinesources or both? Cytokine Growth Factor Rev2009;20:429–434.25 Crisan M, Chen CW, Corselli M et al.

Perivascular multipotent progenitor cells in hu-man organs. Ann NY Acad Sci 2009;1176:118–123.26 Corselli M, Chen CW, Sun B et al. The

tunica adventitia of human arteries and veinsas a source of mesenchymal stem cells. StemCells Dev 2012;21:1299–1308.27 James AW, Levi B, Nelson ER et al. Dele-

terious effects of freezing on osteogenic differ-entiation of human adipose-derived stromalcells in vitro and in vivo. Stem Cells Dev 2011;20:427–439.28 James AW, Pan A, Chiang M et al. A new

function of Nell-1 protein in repressing adipo-genic differentiation. Biochem Biophys ResCommun 2011;411:126–131.29 Tsai CH, Chou MY, Jonas M et al. A com-

posite graft material containing bone particlesand collagen in osteoinduction in mouse.J Biomed Mater Res 2002;63:65–70.30 Li W, Lee M, Whang J et al. Delivery of

lyophilized Nell-1 in a rat spinal fusion model.Tissue Eng Part A 2010;16:2861–2870.31 Boden SD, Kang J, Sandhu H et al. Use of

recombinant human bone morphogenetic pro-tein-2 to achieve posterolateral lumbar spine fu-

sion in humans: A prospective, randomized clini-calpilot trial: 2002VolvoAward inclinical studies.Spine (Phila Pa 1976) 2002;27:2662–2673.32 Zhang X, Peault B, Chen W et al. The

Nell-1 growth factor stimulates bone forma-tion by purified human perivascular cells. Tis-sue Eng Part A 2011;17:2497–2509.33 Parfitt AM, Drezner MK, Glorieux FH et al.

Bone histomorphometry: Standardization of no-menclature, symbols, andunits.Reportof theAS-BMR Histomorphometry Nomenclature Com-mittee. J BoneMiner Res 1987;2:595–610.34 James AW, Theologis AA, Brugmann SA

et al. Estrogen/estrogen receptor alpha signal-ing in mouse posterofrontal cranial suture fu-sion. PLoS One 2009;4:e7120.35 Bueno EM, Glowacki J. Cell-free and cell-

based approaches for bone regeneration. NatRev Rheumatol 2009;5:685–697.36 Neumann K, Endres M, Ringe J et al.

BMP7 promotes adipogenic but not osteo-/chondrogenic differentiation of adult humanbone marrow-derived stem cells in high-den-sity micro-mass culture. J Cell Biochem 2007;102:626–637.37 Tseng YH, Kokkotou E, Schulz TJ et al.

New role of bone morphogenetic protein 7 inbrown adipogenesis and energy expenditure.Nature 2008;454:1000–1004.38 Zhang X, Zara J, Siu RK et al. The role of

NELL-1, a growth factor associatedwith cranio-synostosis, in promoting bone regeneration. JDent Res 2010;89:865–878.39 Rajashekhar G, Traktuev DO, Roell WC

et al. IFATS collection: Adipose stromal cell dif-ferentiation is reduced by endothelial cell con-tact and paracrine communication: Role of ca-nonical Wnt signaling. STEM CELLS 2008;26:2674–2681.40 Meury T, Verrier S, Alini M. Human en-

dothelial cells inhibit BMSC differentiation intomature osteoblasts in vitro by interfering withosterix expression. J Cell Biochem 2006;98:992–1006.41 Schultz D. FDA public health notification:

Life-threatening complications associated withrecombinant human bone morphogeneticprotein in cervical spine fusion. Available athttp://www.fda.gov/MedicalDevices/Safety/AlertsandNotices/PublicHealthNotifications/ucm062000.htm. Accessed March 9, 2012.42 Kaneko H, Arakawa T, Mano H et al. Di-

rect stimulation of osteoclastic bone resorp-tion by bone morphogenetic protein (BMP)-2and expression of BMP receptors inmature os-teoclasts. Bone 2000;27:479–486.43 Moerman EJ, Teng K, Lipschitz DA et al.

Aging activates adipogenic and suppresses os-teogenic programs in mesenchymal marrowstroma/stem cells: The role of PPAR-gamma2transcription factor and TGF-beta/BMP signal-ing pathways. Aging Cell 2004;3:379–389.

518 Perivascular Stem Cells in Bone Formation

STEM CELLS TRANSLATIONAL MEDICINE

Page 11: Edinburgh Research Explorer · bone chips with sodium hyaluronate, and it was chosen for its proven osteoinductive characteristics and clinical translation (human demineralized bone

44 Jeppsson C, Aspenberg P. BMP-2 caninhibit bone healing. Bone-chamber study inrabbits. Acta Orthop Scand 1996;67:589–592.45 Aghaloo T, Jiang X, Soo C et al. A study of

the role of Nell-1 gene modified goat bonemarrow stromal cells in promoting new boneformation. Mol Ther 2007;15:1872–1880.46 Takada I, Kouzmenko AP, Kato S. Wnt

and PPARgamma signaling in osteoblastogen-esis and adipogenesis. Nat Rev Rheumatol2009;5:442–447.

47 Takada I, Suzawa M, Matsumoto K et al.Suppression of PPAR transactivation switchescell fate of bone marrow stem cells from adi-pocytes into osteoblasts. Ann NY Acad Sci2007;1116:182–195.48 Li W, Zara JN, Siu RK et al. Nell-1 en-

hances bone regeneration in a rat critical-sizedfemoral segmental defect model. Plast Recon-str Surg 2011;127:580–587.49 Lu SS, Zhang X, Soo C et al. The osteoin-

ductive properties of Nell-1 in a rat spinal fu-sion model. Spine J 2007;7:50–60.

50 Siu RK, Lu SS, Li W et al. Nell-1 proteinpromotes bone formation in a sheep spinal fu-sion model. Tissue Eng Part A 2011;17:1123–1135.51 Park TS, Gavina M, Chen CW et al. Pla-

cental perivascular cells for human muscle re-generation. Stem Cells Dev 2011;20:451–463.52 Montemurro T, AndrioloG,Montelatici E

et al. Differentiation and migration propertiesof human foetal umbilical cord perivascularcells: Potential for lung repair. J Cell Mol Med2011;15:796–808.

See www.StemCellsTM.com for supporting information available online.

519James, Zara, Zhang et al.

www.StemCellsTM.com