angiogenic and osteogenic potential of bone repair cells

13
Angiogenic and Osteogenic Potential of Bone Repair Cells for Craniofacial Regeneration Darnell Kaigler, D.D.S., M.S., Ph.D., 1–3 Giorgio Pagni, D.D.S., 1 Chan-Ho Park, M.S., 1 Susan A. Tarle, B.S., 1 Ronnda L. Bartel, Ph.D., 4 and William V. Giannobile, D.D.S., DMSc 1–3 There has been increased interest in the therapeutic potential of bone marrow derived cells for tissue engineering applications. Bone repair cells (BRCs) represent a unique cell population generated via an ex vivo, closed-system, automated cell expansion process, to drive the propagation of highly osteogenic and angiogenic cells for bone engineering applications. The aims of this study were (1) to evaluate the in vitro osteogenic and angiogenic potential of BRCs, and (2) to evaluate the bone and vascular regenerative potential of BRCs in a craniofacial clinical application. BRCs were produced from bone marrow aspirates and their phenotypes and multipotent potential characterized. Flow cytometry demonstrated that BRCs were enriched for mesenchymal and vascular phenotypes. Alkaline phosphatase and von Kossa staining were performed to assess osteogenic differentiation, and reverse transcriptase–polymerase chain reaction was used to determine the expression levels of bone specific factors. Angiogenic differentiation was determined through in vitro formation of tube-like structures and fluo- rescent labeling of endothelial cells. Finally, 6 weeks after BRC transplantation into a human jawbone defect, a biopsy of the regenerated site revealed highly vascularized, mineralized bone tissue formation. Taken together, these data provide evidence for the multilineage and clinical potential of BRCs for craniofacial regeneration. Introduction T he demand for tissue replacements has led to the emergence and significant expansion of the field of tissue engineering. 1 Craniofacial regenerative medicine applications have demonstrated significant impacts for oral soft and hard tissue repair. 2,3 In preclinical model systems, multipotent cells derived from bone marrow have become a popular source of cells for regenerating bone, ligament, tendon, and cartilage. 4–8 More recently, autologous grafts utilizing various formula- tions of bone marrow or bone-marrow-derived cells have been investigated in clinical studies for skeletal bone repair 9,10 and craniofacial regeneration. 11–14 Although there has been modest success achieved in these approaches, major limita- tions still include crude isolation techniques and poorly de- fined cell preparations appropriate for grafting, inability to produce sufficient cell numbers for transplantation without multiple passages in traditional open tissue culture systems, and a lack of identification of an ideal cell type or cell pop- ulation for transplantation. Despite the specific limitations of currently defined cell isolation and preparation protocols, an overarching challenge common to all cell and tissue trans- plantation strategies is the inability to produce a supportive vasculature for graft incorporation and cell survival. It has been well-established that key to the development of bone tissue is not only the formation of bone but also the coordinated development of a supportive blood supply. 15 Thus, when employing cell transplantation strategies, not only does the osteoprogenitor cell type need to be consid- ered, but formation of a functional vasculature to support cell viability and maturation of the tissue warrants serious consideration as well. As a result, some strategies used to engineer and regenerate bone tissue employ cotransplanta- tion of osteoprogenitor cells with either hematopoietic or endothelial cells to help establish a supportive blood supply to the transplanted cells. 16–19 Although these approaches all hold great promise, it would be more desirable to transplant a cell population that contains cells capable of establishing both a blood supply and regenerating bone. An automated cell-manufacturing process has been de- veloped that utilizes a single-pass perfusion (SPP) process; this enables a clinical-scale expansion of autologous, pri- mary, human bone repair cells (BRCs) derived from bone marrow. In SPP, the culture medium is continuously re- placed with a fresh medium at a slow, controlled rate without the disturbance, removal, or passaging of cells. This technology results in significant expansion of primary human cells 20,21 and has previously demonstrated clinical Departments of 1 Periodontics and Oral Medicine and 2 Biomedical Engineering, University of Michigan, Ann Arbor, Michigan. 3 University of Michigan Center for Oral Health Research, Ann Arbor, Michigan. 4 Aastrom Biosciences, Inc., Ann Arbor, Michigan. TISSUE ENGINEERING: Part A Volume 16, Number 9, 2010 ª Mary Ann Liebert, Inc. DOI: 10.1089/ten.tea.2010.0079 2809

Upload: others

Post on 26-May-2022

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Angiogenic and Osteogenic Potential of Bone Repair Cells

Angiogenic and Osteogenic Potential of Bone Repair Cellsfor Craniofacial Regeneration

Darnell Kaigler, D.D.S., M.S., Ph.D.,1–3 Giorgio Pagni, D.D.S.,1 Chan-Ho Park, M.S.,1 Susan A. Tarle, B.S.,1

Ronnda L. Bartel, Ph.D.,4 and William V. Giannobile, D.D.S., DMSc1–3

There has been increased interest in the therapeutic potential of bone marrow derived cells for tissue engineeringapplications. Bone repair cells (BRCs) represent a unique cell population generated via an ex vivo, closed-system,automated cell expansion process, to drive the propagation of highly osteogenic and angiogenic cells for boneengineering applications. The aims of this study were (1) to evaluate the in vitro osteogenic and angiogenicpotential of BRCs, and (2) to evaluate the bone and vascular regenerative potential of BRCs in a craniofacialclinical application. BRCs were produced from bone marrow aspirates and their phenotypes and multipotentpotential characterized. Flow cytometry demonstrated that BRCs were enriched for mesenchymal and vascularphenotypes. Alkaline phosphatase and von Kossa staining were performed to assess osteogenic differentiation,and reverse transcriptase–polymerase chain reaction was used to determine the expression levels of bone specificfactors. Angiogenic differentiation was determined through in vitro formation of tube-like structures and fluo-rescent labeling of endothelial cells. Finally, 6 weeks after BRC transplantation into a human jawbone defect, abiopsy of the regenerated site revealed highly vascularized, mineralized bone tissue formation. Taken together,these data provide evidence for the multilineage and clinical potential of BRCs for craniofacial regeneration.

Introduction

The demand for tissue replacements has led to theemergence and significant expansion of the field of tissue

engineering.1 Craniofacial regenerative medicine applicationshave demonstrated significant impacts for oral soft and hardtissue repair.2,3 In preclinical model systems, multipotent cellsderived from bone marrow have become a popular source ofcells for regenerating bone, ligament, tendon, and cartilage.4–8

More recently, autologous grafts utilizing various formula-tions of bone marrow or bone-marrow-derived cells havebeen investigated in clinical studies for skeletal bone repair9,10

and craniofacial regeneration.11–14 Although there has beenmodest success achieved in these approaches, major limita-tions still include crude isolation techniques and poorly de-fined cell preparations appropriate for grafting, inability toproduce sufficient cell numbers for transplantation withoutmultiple passages in traditional open tissue culture systems,and a lack of identification of an ideal cell type or cell pop-ulation for transplantation. Despite the specific limitations ofcurrently defined cell isolation and preparation protocols, anoverarching challenge common to all cell and tissue trans-plantation strategies is the inability to produce a supportivevasculature for graft incorporation and cell survival.

It has been well-established that key to the development ofbone tissue is not only the formation of bone but also thecoordinated development of a supportive blood supply.15

Thus, when employing cell transplantation strategies, notonly does the osteoprogenitor cell type need to be consid-ered, but formation of a functional vasculature to supportcell viability and maturation of the tissue warrants seriousconsideration as well. As a result, some strategies used toengineer and regenerate bone tissue employ cotransplanta-tion of osteoprogenitor cells with either hematopoietic orendothelial cells to help establish a supportive blood supplyto the transplanted cells.16–19 Although these approaches allhold great promise, it would be more desirable to transplanta cell population that contains cells capable of establishingboth a blood supply and regenerating bone.

An automated cell-manufacturing process has been de-veloped that utilizes a single-pass perfusion (SPP) process;this enables a clinical-scale expansion of autologous, pri-mary, human bone repair cells (BRCs) derived from bonemarrow. In SPP, the culture medium is continuously re-placed with a fresh medium at a slow, controlled ratewithout the disturbance, removal, or passaging of cells. Thistechnology results in significant expansion of primaryhuman cells20,21 and has previously demonstrated clinical

Departments of 1Periodontics and Oral Medicine and 2Biomedical Engineering, University of Michigan, Ann Arbor, Michigan.3University of Michigan Center for Oral Health Research, Ann Arbor, Michigan.4Aastrom Biosciences, Inc., Ann Arbor, Michigan.

TISSUE ENGINEERING: Part AVolume 16, Number 9, 2010ª Mary Ann Liebert, Inc.DOI: 10.1089/ten.tea.2010.0079

2809

Page 2: Angiogenic and Osteogenic Potential of Bone Repair Cells

success in the expansion of bone marrow and blood cells forreplenishment of hematopoietic cells after treatment of var-ious blood dyscrasias.22–25 Additionally, results of recentin vitro and in vivo studies have generated interest in usingthis process to produce cells for bone tissue regeneration.26

The hypothesis underlying the current study is that BRCshave osteogenic and angiogenic potential that could manifestin their ability to regenerate bone and vascular tissue in aclinical craniofacial application. To address this hypothesis, weaimed to first examine the multipotency of BRCs in vitro, withparticular emphasis on elucidation of their angiogenic andosteogenic capacities. Second, we sought to determine if BRCscould form bone and accelerate osteogenesis clinically whenimplanted within an osseous defect in the jaw.

Materials and Methods

Bone marrow harvest and BRC production

The production of BRCs has been previously described.26

Briefly, after U.S. Food and Drug Administration (FDA) andUniversity of Michigan Institutional Review Board approval,10 healthy subjects giving informed consent underwent abone marrow aspiration of the posterior ilium under con-scious sedation and local anesthetic. Of these 10 subjects,where indicated, subsets of them (marrow) were used forphenotypic characterization of cells. Collected marrow wastransferred to a sterile blood bag and bone marrow mono-nuclear cells (BMMNCs) were purified by Ficoll densitygradient centrifugation. BMMNCs were then inoculated intoa bioreactor, which is a proprietary computer-controlled,automated cell-processing unit, the Aastrom Replicell� Sys-tem (Aastrom Biosciences). The Cell Cassette within thissystem provides a functionally closed, sterile environment inwhich cell production occurs. The fluid pathway in the CellCassette includes the cell growth chamber, a medium supplycontainer, a mechanism for medium delivery, a waste me-dium collection container, and a container for the collectionof harvested cells. All components are interconnected withsterile barrier elements throughout to protect the culturefrom contamination during use. The culture medium consistsof Iscove’s modified Dulbecco’s medium, 10% fetal bovineserum, 10% horse serum, and 5 mM hydrocortisone. Thissystem incorporates SPP in which a fresh medium flowsslowly over cells without retention of waste metabolites ordifferentiating cytokines.20 In-process safety test (absence ofbacterial and fungal contaminants and endotoxins) wasconducted on an sampled effluent medium at 48 h beforeharvest. After cultivation for 12 days at 378C and 5% CO2,the BRC product was harvested by trypsinization, washed ina physiologic buffer, and collected into a sterile bag and re-suspended in Isolyte and 0.5% human serum albumin, whereit was stored until the time of transplantation (Fig. 1).Sterility, endotoxin, and mycoplasma testing were all con-ducted on the BRCs.

Cell culture

The Replicell System is an automated one-cycle systemthat requires the input to be derived from fresh bone marrowaspirates; as a result, once BRCs are removed from the sys-tem, it is not possible to regrow or resume culture of cellswithin this same system. Thus, further culture of BRCs for

in vitro assays required conventional tissue culturing tech-niques in both experimental and control conditions. ExcessBRC product not utilized for transplantation was cultured ina medium consistent with culture of bone-marrow-derivedstem cells,27 consisting of minimum essential alpha medium(aMEM) (Gibco–Invitrogen #12571) with 15% fetal bovineserum (Gibco-Invitrogen-16000), 100mM ascorbic acid 2phosphate (Sigma A-8960), and 5mg/mL Gentamicin (In-vitrogen #15750060), and grown in a 378C humidified tissueculture incubator under 5% CO2. This medium was used forcontrol conditions (without incorporation of differentiation/inductive factors) in the in vitro assays and it should also benoted that this open culture system and media used weredistinctly different from those used to produce the BRCproduct. Cell culture media were changed every 2 or 3 days.Cells were grown in T-150 flasks to *80% confluence, fol-lowed by cell washing with phosphate-buffered saline (PBS)and trypsinization before being split into 12-well plates or60 mm tissue culture dishes for differentiation assays.

Flow cytometry

Phenotypic comparison of cell surface marker expressionwas made between the starting BMMNCs and the final BRCproduct from a subset of four subjects who had marrowharvested for generation of BRCs. Flow cytometry was per-formed on the same day of isolation of the BMMNCsand immediately after production of BRCs (i.e., neitherBMMNCs nor BRCs were cultured, as described in the CellCulture section, before performance of the flow cytometry).BMMNCs or BRCs were washed and resuspended in1�Dulbecco’s PBS (Gibco) containing 1% bovine serum albu-min. Samples were Fc receptor blocked with normal mouseIgG for approximately 10 min. Tubes containing 0.5�106 cellsin approximately 0.1 mL were then stained at 28C–88C withphycoerythrin (PE) or PE cyanin 5-conjugated anti-CD90(Thy1) antibodies, PE-conjugated anti-CD11b, anti-Gly-A,anti-CD34, anti-AC133, anti-CD19, anti-vascular endothelial

FIG. 1. Bone repair cell (BRC) production. After harvest ofbone marrow aspirates, cells are cultured using an auto-mated, closed-system, single-pass perfusion (SPP) process.After 12 days of cell expansion in this bioreactor system,cells are packaged and delivered to the bone regenerative siteon a biodegradable sponge. Color images available online atwww.liebertonline.com/ten.

2810 KAIGLER ET AL.

Page 3: Angiogenic and Osteogenic Potential of Bone Repair Cells

growth factor (VEGF)R1, anti-Tie2, anti-CD145, fluouresceinisothyocyanate-conjugated anti-CD66b, anti-CD14, anti-CD45, anti-CD3, anti-CD144 antibodies, and PE cyanin5-conjugated anti-CXCR4 (CD184) (Beckman Coulter). After15 min, cells were washed and resuspended in 0.5 mL PBS/bovine serum albumin for analysis on the Epics XL-MCL(Beckman Coulter) or FC500 flow cytometer.

In vitro multilineage differentiation

Multipotency of BRC cultures was determined by in vitrodifferentiation assays of osteogenesis, chondrogenesis, andadipogenesis. BRCs were plated at a density of 30,000 cellsper well in 12-well plates. At >90% confluency, cells wereinduced, according to previously described protocols,28 withosteogenic (BRC medium including 5 mM b-glycerol phos-phate, 100 nM dexamethasone, and 50mM ascorbic acid2-phosphate), chondrogenic (BRC medium including 50mMascorbic acid 2-phosphate, 100 nM dexamethasone, 5 mg/mLhuman insulin, 1 ng/mL transforming growth factor-b, 400mMproline, and 1� Nonessential amino acids), or adipogenic(BRC medium including 0.5 mM isobutylmethylxanthine,1 mM dexamethasone, 10 mg/mL human insulin, and 200mMindomethacin) medium. BRCs were grown at 378C in a hu-midified 5% CO2 incubator. The medium was changed every2–3 days. After 2 weeks, the cells were fixed and stained asoutlined below.

Mesenchymal differentiation staining

To detect chondrogenic differentiation, induced BRCswere fixed in cold 100% methanol for 30 min and then ex-posed to 1% Alcian blue in 0.1 N HCl for 30 min. Cells werewashed twice with 0.1 N HCl. Cells in PBS were viewed orstored at 48C.

To detect mineralized matrix formation indicative ofosteogenic differentiation, induced BRCs were fixed in 4%paraformaldehyde for 30 min, immersed in fresh 5% silvernitrate, and incubated in the dark for 30 min. After washingin water, the BRCs were exposed to ultraviolet light for30 min followed by a 4-min incubation in 1% sodium thio-sulfate to neutralize the silver nitrate. Cells were washed andstored in PBS at 48C.

To detect adipogenic differentiation by identifying lipidvesicles, induced BRCs were fixed in 4% paraformaldehydefor 30 min and then immersed in 0.3% Oil Red O solution (inisopropanol) for 30 min. Cells were washed and stored inPBS at 48C.

Alkaline phosphatase activity and detection

Early osteogenic differentiation was detected and quanti-fied by alkaline phosphatase (ALP) staining and ALP en-zyme assays. ALP activity was confirmed with reversetranscriptase–polymerase chain reaction (PCR). Cells wereplated at a density of 30,000 cells per well in 12-well plates.At 80% confluence, cells were induced with the osteogenicmedium as described above. The medium was changedevery 2–3 days and after 1 week, and ALP activity wasmeasured.

To detect ALP activity, the BRCs were fixed in 70% etha-nol for 30 min. They were then incubated with freshly madesubstrate containing naphthol AS-TR phosphate (Sigma) andFast blue for 30 min. Cells were washed twice with PBS andthen viewed or stored at 48C.

To quantify the ALP activity and normalize the results,BRCs were lysed in Passive Lysis Buffer (Promega) accord-ing to manufacturer’s instructions. Cell lysates were thensonicated, and centrifuged (10,000 rpm for 10 min at 48C).The supernatant was recovered for the quantitative color-metric ALP assay,29 and the cell pellet was used for DNAisolation and the determination of the DNA concentrationusing the Quant-iT� dsDNA BR Assay (Invitrogen) per themanufacturer’s instructions.

Reverse transcriptase–PCR

To further confirm osteogenic differentiation, after 1 weekof osteogenic induction total BRC cellular RNA was ex-tracted, reverse transcribed, and amplified using osteoblast-specific gene primers: Runx2, osteocalcin (OCN), and bonesialoprotein (BSP). Media from the wells of induced BRCswere aspirated. Cells were immediately resuspended in 1 mLof Trizol (Invitrogen) and RNA was isolated according to themanufacturer’s instructions. cDNA was synthesized usingInvitrogen’s SuperScriptII kit and oligo dT. PCR reactioncomponents and concentrations were as described in theInvitrogen Platinum Taq polymerase instructions using theprimer sets below. An MJ themorcycler (MJ Resarch ModelPTC-200) was used for the two PCR reaction conditions(Table 1).

Angiogenic potential

The angiogenic potential of BRCs was investigatedthrough the fluorescent labeling of endothelial cells andcapillary tube formation in culture. Collagen culture disheswere prepared by dispensing 2.5 mL of an ice-cold collagen

Table 1. Polymerase Chain Reaction Primer Pairs

Primer name Primer sequence Product size (bp) Accession number

BSP BSP FWDb CTATGGAGAGGACGCCACGCCTGG 586 NM_004967BSP REVb CATAGCCATCGTAGCCTTGTCCTGAPDH FWDa AGCCGCATCTTCTTTTGCGTC 815 NM_002046GAPDH REVa TCATATTTGGCAGGTTTTTCTOCN FWDa CATGAGAGCCCTCACA 314 NM_199173OCN REVa AGAGCGACACCCTAGACRunx2 FWDb CCCGTGGCCTTCAAGGT 76 NM_004348Runx2 REVb CGTTACCCGCCATGACAGTA

a948C 2 min [948C 45@ 568C 45@ 728C 10]�35 cycles 728C 150.b948C 2 min [948C 45@ 678C 45@ 728C 10]�35 cycles 728C 150.

BONE REPAIR CELLS FOR CRANIOFACIAL REGENERATION 2811

Page 4: Angiogenic and Osteogenic Potential of Bone Repair Cells

solution, containing 2.4 mg/mL PureCol� bovine collagen(Advanced BioMatrix) in PBS at pH 7.4 into 60 mm plates.Collagen plates were solidified at 378C for 90 min and thenequilibrated in the growth medium for 2 h. BRC cultureswere plated with 200,000 BRCs and incubated overnight at378C. One day after plating the cells, the medium waschanged with a medium containing 50 ng/mL VEGF (R&D#293-VE). The medium was changed every 2–3 days. After1 week, cells were fed the medium containing 10 mg/mLfluorescently labeled lipoprotein (DiI-Ac-LDL; BiomedicalTechnologies) for 4 h at 378C to specifically label the endo-thelial cells. The medium was aspirated and the cells werewashed three times with PBS before being fixed in 4%paraformaldehyde for 60 min. After fixing, the cells werewashed three times with water before 3 mL of PBS was ad-ded to each plate, and then viewed or stored at 48C.

To measure angiogenic cytokine production, BRCs from asubset of eight subjects were centrifuged to pellet the cells,and resuspended in X-Vivo 15 (Lonza) medium. X-Vivo 15 isa serum-free medium that was used in these assays toeliminate potential serum interference and nonspecificbinding with some of the reagents (i.e., lack of human spe-cific antibodies) used in the assays. All fractions werequantified and diluted to 1�106 mL�1 in X-VIVO 15 me-dium. The cells (100mL) were added to the wells of a 96-wellround-bottom plate in triplicate. The culture mediumalone or supplemented with 300 ng/mL endotoxin lipo-polysaccharide was added in a volume of 50 mL to all wells.Twent-four hours later, 100 mL of supernatant from each wellwas collected and stored at �708C until analysis. HumanCytokine/Chemokine LINCOplex kits (Millipore Corpora-tion) were used for multiplex analysis of VEGF and IL-8concentrations in BRC supernatant fluids. The kits were usedfor cytokine determination as defined by the manufacturer’sprotocol (HCYTO-60K-Rev. 12/07/05) located at the fol-lowing link:

www.millipore.com/userguides.nsf/a73664f9f981af8c852569b9005b4eee/09dc59783f6430ab85257259004f0203/$FILE/hcyto-60k.pdf.

ELISA kits (R&D Systems) were used to quantify the in-dividual analytes Tie-1 and Tie-2 according to the manu-facturer’s protocol.

Surgical transplantation of BRCsto a localized craniofacial osseous defect

A nonrestorable tooth was extracted under local anes-thesia from a patient in need of tooth extraction due todental decay and periodontal disease associated with thetooth. Due to the preexisting condition of the tooth, itsremoval resulted in an area of a localized bone defect.According to a protocol approved by the U.S. FDA and theUniversity of Michigan’s Institutional Review Board, afull-thickness mucoperiosteal flap was elevated for accessand hemostasis of the surgical defect achieved. One mil-liliter of the BRC cell suspension (approximately 1.5�107

cells/mL) was absorbed onto a gelatin sponge (Gelfoam;Pfizer) sized to a dimension of *2 cm3. The sponge was thentransplanted into the extraction site to the level of the in-terproximal bone. The flap was then coronally repositioned,and a bioabsorbable collagen barrier membrane (Biomend�;Zimmer Dental) was placed over the sponge to contain the

cell construct. The tissues were approximated and closedwith a 4-0 bioabsorbable suture material. The subject wasprescribed 500 mg amoxicillin, three times a day (t.i.d.) for 7days, decreasing doses of dexamethasone (8, 4, 2, 2 mg) for4 days, and 600 mg Ibuprofen four times a day (q.i.d.) for3 days. Surgical reentry of the treated osseous defect wasperformed 6 weeks postsurgery, and a bone core of 2�7 mmwas harvested with a trephine drill (Ace Surgical Supply Co.,Inc.). To ensure harvesting of bone from the area where theBRCs were grafted, pre- and postsurgical measurementtemplates were used to identify the area corresponding tothe center of the previous extraction socket. The core wasprepared for microcomputed tomography (mCT) imagingand descriptive histological evaluation.

mCT and histology

The nondecalcified core was captured with the scanningdirection parallel to the longitudinal axis of the core speci-men. High-resolution scanning with an in-plane pixel sizeand slice thickness of 24mm was performed. To cover theentire thickness of the bone core biopsy, the number of sliceswas set at 400. GEMS MicroView� software was used tomake three-dimensional (3D) reconstructions from the set ofscans as previously described.30

After scanning, the core was decalcified for 2 weeks in10% ethylenediaminetetraacetic acid. After decalcification,the core was transferred to 70% ethanol stored at 48C untilready for embedding in paraffin. Standard hematoxylin andeosin staining was performed to assess bone morphologyand blood vessel formation.

Statistical analysis

Statistical analysis was performed with the use of Instatsoftware (GraphPad Software). All data were plotted asmean� standard error of the mean, unless otherwise noted.Statistically significant differences were determined by two-tailed Student’s t-tests, and statistical significance was de-fined as p< 0.05.

Results

Phenotypic characterization of BRC population

For all BRC formulations produced after cell processing,flow cytometry was performed to characterize the phenotypeof the BRCs, and in some instances, the initial BMMNCswere also assessed before production of BRCs. For 10 sub-jects from whom BRCs were produced, including the subjecttreated with BRCs in this study, cell surface marker expres-sion of the final BRC product is shown (Table 2). There arehigh percentages of CD31þ, CD90þ (Thy1), and CD105þcells. When compared to cell surface marker expression fromtypical BMMNC fractions used to produce these cells (un-published proprietary data), CD90þ (Thy1) and CD105þ arehighly enriched in the BRC product.

Mesenchymal potential of BRCs

BRCs were assessed for their capacity to differentiate to-ward different cellular lineages after culture under adi-pogenic, chondrogenic, and osteogenic conditions. After 2weeks of culture, BRCs were subjected to Oil Red O, Alcian

2812 KAIGLER ET AL.

Page 5: Angiogenic and Osteogenic Potential of Bone Repair Cells

blue, and von Kossa staining. In control culture conditions,Oil Red O, Alcian blue, and von Kossa staining were allnegative. In adipogenic conditions, Oil Red O staining wasused to detect intracellular lipid-rich vacuoles and morpho-logical changes in cell shape. The results confirmed that cellswere differentiated toward an adipogenic lineage (Fig. 2a).Similarly, in cells grown under chondrogenic conditions, thepresence of chondrogenic proteoglycans was indicated bypositive Alcian blue staining (Fig. 2a), confirming chondro-genic differentiation. Under osteogenic culture conditions,deposition of mineralized matrix indicative of osteoblastswas evident through positive von Kossa staining (Fig. 2a). Tofurther evaluate osteogenic differentiation, alkaline phopha-tase activity was also analyzed qualitatively and quantita-tively after 1 week of culture in osteogenic conditions (Fig.2b). With ALP staining, there was a significantly more robustand widespread degree of staining in the osteogenic condi-tions relative to the control culture conditions. When thisdifference was measured quantitatively, there was a signifi-cant threefold increase ( p< 0.05) in ALP activity in osteo-genic relative to control conditions. Commitment toward anosteogenic lineage was further confirmed through gene ex-pression of Runx2, OCN, and BSP (Fig. 2c), and althoughRunx2 was expressed at a lower level in the control condition(data not shown), OCN and BSP were not detectable incontrol conditions at this timepoint.

Angiogenic potential of BRCs

In addition to standard phenotypic marker characteriza-tion of the final BRC product (shown from 10 patients treatedwith BRCs in Table 2), further analysis was performed onBRC products produced from smaller subsets of healthyvolunteer subjects who provided donor marrow specificallyto evaluate marker expression of angiogenic phenotypes.First, phenotypic comparison of cell surface marker expres-sion was made between the starting BMMNCs and the finalBRC product from a subset of four subjects. After expansion

of the BMMNCs using the BRC process, the enrichment ofvascular phenotypes in BRCs is shown in Table 3. Cell sur-face expression of vascular phenotypes associated with en-dothelial cells (Angiopoietin and flt1) and pericytes (Thy1þand MUC-18) are all highly enriched. Next, when BRCs froma subset of eight volunteer donors (from the 10) were cul-tured in a medium (X-VIVO) without addition of VEGF, cellswere shown to produce appreciable levels of potent angio-genic cytokines VEGF, and angiopoietin (Ang)-1 and -2(Fig. 3a). It should be noted that none of these cytokines weredetected in the supernatant from the initial BMMNCs used toproduce BRCs. Another angiogenic cytokine, interleukin-8(Il-8), was produced by BRCs at a concentration (7.5 ng/mL/100,000 cells) significantly higher than that in the initialBMMNCs (4.6 ng/mL/100,000).

To examine the differentiation of BRCs into endothelialand vascular cells, BRCs were cultured in 3D collagen gels inthe presence of VEGF. In these assays, VEGF is typicallyincorporated to induce endothelial cell differentiation.31–33

Over the course of 1 week, cells elongated and formedspindle-like interconnecting structures resembling those seenby endothelial cells forming capillary tubes when grownunder similar culture conditions (i.e., 3D extracellular ma-trices) (Fig. 3b). To further confirm the differentiation ofBRCs into endothelial cells, a fluorescently labeled lipopro-tein (DiI-Ac-LDL) that is metabolized specifically by endo-thelial cells was added to BRC 3D cultures. After 4 h ofincubation with DiI-Ac-LDL, cells were examined underfluorescence microscopy and demonstrated uptake of thefluorescently labeled dye (Fig. 3c, d), characteristically seenby endothelial cells.34 Taken together, these data demon-strate that BRCs have the capacity to differentiate into en-dothelial cells in 3D extracellular matrices and producesoluble angiogenic factors.

Clinical osteogenic and vascular regenerativepotential of BRCs

The ultimate test of the osteogenic and angiogenic po-tential of BRCs is their ability to regenerate bone and vas-cular structures in a clinical model. To address this clinicalsituation, we grafted BRCs into an osseous defect of thejawbone. After tooth extraction, a bone defect resulted in thearea that the tooth previously occupied. The BRCs (in liquidsuspension) were carefully placed (Fig. 4a) onto a resorbablematerial matrix (gelatin sponge) to the point of saturation(Fig. 4b). Cells were allowed 15 min to adhere to the gelatinsponge before their placement into the recipient graft site.After the extent of the bone defect (created as a result of toothremoval) was assessed (Fig. 4c), the sponge matrix contain-ing the BRCs was grafted into the bone defect (Fig. 4d), acollagen membrane was placed for containment of the graft(Fig. 4e, f ), and the area then closed with sutures and al-lowed to heal for 6 weeks.

After 6 weeks of healing, the grafted defect site was re-entered for examination of regenerated tissue and dentalimplant placement. Clinical examination revealed the ap-pearance of bone tissue, with no evidence of the previousbone defect (Fig. 5a), and a core biopsy was harvested fromthe center of the area in which the BRCs were grafted. Uponretrieval of the specimen with the trephine drill, it was foundthat the tissue was very dense, indicative of mature bone

Table 2. Frequency of Cell Phenotypes in Bone

Repair Cells (n¼ 10) Produced by Single-Pass

Perfusion (Mean� Standard Deviation)

Phenotype BRC SD

% Viable (% 7AAD�) 92.69 2.16% CD11bþ 61.17 5.70% CD66bþ 28.20 7.50% Lin-CD34þ 0.61 0.35% CD133þ 0.38 0.13% VEGFR2 6.15 2.39% CD14þAuto� 10.57 2.57% CD14þAutoþ 16.81 6.89% CD45þ 79.24 3.39% CD3þ 14.24 5.41% CD19þ 0.81 0.47% CD31þ 65.61 5.69% CD90 (Thy-1)þ 17.00 2.85% CD105þ 20.20 1.85% CD105þCD90þ 15.79 2.98% CD90þ s that are 105þ 94.44 3.05

BRC, bone repair cell; SD, standard deviation; VEGF, vascularendothelial growth factor.

BONE REPAIR CELLS FOR CRANIOFACIAL REGENERATION 2813

Page 6: Angiogenic and Osteogenic Potential of Bone Repair Cells

FIG. 2. Multipotent and osteogenic potential of BRCs. (a) After induction of BRCs in osteogenic, chondrogenic, and adi-pogenic media, photographs and corresponding photomicrographs show multipotent mesenchymal differentiation asmeasured by phenotypic expression of osteogenic mineralized matrix (von Kossa), chondrogenic proteoglycans (Alcian blue),and adipogenic lipid vacuoles (Oil Red O); low magnification images were taken at 40�; high magnification images areshown at 200�. (b) Osteogenic induction potential of BRCs is further evaluated qualitatively (staining) and (c) Quantitativelythrough measurement of alkaline phosphatase (ALP) activity of cells grown in the control medium versus cells grown in theosteogenic medium (*p< 0.05; alkaline phosphatase assays were performed in triplicate, and high-magnification non-osteogenic and osteogenic panels shown on the ends represent the same wells shown at lower magnification in middle panel);low magnification images were taken at 40�; high magnification images are shown at 200�. (d) BRC expression of bone-specific transcripts (Runx2, osteocalcin [OCN], and bone sialoprotein [BSP]) was also measured with reverse transcriptase–polymerase chain reaction. Color images available online at www.liebertonline.com/ten.

2814 KAIGLER ET AL.

Page 7: Angiogenic and Osteogenic Potential of Bone Repair Cells

tissue (Fig. 5b). The clinical appearance of the biopsy speci-men clearly showed a highly vascular tissue that, consistentwith retrieval of viable bone tissue, produced bleeding afterharvest (Fig. 5c). mCT analysis was performed on the biop-sied tissue, and 3D reconstruction of the specimen showed

highly mineralized tissue throughout the entire length of thecore (Fig. 5d). After this analysis, histology was performedon the specimen and hematoxylin and eosin staining clearlyshowed widespread distribution of mature bone tissuewith an abundance of blood vessels distributed throughout

Table 3. Presence and Enrichment of Vascular Phenotypes in Bone Marrow

Mononuclear Fraction and Bone Repair Cell Product

Phenotype Common name Cell type recognized BMMNC (mean� SD) BRC (mean� SD) Fold enrichment

CD34þ — HSCs/ECs progenitors 3.5þ 1.2 0.6þ 0.3 <1CD133þ AC133 HSCs/ECs progenitors 1.2þ 0.2 0.5þ 0.2 <1CD90þ Thy1 SSCs/pericytes 0.3þ 0.2 28.2þ 10.7 107.5CD202bþ Tie2/Angiopoietin receptor HSCs/ECs 7.3þ 4.8 35.7þ 6.3 4.9— VEGFR1/flt 1 Hematopoietic 6.5þ 2.0 38.9þ 4.7 6.0CD144þ VE-Cadherin Vascular ECs 2.5þ 2.4 0.9þ 0.6 <1CD146þ MUC18, S-endo ECs/pericytes 1.2þ 0.5 27.1þ 8.5 23.4

Values represent means taken from four different subjects. BMMNC, bone marrow mononuclear cell; HSCs, hematopoietic stem cells; ECs,endothelial cells; SSCs, skeletal (mesenchymal) stem cells.

FIG. 3. In vitro angiogenic phenotype of BRCs. (a) BRCs (n¼ 8) were shown to produce clinically appreciable levels of potentangiogenic cytokines, vascular endothelial growth factor (VEGF), and angiopoietin (Ang)-1 and -2. (b) After BRCs were culturedover 5 days in the presence of 50 ng/mL VEGF within three-dimensional collagen extracellular matrices, photomicrographs weretaken and show sprouting structures indicative of endothelial cell capillary tube formation (100�magnification). (c) 100� and (d)200� photomicrographs of fluorescently labeled BRCs after 4-h incubation with a (w/a) fluorescent-labeled lipoprotein (DiI-Ac-LDL) metabolized specifically by endothelial cells. Color images available online at www.liebertonline.com/ten.

BONE REPAIR CELLS FOR CRANIOFACIAL REGENERATION 2815

Page 8: Angiogenic and Osteogenic Potential of Bone Repair Cells

(Fig. 5e). These results provide preliminary evidence thatBRCs, produced from BMMNCs, have the regenerative ca-pacity to produce highly vascular bone tissue in a humancraniofacial bone defect.

Discussion

Cell transplantation of stem cells has tremendous potentialfor craniofacial regenerative applications; yet, identification

of the appropriate cell types and cell processing protocols aretwo of the most critical determinants in producing successfuloutcomes. In the present study, our aim was to assess thecapacity of a cell production process, which utilizes an au-tomated closed-system bioreactor, to produce clinical-scalenumbers of autologous cells (BRCs) capable of regeneratingclinically viable bone. Through cell surface marker charac-terization of BRCs, it was determined that the SPP ex vivo cellexpansion processing of bone marrow aspirate was capable

FIG. 4. Grafting of BRCs into localized jawbone defects. (a) Initial placement of BRCs onto gelatin sponge and (b) saturationof sponge with BRCs just before implantation into jawbone defect. Photographs of (c) jawbone defect after tooth removal, (d)placement of BRC-loaded gelatin into jawbone defect (arrows point to cell loaded gelatin), and (e, f) placement of a protectivecollagen membrane (arrows) for graft containment. Color images available online at www.liebertonline.com/ten.

2816 KAIGLER ET AL.

Page 9: Angiogenic and Osteogenic Potential of Bone Repair Cells

FIG. 5. BRC regeneration of highly vascular bone in jawbone defect. Photomicrographs of (a) regenerated jawbone defect6 weeks after grafting of BRCs, (b) harvesting of bone core biopsy using trephine bur (just before placement of dental implant), and(c) bone core biopsy. (d) Microcomputed tomography three-dimensional reconstruction of bone core biopsy from BRC-regeneratedbone demonstrating mineralized tissue formation throughout specimen. (e) Histological evaluation (hematoxylin and eosin staining)of bone formation showing areas of mature cortical bone (B) with high vascularity, as indicated by abundance of blood vessels(arrows; low magnification at 4� and high magnification at 200�). Color images available online at www.liebertonline.com/ten.

2817

Page 10: Angiogenic and Osteogenic Potential of Bone Repair Cells

of producing cell populations highly enriched for mesen-chymal and vascular progenitor cells. This was confirmedthrough cell characterization of BRCs, where cells within thisheterogeneous population demonstrated the capacity to beinduced to differentiate down chondrogenic, adipogenic,osteogenic, and angiogenic lineages. Finally, the test of theclinical regenerative capacity of BRCs demonstrated theirability to regenerate highly vascular bone in a human jaw-bone defect.

The cell-processing system employed to produce thesecells utilizes an SPP protocol.20,22,25,35 Gastens et al.35 exam-ined this system and its ability to expand bone-marrow-derived cells to produce clinical-scale cell numbers. Theseinitial studies compared cell phenotypes of cells producedwith this closed SPP system to phenotypes of cells culturedwith an open system utilizing conventional tissue cultureparameters. Although mesenchymal cell numbers were ob-served to be higher in cells cultured with the SPP closedsystem, the limitation of the study was that the initialBMMNC fractions tested in the two different systems camefrom different donors; thus, differences observed betweenthe final cell products could have been, at least in part, at-tributed to donor–donor variability. In the present study,although we did not compare cell populations from closedand open culture systems, we did compare BRCs directly tothe phenotypes of the BMMNCs from which they wereproduced. The final BRC population showed marked en-richment for mesenchymal and vascular cell phenotypes,suggesting that for therapeutic regenerative strategies, theSPP process supports the production of a more favorable cellpopulation for transplantation than protocols using trans-plants comprised of unfractioned, whole bone marrow. Al-though there is often a wide degree of variability in thephenotypic expression of these markers from donor to donor,the relative differences in cell phenotype before and aftercell processing is consistent in that the final product is en-riched in expression of mesenchymal and angiogenic phe-notypes after cell processing with the SPP system. Additionalstudies need to be performed comparing cell phenotypes anddifferentiation potential of cells derived from the samedonor, when processed with either this closed system or atraditional tissue culture open system technique. It is alsoimportant to note that although the objectives of the differ-entiation assays were to examine the osteogenic and angio-genic potential of the BRCs, it is not possible to regrow orresume culture of cells in the Replicell system once they havebeen removed from the system. As such, further culture ofBRCs required conventional tissue culturing techniques.However, it is recognized that this additional culture stepcould have potentially resulted in a cell population notidentical to the population produced from the Replicell sys-tem; yet, because the Replicell system is a closed system,there is not a viable alternative to performing or adapting theaforementioned assays to cells cultured while in the Replicellsystem.

The ability of the SPP process to form bone-forming cellshas also been recently studied in vitro and in ectopic animalmodels.26 In these studies, the levels of bone formation seenin vivo followed the same trends of the osteogenic differen-tiation observed in vitro. Additionally, cell surface markers,including CD90þ (Thy1) and CD105þ (endoglin), werepositively correlated with ectopic bone formation in mice.

CD105, originally identified as a marker of mesenchymalstem cells,36 has more recently been associated with vascularendothelium in angiogenic tissues37 and expression of CD90has been linked to bone marrow subpopulations of colony-forming mesenchymal stem cells (CFU-F, colony-formingunit–fibroblasts).38 In our study, cell surface marker expres-sion of BRCs from the subject who underwent cell trans-plantation in the bone defect showed 65-fold and 5-foldincreases in CD90þ and CD105þ, respectively (data notshown). This served as an indication that the BRC productwas highly enriched with cells possessing mesenchymal andangiogenic potential and is in accordance with previous re-ports demonstrating enrichment of theses cell types with thiscell-processing protocol.26,35

Although the cell product is highly enriched for vascularand mesenchymal cells as indicated by cell surface markerexpression and in vitro differentiation capacity, it is clear thatin vitro osteogenic differentiation of cell populations does notguarantee bone-forming capacity in vivo. Meijer et al. per-formed a clinical study evaluating the repair of jawbonedefects in six subjects treated with autologous cells expandedusing an open system,39 similar to the protocol used in tra-ditional tissue culture of mesencyhmal stem cells.40,41 In thisstudy, cells were cultured anywhere from 12 to 25 days be-fore implantation and the last 7 days in culture cells weregrown on a mineral substrate, hydroxyapatite (HA) particles,in the presence of the osteoinductive agent dexamethasone.When cells grown under these osteogenic conditions wereanalyzed for their osteogenic capacity, all six bone marrowspecimens produced cells capable of osteogenic differentia-tion in vitro (as determined by ALP expression) and boneformation in vivo (subcutaneous implants of HA/cell con-structs in athymic mice). However, after implantation ofthese HA/cell constructs into various human jawbone de-fects of the six patients, biopsy specimens taken at 4 monthsshowed that bone formation by implanted cells was able toregenerate bone in only one of the six patients treated. Theauthors made the important observation that in vitro osteo-genic assays and bone formation in preclinical mouse modelsmay not necessarily correlate to successful bone regenerationin the more challenging clinical applications. They concludedfurther that inadequate vascularity could have resulted in thereported disappointing outcome of the study.39 In accor-dance with the authors’ conclusion from this study, it is ourbelief that not only is vascularization from the host envi-ronment essential to clinical bone regeneration, but evenfurther, that the angiogenic potential of the transplanted cellsthemselves should play an active role in this vascularizationprocess.

The BRCs used to treat the human jawbone defect in ourstudy were not produced in the presence of any osteogenicfactors, and a gelatin sponge (with no known osteoinductiveor osteoconductive properties) was used as a carrier matrixto transplant the cells, as opposed to a mineralized matrixsuch as HA. Additionally, the biopsy specimen harvested at6 weeks showed significant new bone formation containingabundant blood vessels. Although no direct evidence (i.e.,labeling) is provided relative to the source of cells that pro-duced the regenerated tissue, we make the assumption thatthe transplanted cells at least partly contributed to the re-generation because the bone core specimen analyzed washarvested from the central region of the defect and graft site.

2818 KAIGLER ET AL.

Page 11: Angiogenic and Osteogenic Potential of Bone Repair Cells

We were able to identify this exact region through the use ofsurgical measurement templates/guides. Yet, even still, de-spite these promising clinical results, the fact that they wereobtained in a single patient can be viewed as a study limi-tation and minimizes the general conclusions that can bedrawn. An additional note is that this case presentation ispart of a larger, U.S. FDA-regulated, randomized, controlledPhase I/II trial where a larger number of patients are to betreated with BRCs. This larger study is still ongoing as itincludes a 1-year patient follow-up; however, upon studycompletion, all the clinical data will be analyzed and theresults outlined in a future report. While it is realized that thefeasibility of this process for routine tooth extraction sur-geries is most likely not practical, this study was conductedas an FDA Phase I/II study to examine safety and efficacy ofthis therapy for regeneration of craniofacial bone. If resultsare favorable, this type of therapy may certainly be feasiblefor larger, more challenging craniofacial reconstructions.

Conclusion

There is a growing interest in cell therapy strategies toregenerate craniofacial tissues, particularly bone. However,key questions to be considered in utilizing these strategiesare the following: What is the source of cells used in theseapproaches? How will the cells be processed and expandedto reach appropriate cell numbers for clinical applications?What is the phenotype and regenerative capacity of the cellsproduced? Through the current investigation, we report anovel approach to craniofacial regeneration with the utili-zation of an automated, closed-system, cell expansion pro-cess for the clinical-scale production of autologous cells,enriched in mesenchymal and vascular cell phenotypes.Additionally, it was demonstrated clinically that these cellspossess the capacity to give rise to highly vascular bone,6 weeks after transplantation into a jawbone defect. Althoughthe findings presented herein cannot fully elucidate the an-swers to the aforementioned questions, they provide im-portant insight toward that end.

Acknowledgments

The authors would like to acknowledge Brad Martin, JanetHock, and Niklaus Lang for their advisement during thecourse of the study, as well as Judy Douville, Tina Huffman,Emily Thorpe, Emily Hall, and Lea Franco for their admin-istrative and technical assistance with the clinical arm of thestudy. Additional acknowledgements to Chris Jung for ar-tistic development of Figure 1 and Sasha Meshinchi fortechnical support. This study was supported by a CareerAward for Medical Scientists given by the Burroughs Well-come Fund and the Michigan Institute for Clinical ResearchPilot Grant Program, and NIH/NCRRUL1RR024986.

Disclosure Statement

The authors would like to disclose that the author RonndaL. Bartel is an employee of Aastrom Biosciences.

References

1. Langer, R., and Vacanti, J.P. Tissue engineering. Science 260,

920, 1993.

2. Alsberg, E., Hill, E.E., and Mooney, D.J. Craniofacial tissueengineering. Crit Rev Oral Biol Med 12, 64, 2001.

3. Kaigler, D., Cirelli, J.A., and Giannobile, W.V. Growth factordelivery for oral and periodontal tissue engineering. ExpertOpin Drug Deliv 3, 647, 2006.

4. Shi, S., Bartold, P.M., Miura, M., Seo, B.M., Robey, P.G., andGronthos, S. The efficacy of mesenchymal stem cells to re-generate and repair dental structures. Orthod Craniofac Res8, 191, 2005.

5. Krebsbach, P.H., Kuznetsov, S.A., Satomura, K., Emmons,R.V., Rowe, D.W., and Robey, P.G. Bone formation in vivo:comparison of osteogenesis by transplanted mouse andhuman marrow stromal fibroblasts. Transplantation 63,

1059, 1997.6. Friedenstein, A.J., Ivanov-Smolenski, A.A., Chajlakjan, R.K.,

Gorskaya, U.F., Kuralesova, A.I., Latzinik, N.W., and Ger-asimow, U.W. Origin of bone marrow stromal mechanocytesin radiochimeras and heterotopic transplants. Exp Hematol6, 440, 1978.

7. Ishaug-Riley, S.L., Okun, L.E., Prado, G., Applegate, M.A.,and Ratcliffe, A. Human articular chondrocyte adhesion andproliferation on synthetic biodegradable polymer films.Biomaterials 20, 2245, 1999.

8. Chang, F., Ishii, T., Yanai, T., Mishima, H., Akaogi, H.,Ogawa, T., and Ochiai, N. Repair of large full-thickness ar-ticular cartilage defects by transplantation of autologousuncultured bone-marrow-derived mononuclear cells. J Or-thop Res 26, 18, 2008.

9. Marcacci, M., Kon, E., Moukhachev, V., Lavroukov, A.,Kutepov, S., Quarto, R., Mastrogiacomo, M., and Cancedda,R. Stem cells associated with macroporous bioceramics forlong bone repair: 6- to 7-year outcome of a pilot clinicalstudy. Tissue Eng 13, 947, 2007.

10. Faundez, A.A., Taylor, S., and Kaelin, A.J. Instrumentedfusion of thoracolumbar fracture with type I mineralizedcollagen matrix combined with autogenous bone marrow asa bone graft substitute: a four-case report. Eur Spine J 15

Suppl 5, 630, 2006.11. Gimbel, M., Ashley, R.K., Sisodia, M., Gabbay, J.S., Wasson,

K.L., Heller, J., Wilson, L., Kawamoto, H.K., and Bradley,J.P. Repair of alveolar cleft defects: reduced morbidity withbone marrow stem cells in a resorbable matrix. J CraniofacSurg 18, 895, 2007.

12. Ueda, M., Yamada, Y., Kagami, H., and Hibi, H. Injectablebone applied for ridge augmentation and dental implantplacement: human progress study. Implant Dent 17, 82,2008.

13. Velardi, F., Amante, P.R., Caniglia, M., De Rossi, G., Gaglini,P., Isacchi, G., Palma, P., Procaccini, E., and Zinno, F. Os-teogenesis induced by autologous bone marrow cells trans-plant in the pediatric skull. Childs Nerv Syst 22, 1158, 2006.

14. Soltan, M., Smiler, D., and Choi, J.H. Bone marrow:orchestrated cells, cytokines, and growth factors for boneregeneration. Implant Dent 18, 132, 2009.

15. Gerber, H.P., Vu, T.H., Ryan, A.M., Kowalski, J., Werb, Z.,and Ferrara, N. VEGF couples hypertrophic cartilage re-modeling, ossification and angiogenesis during endochon-dral bone formation. Nat Med 5, 623, 1999.

16. Kaigler, D., Krebsbach, P.H., Wang, Z., West, E.R., Horger,K., and Mooney, D.J. Transplanted endothelial cells enhanceorthotopic bone regeneration. J Dent Res 85, 633, 2006.

17. Grellier, M., Granja, P.L., Fricain, J.C., Bidarra, S.J., Renard,M., Bareille, R., Bourget, C., Amedee, J., and Barbosa, M.A.The effect of the co-immobilization of human osteoprogenitors

BONE REPAIR CELLS FOR CRANIOFACIAL REGENERATION 2819

Page 12: Angiogenic and Osteogenic Potential of Bone Repair Cells

and endothelial cells within alginate microspheres on min-eralization in a bone defect. Biomaterials 30, 3271, 2009.

18. Kim, S.S., Park, M.S., Cho, S.W., Kang, S.W., Ahn, K.M., Lee,J.H., and Kim, B.S. Enhanced bone formation by marrow-derived endothelial and osteogenic cell transplantation.J Biomed Mater Res A 92, 246, 2010.

19. Moioli, E.K., Clark, P.A., Chen, M., Dennis, J.E., Erickson,H.P., Gerson, S.L., and Mao, J.J. Synergistic actions of he-matopoietic and mesenchymal stem/progenitor cells invascularizing bioengineered tissues. PLoS ONE 3, e3922, 2008.

20. Schwartz, R.M., Palsson, B.O., and Emerson, S.G. Rapidmedium perfusion rate significantly increases the produc-tivity and longevity of human bone marrow cultures. ProcNatl Acad Sci USA 88, 6760, 1991.

21. Caldwell, J., Palsson, B.O., Locey, B., and Emerson, S.G.Culture perfusion schedules influence the metabolic activityand granulocyte-macrophage colony-stimulating factorproduction rates of human bone marrow stromal cells. J CellPhysiol 147, 344, 1991.

22. Stiff, P., Chen, B., Franklin, W., Oldenberg, D., Hsi, E., Bayer,R., Shpall, E., Douville, J., Mandalam, R., Malhotra, D.,Muller, T., Armstrong, R.D., and Smith, A. Autologoustransplantation of ex vivo expanded bone marrow cellsgrown from small aliquots after high-dose chemotherapy forbreast cancer. Blood 95, 2169, 2000.

23. Pecora, A.L., Stiff, P., LeMaistre, C.F., Bayer, R., Bachier, C.,Goldberg, S.L., Parthasarathy, M., Jennis, A.A., Smith, A.K.,Douville, J., Chen, B., Armstrong, R.D., Mandalam, R.K.,and Preti, R. A phase II trial evaluating the safety and ef-fectiveness of the AastromReplicell system for augmentationof low-dose blood stem cell transplantation. Bone MarrowTransplant 28, 295, 2001.

24. Whyte, M.P., Kurtzberg, J., McAlister, W.H., Mumm, S.,Podgornik, M.N., Coburn, S.P., Ryan, L.M., Miller, C.R.,Gottesman, G.S., Smith, A.K., Douville, J., Waters-Pick, B.,Armstrong, R.D., and Martin, P.L. Marrow cell transplan-tation for infantile hypophosphatasia. J Bone Miner Res 18,

624, 2003.25. Jaroscak, J., Goltry, K., Smith, A., Waters-Pick, B., Martin,

P.L., Driscoll, T.A., Howrey, R., Chao, N., Douville, J., Bur-hop, S., Fu, P., and Kurtzberg, J. Augmentation of umbilicalcord blood (UCB) transplantation with ex vivo-expandedUCB cells: results of a phase 1 trial using the Aas-tromReplicell System. Blood 101, 5061, 2003.

26. Dennis, J.E., Esterly, K., Awadallah, A., Parrish, C.R.,Poynter, G.M., and Goltry, K.L. Clinical-scale expansion of amixed population of bone-marrow-derived stem and pro-genitor cells for potential use in bone-tissue regeneration.Stem Cells 25, 2575, 2007.

27. Krebsbach, P.H., and Robey, P.G. Dental and skeletal stemcells: potential cellular therapeutics for craniofacial regen-eration. J Dent Educ 66, 766, 2002.

28. Pittenger, M.F., Mackay, A.M., Beck, S.C., Jaiswal, R.K.,Douglas, R., Mosca, J.D., Moorman, M.A., Simonetti,D.W., Craig, S., and Marshak, D.R. Multilineage potentialof adult human mesenchymal stem cells. Science 284, 143,1999.

29. Manolagas, S.C., Burton, D.W., and Deftos, L.J. 1,25-Dihy-droxyvitamin D3 stimulates the alkaline phosphatase activ-ity of osteoblast-like cells. J Biol Chem 256, 7115, 1981.

30. Park, C.H., Abramson, Z.R., Taba, M., Jr., Jin, Q., Chang, J.,Kreider, J.M., Goldstein, S.A., and Giannobile, W.V. Three-dimensional micro-computed tomographic imaging ofalveolar bone in experimental bone loss or repair. J Period-ontol 78, 273, 2007.

31. Kaigler, D., Krebsbach, P.H., Polverini, P.J., and Mooney,D.J. Role of vascular endothelial growth factor in bonemarrow stromal cell modulation of endothelial cells. TissueEng 9, 95, 2003.

32. Nor, J.E., Christensen, J., Mooney, D.J., and Polverini, P.J.Vascular endothelial growth factor (VEGF)-mediated an-giogenesis is associated with enhanced endothelial cell sur-vival and induction of Bcl-2 expression. Am J Pathol 154,

375, 1999.33. Folkman, J., and Haudenschild, C. Angiogenesis in vitro.

Nature 288, 551, 1980.34. Voyta, J.C., Via, D.P., Butterfield, C.E., and Zetter, B.R.

Identification and isolation of endothelial cells based on theirincreased uptake of acetylated-low density lipoprotein.J Cell Biol 99, 2034, 1984.

35. Gastens, M.H., Goltry, K., Prohaska, W., Tschope, D.,Stratmann, B., Lammers, D., Kirana, S., Gotting, C., andKleesiek, K. Good manufacturing practice-compliant ex-pansion of marrow-derived stem and progenitor cells for celltherapy. Cell Transplant 16, 685, 2007.

36. Haynesworth, S.E., Baber, M.A., and Caplan, A.I. Cell sur-face antigens on human marrow-derived mesenchymal cellsare detected by monoclonal antibodies. Bone 13, 69, 1992.

37. Fonsatti, E., and Maio, M. Highlights on endoglin (CD105):from basic findings towards clinical applications in humancancer. J Transl Med 2, 18, 2004.

38. Boiret, N., Rapatel, C., Boisgard, S., Charrier, S., Tchirkov,A., Bresson, C., Camilleri, L., Berger, J., Guillouard, L.,Guerin, J.J., Pigeon, P., Chassagne, J., and Berger, M.G.CD34þCDw90(Thy-1)þ subset colocated with mesenchymalprogenitors in human normal bone marrow hematon units isenriched in colony-forming unit megakaryocytes and long-term culture-initiating cells. Exp Hematol 31, 1275, 2003.

39. Meijer, G.J., de Bruijn, J.D., Koole, R., and van Blitterswijk,C.A. Cell based bone tissue engineering in jaw defects.Biomaterials 29, 3053, 2008.

40. Caplan, A.I. Mesenchymal stem cells. J Orthop Res 9, 641,1991.

41. Goshima, J., Goldberg, V.M., and Caplan, A.I. The osteo-genic potential of culture expanded rat marrow mesenchy-mal cells assayed in vivo in calcium phosphate ceramicblocks. Clin Orthop Relat Res 262, 298, 1991.

Address correspondence to:Darnell Kaigler, D.D.S., M.S., Ph.D.

Department of Periodontics and Oral MedicineUniversity of Michigan

1011 N. UniversityAnn Arbor, MI 48109

E-mail: [email protected]

Received: February 8, 2010Accepted: April 20, 2010

Online Publication Date: June 30, 2010

2820 KAIGLER ET AL.

Page 13: Angiogenic and Osteogenic Potential of Bone Repair Cells

This article has been cited by:

1. Hector F. Rios, Zhao Lin, BiNa Oh, Chan Ho Park, William V. Giannobile. 2011. Cell- and Gene-Based Therapeutic Strategiesfor Periodontal Regenerative Medicine. Journal of Periodontology 1-13. [CrossRef]

2. P Jayakumar, L Di Silvio. 2010. Osteoblasts in bone tissue engineering. Proceedings of the Institution of Mechanical Engineers, PartH: Journal of Engineering in Medicine 224:12, 1415-1440. [CrossRef]

3. Arnaud Scherberich, Andreas M. Müller, Dirk J. Schäfer, Andrea Banfi, Ivan Martin. 2010. Adipose tissue-derived progenitorsfor engineering osteogenic and vasculogenic grafts. Journal of Cellular Physiology 225:2, 348-353. [CrossRef]