ArticlePDF Available

Stem Cells from Deciduous Tooth Repair Mandibular Defect in Swine

Authors:

Abstract and Figures

Stem cells from human exfoliated deciduous teeth have been identified as a new post-natal stem cell population with multipotential differentiation capabilities, including regeneration of mineralized tissues in vivo. To examine the efficacy of utilizing these stem cells in regenerating orofacial bone defects, we isolated stem cells from miniature pig deciduous teeth and engrafted the critical-size bone defects generated in swine mandible models. Our results indicated that stem cells from miniature pig deciduous teeth, an autologous and easily accessible stem cell source, were able to engraft and regenerate bone to repair critical-size mandibular defects at 6 months post-surgical reconstruction. This pre-clinical study in a large-animal model, specifically swine, allows for testing of a stem cells/scaffold construct in the restoration of orofacial skeletal defects and provides rapid translation of stem-cell-based therapy in orofacial reconstruction in human clinical trials.
Content may be subject to copyright.
249
RESEARCH REPORTS
Biological
DOI: 10.1177/0022034509333804
Received April 19, 2008; Last revision January 6, 2009;
Accepted January 30, 2009
A supplemental appendix to this article is published elec-
tronically at http://jdr.sagepub.com/supplemental.
Y. Zheng1, Y. Liu1, C.M. Zhang1, H.Y.
Zhang2, W.H. Li2, S. Shi3, A.D. Le3*,
and S.L. Wang1, 4*
1Salivary Gland Disease Center and the Molecular
Laboratory for Gene Therapy & Tooth Regeneration, Capital
Medical University School of Stomatology, Tian Tan Xi Li
No.4, Beijing 100050, China; 2Department of Cell Biology,
Municipal Laboratory for Liver Protection and Regulation
of Regeneration, Capital Medical University School of
Basic Medical Sciences, You An Men Wai No.10, Beijing
100069, China; 3The Center for Craniofacial Molecular
Biology, University of Southern California School of
Dentistry, 2250 Alcazar Street, CSA 103, Los Angeles, CA
90033, USA; and 4Department of Biochemistry and
Molecular Biology, Capital Medical University School of
Basic Medical Sciences, You An Men Wai No.10, Beijing
100069, China; *corresponding authors, slwang@ccmu.
edu.cn and anhle@usc.edu
J Dent Res 88(3):249-254, 2009
ABSTRACT
Stem cells from human exfoliated deciduous teeth
have been identified as a new post-natal stem cell
population with multipotential differentiation
capabilities, including regeneration of mineralized
tissues in vivo. To examine the efficacy of utilizing
these stem cells in regenerating orofacial bone
defects, we isolated stem cells from miniature pig
deciduous teeth and engrafted the critical-size
bone defects generated in swine mandible models.
Our results indicated that stem cells from minia-
ture pig deciduous teeth, an autologous and easily
accessible stem cell source, were able to engraft
and regenerate bone to repair critical-size man-
dibular defects at 6 months post-surgical recon-
struction. This pre-clinical study in a large-animal
model, specifically swine, allows for testing of a
stem cells/scaffold construct in the restoration of
orofacial skeletal defects and provides rapid trans-
lation of stem-cell-based therapy in orofacial
reconstruction in human clinical trials.
KEY WORDS: deciduous tooth, stem cell,
bone, tissue engineering, miniature pig
Stem Cells from Deciduous
Tooth Repair Mandibular
Defect in Swine
INTRODUCTION
Reconstruction of orofacial defects secondary to tumors and trauma relies on
different sources of bone grafts with inherent morbidity. Stem-cell-based
tissue engineering is a promising alternative for bone regeneration (Petite et al.,
2000; Bianco et al., 2001; Rose and Oreffo, 2002). The stem-cell-based therapeu-
tic approach can restore bone defects without incurring graft donor site morbidity.
Bone marrow mesenchymal stem cells (BMMSCs) have emerged as an important
cell source for bone regeneration (Gronthos et al., 2003; Mankani et al., 2006;
Mastrogiacomo et al., 2007), as has been demonstrated clinically in femur fracture
repair and regeneration (Shao et al., 2006). Previous studies have indicated that
the orofacial bone and dental tissues contain a variety of stem cells, including
dental pulp stem cells (DPSCs), stem cells from human exfoliated deciduous teeth
(SHED), periodontal ligament stem cells (PDLSCs), and orofacial mesenchymal
stem cells (Akintoye et al., 2006). These orofacial stem cells possess a higher
proliferation capacity when compared with BMMSCs, and their origin may be
associated with neural crest cells (Gronthos et al., 2000; Shi et al., 2002; Shi and
Gronthos, 2003; Seo et al., 2004; Akintoye et al., 2006). When transplanted sub-
cutaneously into immunocompromised mice, SHEDs, an easily accessible stem
cell source, are capable of generating robust amounts of bone in vivo (Miura et al.,
2003; Seo et al., 2008), suggesting a potential for bone regeneration. To explore
the feasibility of using SHED-based bone regeneration to treat orofacial bone
defects, we utilized swine as a pre-clinical animal model to test the regeneration
of critical-size mandibular defects using SHEDs. Our previous studies demon-
strated that miniature pigs (minipig) are appropriate large-animal models for oral
and craniofacial tissue engineering and therapies (Shan et al., 2005; Sonoyama
et al., 2006; Wang et al., 2007; Yan et al., 2007; Liu et al., 2008). In this study, we
utilized minipigs as a large-animal model to examine the feasibility of using autol-
ogous stem cells derived from miniature pig deciduous teeth—that is, SPD—to
repair critical-size mandibular bone defects.
MATERIALS & METHODS
Animals
Sixteen inbred female minipigs (4–6 mos old, weighing 20–30 kg each) were obtained
from the Institute of Animal Science of the Chinese Agriculture University. Minipigs
were kept under conventional conditions, with free access to water and a regular supply
of soft food diet. The study was performed in accordance with a protocol approved by
the Animal Care and Use Committees of Capital Medical University.
Cell Culture
Deciduous incisor pulp tissues from first and second deciduous incisors were harvested
from 16 inbred female minipigs. SPDs were isolated and cloned following established
250 Zheng et al. J Dent Res 88(3) 2009
protocols (Gronthos et al., 2000; Miura et al., 2003; Seo et al., 2008;
Appendix).
Immunocytochemistry
SPDs were subcultured in 24-chamber slides. Cells were fixed in 4%
paraformaldehyde for 15 min, blocked with non-specific antibodies,
and incubated with either anti-STRO-1 (R&D, Minneapolis, MN, USA)
at dilutions of 1:200 to 1:500 or anti-vimentin (Chemicon, Temecula,
CA, USA) at a dilution of 1:500 for 1 hr according to the manufactur-
er’s protocol. To test mouse anti-human STRO-1 antibody cross-
reactivity with pig tissues, we performed immunocytochemical stain on
human SHEDs, minipig SPDs, and lymphocytes, and found that only
human SHEDs and pig SPDs were specifically stained with human
STRO-1 antibody. Based on these findings, we utilized mouse anti-
human monoclonal antibody to STRO-1 in this study. Samples were
subsequently incubated with goat secondary antibodies for 45 min, and
observed by fluorescence microscopy. Non-immune serum served as
negative control. Subsequently, sections were counterstained with
DAPI. We used an alkaline phosphatase detection kit (Chemicon) to
examine the expression of ALP according to the manufacturer’s proto-
col, and the result was observed by light microscopy.
Flow Cytometric Analysis
Detached cells were permeabilized with PBS containing 0.1% (wt/v)
saponin at room temperature for 20 min. After being blocked with nor-
mal serum, the cells were incubated with fluorescein isothiocyanate
(FITC)-conjugated STRO-1 antibodies (R&D Systems; clone STRO-1)
or phycoerythrin (PE)-conjugated ALP antibodies (R&D systems; clone
B4-78) for 30 min at room temperature. After 3 washes with PBS con-
taining 0.1% saponin, fluorescence was analyzed by a FACSCalibur
flow cytometer with CellQuest software (BD Bioscience, Palo Alto,
CA, USA). Positive cells were identified by comparison with the cor-
responding isotype controls (FITC- or PE-conjugated IgG) in which a
false-positive rate of less than 2% was accepted.
Transfection of eGFP Genes
Conditional retroviral supernatants were produced by the stable retrovirus-
producing cell lines PT67/eGFP as described previously (Brazelton and
Blau, 2005; Zhang et al., 2005). For transfection, about 1 x 105 SPD
grown in 6-well plates were incubated for approximately 20 hrs with a
mixture of 1 vol of viral supernatant and growth medium at equal vols
and in the presence of 8 μg/mL polybrene (Sigma, St. Louis, MO,
USA). A repeated transfection was performed in a period of 72 hrs, and
the transfected cultures were selected with G418 (100 μg/mL, Sigma).
The transfection efficiency of the cells was 80%.
Scanning Electron Microscopy
GFP-positive SPDs were grown on β-TCP carrier (Biomedical Materials
and Engineering Center of Wuhan University of Technology, China) for
7 days and fixed with 2.5% glutaraldehyde in 0.1 M sodium cacodylate
buffer (pH 7.2) for 2 hrs at 4˚C. The samples were examined under a
Hitachi S-520 scanning electron microscope (Hitachi, Tokyo, Japan;
http://www.hitachi.com/).
Reconstruction of Mandibular Bone Defects
in Swine Model with SPDs
A critical-size defect of 2.5 x 1.5 x 1.5 cm3 was surgically created in the
parasymphyseal region of the mandible in 10 minipigs for a long-term
(24 wks) follow-up so that we could evaluate bone regeneration (Henkel
et al., 2005). In another 6 minipigs, we created 2 smaller defects of 1.0
x 1.0 x 0.5 cm3 bilaterally in the parasymphysis of the mandible and
evaluated short-term post-surgical follow-ups, at 2 wks (n = 3) and 4
wks (n = 3) (Appendix Fig. 1).
Computed Tomography (CT) Assessment
of Bone Formation
CT imaging of the minipig’s mandible (Siemens Company, Bensheim,
Germany) was carried out at 2, 4, 12, and 24 wks after transplantation
at a scanning length of 0.75 mm.
Quantitative and Histological Evaluation
of Regenerated Bone
Bone specimens were fixed in 10% buffered formalin. Half of the
specimens were decalcified and embedded in paraffin, while the other
half were processed as non-decalcified. Sections of 5- to 6-μm thickness
from the embedded specimen were stained with H&E. The extent of
ossification within each section was analyzed semi-quantitatively by
histomorphometric techniques (Appendix). The non-decalcified sample
was sectioned into a 400-μm series of slices (Donath and Breuner,
1982), and GFP-labeled SPDs were observed by fluorescence micros-
copy (Olympus BX/TF, U-LH100HG, Tokyo, Japan). For pair-wise
comparison, data were analyzed by one-way ANOVA and the Bonferroni
method.
RESULTS
Isolation and Characterization of SPDs
In culture, SPDs form adherent clonogenic cell clusters of
fibroblast-like cells, similar to the morphology described in
SHEDs; approximately 67 single colonies were generated
from 105 single cells cultured at low cell density. These
colony-forming cells were spindle-shaped (Fig. 1A) and posi-
tively stained for STRO-1, vimentin, and ALP (Figs. 1B-1D).
Flow cytometric analysis showed an average of 10.9% of
SPDs positive for STRO-1 and 21.1% positive for ALP (Figs.
1G, 1H), suggesting a heterogeneic population of cells, as
previously reported in other post-natal mesenchymal stem
cells, including bone marrow (Shi and Gronthos, 2003), dental
pulp (Gronthos et al., 2000), and periodontal ligament stem
cells (Seo et al., 2004).
Expression of eGFP Gene and Fabrication
of an SPD-Scaffold Construct
The retroviral transfection was carried out on day 2 in culture,
followed by a repeated transfection on day 3. Subsequently,
transfected SPDs were selected with G418 (100 μg/mL), and
colonies expressing green fluorescence were expanded in growth
medium (Fig. 1E). After several passages, the progenies of GFP-
positive SPDs continued to express eGFP.
For fabrication of the cell-scaffold construct, approximately
2 x 107-4 x 108 GFP-positive SPDs at third and fourth passages
were seeded onto β-TCP scaffolds and cultured in growth
medium for 7 days in vitro. SEM studies showed that GFP+-
SPDs were able to grow on β-TCP scaffolds (Fig. 1F).
J Dent Res 88(3) 2009 Regeneration of Bone Tissues 251
SPD-mediated Bone
Regeneration in the
Reconstructed Mandibular
Defect
At 24 wks, animals were
killed, and the mandibles were
harvested for CT, gross mor-
phology, and histological anal-
yses (Fig. 2, Appendix Fig. 2).
CT scan analyses showed
nearly complete regeneration
of initial defect sites, with new
bone formation in the SPD/β-
TCP treatment group as shown
in coronal (Fig. 2A) and axial
sections (Appendix Fig.
2A), three-dimensional view
(Appendix Fig. 2B), as well as
gross bone morphology of a
sagittal bone section (Appendix
Fig. 2C). Histological sections
of the SPD/β-TCP grafted site
revealed newly formed lamel-
lar bone and degraded β-TCP
scaffold (Fig. 2B). In contrast,
in the β-TCP group, the defect
site was only partially restored,
with significant remaining
defect in the lateral cortex
of the mandible (Fig. 2C,
Appendix Figs. 2D, 2E, and
2F). Histologically, the scaf-
fold-treated defect was par-
tially filled with some new
bone formation, connective
tissues, and degraded β-TCP
(Fig. 2D). In the control, or
untreated, group, a marked
Figure 1. Characterization of stem cells derived from miniature pig deciduous teeth (SPD). (A) Single
colonies of SPD showed typical fibroblast-like morphology under light microscopy. (B) SPDs expressed
STRO-1 by immunohistochemical staining with anti-STRO-1 antibody and (C) ALP-positive staining with the
alkaline phosphatase detection kit under light microscopy. (D) Immunohistochemical staining with anti-
vimentin antibody showed 99.5% positive staining. (E) GFP-expressed SPDs showed fibroblast-like
morphology. (F) Scanning electron microscopy revealed GFP-labeled SPDs grown on β-TCP scaffold. (G-H)
An average of 10.9% of the 3rd-passage SPDs were positively stained for STRO-1 (G) and 21.1% for ALP
by flow cytometric analysis (H). Abbreviations: SPD, stem cells from pig deciduous teeth; ALP, alkaline
phosphatase. Scale bars: 100 µm in A; 30 µm in B, C, D, E; 4 µm in F.
bone defect remained with predominantly connective tissues
(Figs. 2E, 2F). The regenerated defect site displayed a mean of
83.1% of mineralized matrix in the SPD/β-TCP-treated group,
significantly higher than the 52.2% in the scaffold group (P <
0.01), and 28.4% higher than in the control group (P < 0.01)
(Fig. 2G).
At 4 wks post-transplantation, CT scan revealed partial bridg-
ing of the lateral cortical continuity defect and moderate bone
regeneration in the SPD/β-TCP-treated group, in conjunction with
early scaffold degradation (Fig. 3A). The gross view of the recon-
structed defect site displayed marked absorption of scaffold at the
junction of native bone and graft in the SPD/β-TCP-treated group
at the early stage of bone healing (Appendix Fig. 3B). Relatively
intact scaffold was observed in the β-TCP group (Appendix Fig.
3C), and a large bony defect remained in the untreated group
(Appendix Fig. 3D). Histological sections of decalcified speci-
mens showed abundant new islands of bone and blood vessels
amid degraded scaffold in the SPD-treated group at 4 wks post-
reconstruction (Figs. 3B, 3C). In contrast, in the β-TCP group, the
lateral cortical rim defect remained, with minimal β-TCP absorp-
tion (Fig. 3D). The bone void at the defect site remained unfilled
in the control group (Fig. 3G, Appendix Fig. 3D). Histological
sections showed a lack of new bone formation at the junction of
β-TCP scaffold (Figs. 3E, 3F) and abundant connective tissue
proper in the untreated group (Figs. 3H, 3I). These findings dem-
onstrated that bone regeneration was significantly higher in the
SPD/β-TCP-treated group as compared with the β-TCP group. In
the blank (control) group, limited bone regeneration was observed
in the defect area, which was filled predominantly with connec-
tive tissue proper (Figs. 3G, 3H, 3I, Appendix Fig. 4D).
Detection of Transplanted GFP-positive SPD
in Regenerative Bone
To identify if the GFP-positive SPDs engrafted at the transplanted
bone defect sites had differentiated into osteoblasts, we first
screened non-decalcified sections using light microscopy for loca-
tion of osteoblasts and bone lacunae in the new bone at 2 wks and
252 Zheng et al. J Dent Res 88(3) 2009
4 wks post-operatively (Figs. 4A-4D).
Using the same visual field, we captured
GFP expressing SPDs under a fluorescence
microscope (Figs. 4B, 4D). When both
images in Figs. 4C and 4D were overlaid,
the locations of some GFP-positive SPDs
were superimposed with the osteoblasts
and bone lacunae (Fig. 4E, yellow arrows
showing GFP-negative osteoblasts, red
arrows showing GFP-positive osteoblasts),
suggesting that GFP-positive SPD cells
might have differentiated to osteoblasts. As
expected, the normal bone tissue section
showed a uniformly distributed green fluo-
rescence signal without local accumulation
of GFP-positive cells under immunofluo-
rescence microscopy (Fig. 4F).
DISCUSSION
Craniofacial tissue engineering by stem
cells is a fast-moving field with consider-
able potential clinical applications (Mao
et al., 2006; Kaigler et al., 2006; Zhao
et al., 2007). Deciduous tooth stem cells are
an easily accessible stem cell source and
capable of robust ex vivo expansion for
several potential clinical applications
(Miura et al., 2003; Mao et al., 2006; Seo
et al., 2008). Embryo logically derived from
the neural crest cell, SHEDs and SPDs may
share similar tissue origin with the man-
dibular bone cells, and therefore, may serve
as a better cell source for the regeneration
of alveolar and orofacial bone defects.
A previous study showed that the bone-
regenerative capacity of SHEDs was simi-
lar to that of bone marrow mesenchymal
stem cells (BMMSC) when transplanted
into immunocompromised mice at 8 wks
post-transplantation (33% mineralized
matrix/area in the SHED group vs. 31%
mineralized matrix/area in the BMMSC
group) (Seo et al., 2008). Similar to SHEDs
and other post-natal mesenchymal stem
cells derived from bone marrow (Shi and
Gronthos, 2003), dental pulp (Gronthos
et al., 2000), and periodontal ligament stem
cells (Seo et al., 2004), it is not unexpected
that the SPDs are a heterogeneic population
of cells which, following ex vivo cultures,
may display different percentages of cells
positive for STRO-1, or form mineralized
nodules, or be positive for Oil red O under
differentiation inductive conditions.
In this study, to investigate SPD-
mediated bone formation in vivo, we labeled
SPDs with GFP and used SPDs to repair
critical-size bone defects in the mandibles in
swine. At 2 and 4 wks after transplantation,
Figure 3. SPD-mediated bone regeneration of small-size mandibular defect at the short-term (wks)
follow-up. (A, B, C) SPD/β-TCP-treated group: CT axial image showing replacement of β-TCP scaffold
with new bone restoration (red arrow), and bridging of the lateral cortical defect with new bone
formation (blue arrow). H&E stain of the regenerated defect site at 10X (B) and 40X (C) revealed
several new islands of bones and blood vessels amid degraded scaffold. (D, E, F) β-TCP-treated
group: CT axial scan showing a partial bridging of the cortical defect and no visible bone formation;
H&E stains (E, F) showing sparse new bone formation at the junction of β-TCP scaffold and partially
degraded β-TCP. (G, H, I) Untreated, or control, group: CT axial scan showing a continuity defect in
the lateral cortex and lack of bone regeneration. Histological sections show primarily connective tissues
filling the defect of the control group (H and I). Scale bars: 1cm in A, D, G; 1 mm in B, E, H; 100 µm
in C, F, I. BT, bone tissue; BV, blood vessel; CT, connective tissue.
Figure 2. SPD-mediated bone regeneration of critical-size mandibular defect at long-term follow-up. (A,
B) SPD/β-TCP-treated group: CT coronal image showed a large quantity of new bone formation filling
the bone defect at 24 wks post-transplantation; yellow dot area indicates the original site of the bone
defect (A); the histological section shows that the defect was filled with new bone (B). (C, D) β-TCP-treated
group: CT coronal image shows remaining smaller bone defect at the reconstructed defect site;
histological section shows that the defect was partially filled with connective tissues, β-TCP scaffold, and
new bone formation (D). (E, F) Untreated, or control, group: CT coronal image shows limited bone
regeneration and large bone defect remaining; histological section shows that the defect was filled
primarily with connective tissues. (G) Semi-quantitative analysis of bone formation showed a statistically
significant increase in mineralized matrix formation at the regenerated defect site, 83.1 ± 5.75% (mean
± SD, n = 4) in the SPD/β-TCP treated group, compared with 52.2 ± 4.54% (n = 3) in the scaffold group
(P < 0.01), and 28.4 ± 2.79% (n = 3) in the control group (P < 0.01). Scale bars: 1 cm in A, C, E; 50
µm in B, F; 30 µm in D. BT, bone tissue; BV, blood vessel; CT, connective tissue.
J Dent Res 88(3) 2009 Regeneration of Bone Tissues 253
green fluorescence signals were
detected by fluorescence microscopy
within newly formed woven bone.
Photomicrographs of the same visual
fields confirmed that GFP-labeled
SPDs had differentiated directly into
new bone, while the normal bone tis-
sue section was non-specifically labeled
with green fluorescence. These find-
ings suggested that SPDs were
engrafted to some extent at the treated
site and contributed to new bone regen-
eration in the restoration of the bone
defect in the swine mandible model.
The results of CT scan, gross view, and
histological analyses consistently
showed that the SPD group had the
earliest and strongest capacity of bone
regeneration compared with other
groups. At 4 wks post-transplantation,
β-TCP was partly degraded and was
replaced with a large quantity of new
bone formation. At 6 mos post-recon-
struction, the defects in the SPD group
were markedly restored with new bone,
while in the β-TCP group and control
group, much less bone regeneration
and predominantly connective tissue
granulation were evident at the defect
sites. Further studies to regulate bone
regeneration are under way to optimize
the transplanted stem cell numbers,
scaffolds, and their immediate niche
component.
Overall, our study provides the
first evidence that SPDs are capable of
regenerating critical-size defects in
the orofacial bone in a large-animal
model, specifically swine, and may
potentially serve as an alternative
stem-cell-based approach in the recon-
struction of alveolar and orofacial
bone defects.
ACKNOWLEDGMENTS
This work was supported by
grants from the National Basic
Research Program of China (No.
2007CB947304), the Beijing Major
Scientific Program (D090600700
0091), the National Natural Science
Foundation of China (Grants 3042
8009 and 30801297), and the US National Institutes of Health
(NIDCR R01DE17449 to S.S.).
REFERENCES
Akintoye SO, Lam T, Shi S, Brahim J, Collins MT, Robey PG (2006). Skeletal
site-specific characterization of orofacial and iliac crest human bone
marrow stromal cells in same individuals. Bone 38:758-768.
Figure 4. Tracking GFP-labeled SPD in new bone tissue regeneration by immunofluorescence microscopy.
(A, B) Tracking GFP-positive cells at 2 wks post-transplantation. Trabecular bone/bone lacunae and β-TCP
scaffold in the restored defect area were observed under light microscopy (A). In the same visual field, the
cells from newly formed bone tissue and β-TCP scaffold area were screened for GFP-labeled SPD by
fluorescence microscopy (B). (C, D, E) Tracking GFP-positive cells at 4 wks post-transplantation. New
trabecular bone and lacunae were observed in the defect area (C). In the same visual field, the cells in
newly regenerated bone tissue were shown to express GFP label by fluorescence microscopy (D). When
Figs. 4C and 4D were overlaid, the locations of some GFP-positive SPDs were superimposed within the
area of bone lacunae (yellow arrow shows GFP-negative bone cells, red arrows show GFP-positive bone
cells), suggesting that new immature bone tissues may be derived from both GFP-labeled SPDs and host
osteoblasts. (F) Normal bone tissue section of miniature pig’s mandible showing non-specific distribution
of green fluorescence by immunofluorescence microscopy. Scale bars: 50 µm.
Bianco P, Riminucci M, Gronthos S, Robey PG (2001). Bone marrow
stromal stem cells: Nature, biology, and potential applications. Stem
Cells 19:180-192.
Brazelton TR, Blau HM (2005). Optimizing techniques for tracking trans-
planted stem cells in vivo. Stem Cells 23:1251-1265.
Donath K, Breuner G (1982). A method for the study of undecalcified bones
and teeth with attached soft tissues. The Sage-Schliff (sawing and
grinding) technique. J Oral Pathol 11:318-326.
254 Zheng et al. J Dent Res 88(3) 2009
Gronthos S, Mankani M, Brahim J, Robey PG, Shi S (2000). Postnatal
human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc Natl
Acad Sci USA 97:13625-13630.
Gronthos S, Zannettino AC, Hay SJ, Shi S, Graves SE, Kortesidis A, et al.
(2003). Molecular and cellular characterization of highly purified
stromal stem cells derived from human bone marrow. J Cell Sci 116(Pt
9):1827-1835.
Henkel KO, Gerber T, Dorfling P, Gundlach KK, Bienengraber V (2005).
Repair of bone defects by applying biomatrices with and without
autologous osteoblasts. J Craniomaxillofac Surg 33:45-49.
Kaigler D, Krebsbach PH, Wang Z, West ER, Horger K, Mooney DJ (2006).
Transplanted endothelial cells enhance orthotopic bone regeneration. J
Dent Res 85:633-637.
Liu Y, Zhang Y, Ding G, Fang DJ, Zhang CM, Shi S, et al. (2008).
Periodontal ligament stem cells-mediated treatment for periodontitis in
miniature swine. Stem Cells 26:1065-1073.
Mankani MH, Kuznetsov SA, Shannon B, Nalla RK, Ritchie RO, Qin Y,
et al. (2006). Canine cranial reconstruction using autologous bone mar-
row stromal cells. Am J Pathol 168:542-550.
Mao JJ, Giannobile WV, Helms JA, Hollister SJ, Krebsbach PH, Longaker
MT, et al. (2006). Craniofacial tissue engineering by stem cells. J Dent
Res 85:966-979.
Mastrogiacomo M, Papadimitropoulos A, Cedola A, Peyrin F, Giannoni P,
Pearce SG, et al. (2007). Engineering of bone using bone marrow
stromal cells and a silicon-stabilized tricalcium phosphate bioceramic:
evidence for a coupling between bone formation and scaffold resorp-
tion. Biomaterials 28:1376-1384.
Miura M, Gronthos S, Zhao M, Lu B, Fisher LW, Robey PG, et al. (2003).
SHED: stem cells from human exfoliated deciduous teeth. Proc Natl
Acad Sci USA 100:5807-5812.
Petite H, Viateau V, Bensaid W, Meunier A, de Pollak C, Bourguignon M, et al.
(2000). Tissue engineered bone regeneration. Nat Biotechnol 18:959-962.
Rose FR, Oreffo RO (2002). Bone tissue engineering: hope vs hype.
Biochem Biophys Res Commun 292:1-7.
Seo BM, Miura M, Gronthos S, Bartold PM, Batouli S, Brahim J, et al.
(2004). Investigation of multipotent postnatal stem cells from human
periodontal ligament. Lancet 364:149-155.
Seo BM, Sonayama W, Yamaza T, Coppe C, Kikuiri T, Akiyama K, et al.
(2008). SHED repair critical-size calvarial defects in mice. Oral Dis
14:428-434.
Shan Z, Li J, Ou G, Liu X, Zhang C, Baum BJ, et al. (2005). Structural and
functional characteristics of irradiation damage to parotid glands in the
miniature pig. Int J Radiat Oncol Biol Phys 62:1510-1516.
Shao X, Goh JC, Hutmacher DW, Lee EH, Zigang G (2006). Repair of large
articular osteochondral defects using hybrid scaffolds and bone mar-
row-derived mesenchymal stem cells in a rabbit model. Tissue Eng 12:
1539-1551.
Shi S, Gronthos S (2003). Perivascular niche of postnatal mesenchymal
stem cells in human bone marrow and dental pulp. J Bone Miner Res
18:696-704.
Shi S, Gronthos S, Chen S, Reddi A, Counter CM, Robey PG, et al. (2002).
Bone formation by human postnatal bone marrow stromal stem cells is
enhanced by telomerase expression. Nat Biotechnol 20:587-591.
Sonoyama W, Liu Y, Fang D, Yamaza T, Seo BM, Zhang C, et al. (2006).
Mesenchymal stem cell-mediated functional tooth regeneration in
swine. PLoS ONE 1:e79.
Wang SL, Liu Y, Fang DJ, Shi ST (2007). The miniature pig: a useful large
animal model for dental and orofacial research. Oral Dis 13:530-537.
Yan X, Voutetakis A, Zheng C, Hai B, Zhang CM, Baum BJ, et al. (2007).
Sorting of transgenic secretory proteins in miniature pig parotid glands
following adenoviral mediated gene transfer. J Gene Med 9:779-787;
erratum in J Gene Med 9:1106, 2007.
Zhang H, Zhao Y, Zhao C, Yu S, Duan D, Xu Q (2005). Long-term expan-
sion of human neural progenitor cells by epigenetic stimulation in vitro.
Neurosci Res 51:157-165.
Zhao Z, Wang Z, Ge C, Krebsbach P, Franceschi RT (2007). Healing cranial
defects with AdRunx2-transduced marrow stromal cells. J Dent Res
86:1207-1211.
... In addition, this literature review presented the results of MSC surface markers both from flow cytometry and polymerase chain reaction (PCR) results, including CD146 (MSC multipotency), CD44 (cell-matrix interaction), CD29 (cell adhesion), CD73 (cell migration and anti-inflammatory property) CD90 (cell adhesion, migration, homing, proliferation, apoptosis, and differentiation), CD105 (cell migration, proliferation, and differentiation), and CD45 (hematopoietic stem cell (HSC) marker) (43)(44)(45)(46)(47)(48)(49). Stem cells have also been isolated from the oral tissues of non-human mammalian species including canines, felines, equines, chimpanzees, swines, murines, and minipigs (34,43,(50)(51)(52)(53). Among these, canine-derived oral-origin MSCs have been widely studied (41,43,(54)(55)(56)(57)(58)(59)(60)(61)(62)(63)(64). ...
Article
Full-text available
Oral and maxillofacial (OMF) defects are not limited to humans and are often encountered in other species. Reconstructing significant tissue defects requires an excellent strategy for efficient and cost-effective treatment. In this regard, tissue engineering comprising stem cells, scaffolds, and signaling molecules is emerging as an innovative approach to treating OMF defects in veterinary patients. This review presents a comprehensive overview of OMF defects and tissue engineering principles to establish proper treatment and achieve both hard and soft tissue regeneration in veterinary practice. Moreover, bench-to-bedside future opportunities and challenges of tissue engineering usage are also addressed in this literature review.
... In the first in-situ animal study the SHED cells from miniature pigs were seeded on beta-TCP scaffolds and inserted into the critical sized bone defects of mandibular bone. These cells were able to repair the defects six months after surgery [83]. However similar study using human SHED cells with collagen scaffolds got no improvement in bone formation compared to the control group [84]. ...
... 215 Miniature pig stem cells were extracted from the ilium, bone marrow, or deciduous teeth and grafted onto the critical-sized bone lesions created in pig mandible models. 201,202,[216][217][218][219] Models of porcine joints and tissues, such as ligaments, bone, and cartilage, have long been used to study the biomechanics of these joints. 5 The pig has been utilized for direct measurements of TMJ tissue deformation and load during biting, making it the greatest non-primate model for human TMJ diseases. ...
Article
The craniofacial region contains skin, bones, cartilage, the temporomandibular joint, teeth, periodontal tissues, mucosa, salivary glands, muscles, nerves, and blood vessels. Applying tissue engineering therapeutically helps replace lost tissues after trauma or cancer. Despite recent advances, it remains essential to standardize and validate the most appropriate animal models to effectively translate pre-clinical data to clinical situations. Therefore, this review focused on applying various animal models in craniofacial tissue engineering and regeneration. This research was based on PubMed, Scopus, and Google Scholar data available until January 2023. This study included only English-language publications describing animal models' application in craniofacial tissue engineering (in vivo and review studies). Study selection was based on evaluating titles, abstracts, and full texts. The total number of initial studies was 6454. Following the screening process, 295 articles remained on the final list. Numerous in vivo studies have shown that small and large animal models can benefit clinical conditions by assessing the efficacy and safety of new therapeutic interventions, devices, and biomaterials in animals with similar diseases/defects to humans. Different species' anatomical, physiologic, and biological features must be considered in developing innovative, reproducible, and discriminative experimental models to select an appropriate animal model for a specific tissue defect. As a result, understanding the parallels between human and veterinary medicine can benefit both fields.
Article
Objective Pigs are emerging as a preferred experimental in vivo model for bone regeneration. The study objective was to answer the focused PEO question: in the pig model (P), what is the capacity of experimental alveolar bone defects (E) for spontaneous regeneration in terms of new bone formation (O)? Methods Following PRISMA guidelines, electronic databases were searched for studies reporting experimental bone defects or extraction socket healing in the maxillae or mandibles of pigs. The main inclusion criteria were the presence of a control group of untreated defects/sockets and the assessment of regeneration via 3D tomography [radiographic defect fill (RDF)] or 2D histomorphometry [new bone formation (NBF)]. Random effects meta‐analyses were performed for the outcomes RDF and NBF. Results Overall, 45 studies were included reporting on alveolar bone defects or extraction sockets, most frequently in the mandibles of minipigs. Based on morphology, defects were broadly classified as ‘box‐defects’ (BD) or ‘cylinder‐defects’ (CD) with a wide range of healing times (10 days to 52 weeks). Meta‐analyses revealed pooled estimates (with 95% confidence intervals) of 50% RDF (36.87%–63.15%) and 43.74% NBF (30.47%–57%) in BD, and 44% RDF (16.48%–71.61%) and 39.67% NBF (31.53%–47.81%) in CD, which were similar to estimates of socket‐healing [48.74% RDF (40.35%–57.13%) and 38.73% NBF (28.57%–48.89%)]. Heterogeneity in the meta‐analysis was high ( I ² > 90%). Conclusion A substantial body of literature revealed a high capacity for spontaneous regeneration in experimental alveolar bone defects of (mini)pigs, which should be considered in future studies of bone regeneration in this animal model.
Article
Full-text available
Dental stem cells (DSCs) have attracted significant interest as autologous stem cells since they are easily accessible and give a minimal immune response. These properties and their ability to both maintain self-renewal and undergo multi-lineage differentiation establish them as key players in regenerative medicine. While many regulatory factors determine the differentiation trajectory of DSCs, prior research has predominantly been based on genetic, epigenetic, and molecular aspects. Recent evidence suggests that DSC differentiation can also be influenced by autophagy, a highly conserved cellular process responsible for maintaining cellular and tissue homeostasis under various stress conditions. This comprehensive review endeavors to elucidate the intricate regulatory mechanism and relationship between autophagy and DSC differentiation. To achieve this goal, we dissect the intricacies of autophagy and its mechanisms. Subsequently, we elucidate its pivotal roles in impacting DSC differentiation, including osteo/odontogenic, neurogenic, and angiogenic trajectories. Furthermore, we reveal the regulatory factors that govern autophagy in DSC lineage commitment, including scaffold materials, pharmaceutical cues, and the extrinsic milieu. The implications of this review are far-reaching, underpinning the potential to wield autophagy as a regulatory tool to expedite DSC-directed differentiation and thereby promote the application of DSCs within the realm of regenerative medicine.
Preprint
Full-text available
Objective Pigs are emerging as a preferred experimental in vivo model for bone regeneration. The study objective was to answer the focused PEO question: in the pig model (P), what is the capacity in experimental alveolar bone defects (E) for spontaneous regeneration in terms of new bone formation (O)? Methods Following PRISMA guidelines, electronic databases were searched for studies reporting experimental bone defect- or extraction socket-healing in the maxillae or mandibles of pigs. The main inclusion criteria were presence of a control group of untreated defects/sockets and assessment of regeneration via 3D tomography [radiographic defect fill (RDF)] or 2D histomorphometry [new bone formation (NBF)]. Random effects meta-analyses were performed for the outcomes RDF and NBF. Results Overall, 45 studies were included reporting on alveolar bone defects or extraction sockets, most frequently in the mandibles of minipigs. Based on morphology, defects were broadly classified as “box-defects” (BD) or “cylinder-defects” (CD) with a wide range of healing times (10 days to 52 weeks). Meta-analyses revealed pooled estimates (with 95% confidence intervals) of 50% RDF (36.87–63.15%) and 43.74% NBF (30.47–57%) in BD, and 44% RDF (16.48–71.61%) and 39.67% NBF (31.53–47.81%) in CD, which were similar to estimates of socket-healing [48.74% RDF (40.35–57.13%) and 38.73% NBF (28.57–48.89%)]. Heterogeneity in the meta-analysis was high (I² > 90%). Conclusion A substantial body of literature revealed a high capacity for spontaneous regeneration in experimental alveolar bone defects of (mini)pigs, which should be considered in future studies of bone regeneration in this animal model.
Chapter
Among other large experimental animal models, like the dog, the sheep or the monkey, swine has a long history in translational biomedical research as well as for procedural training in different surgical disciplines [1,2,3]. In recent years, swine has attracted attention in musco-skeletal surgery and in oral, maxillofacial bone surgery related research [4, 5]. This chapter will specifically discuss the place of swine as an experimental model for dental implant rehabilitation.
Article
Full-text available
Bone marrow stromal cells are progenitors of skeletal tissue components such as bone, cartilage, the hematopoiesis-supporting stroma, and adipocytes. In addition, they may be experimentally induced to undergo unorthodox differentiation, possibly forming neural and myogenic cells. As such, they represent an important paradigm of post-natal nonhematopoietic stem cells, and an easy source for potential therapeutic use. Along with an overview of the basics of their biology, we discuss here their potential nature as components of the vascular wall, and the prospects for their use in local and systemic transplantation and gene therapy.
Article
Full-text available
Stem cells from human exfoliated deciduous teeth (SHED) are a population of highly proliferative postnatal stem cells capable of differentiating into odontoblasts, adipocytes, neural cells, and osteo-inductive cells. To examine whether SHED-mediated bone regeneration can be utilized for therapeutic purposes, we used SHED to repair critical-size calvarial defects in immunocompromised mice. We generated calvarial defects and transplanted SHED with hydroxyapatite/tricalcium phosphate as a carrier into the defect areas. SHED were able to repair the defects with substantial bone formation. Interestingly, SHED-mediated osteogenesis failed to recruit hematopoietic marrow elements that are commonly seen in bone marrow mesenchymal stem cell-generated bone. Furthermore, SHED were found to co-express mesenchymal stem cell marker, CC9/MUC18/CD146, with an array of growth factor receptors such as transforming growth factor beta receptor I and II, fibroblast growth factor receptor I and III, and vascular endothelial growth factor receptor I, implying their comprehensive differentiation potential. Our data indicate that SHED, derived from neural crest cells, may select unique mechanisms to exert osteogenesis. SHED might be a suitable resource for orofacial bone regeneration.
Article
Full-text available
Bone lesions above a critical size become scarred rather than regenerated, leading to nonunion. We have attempted to obtain a greater degree of regeneration by using a resorbable scaffold with regeneration-competent cells to recreate an embryonic environment in injured adult tissues, and thus improve clinical outcome. We have used a combination of a coral scaffold with in vitro-expanded marrow stromal cells (MSC) to increase osteogenesis more than that obtained with the scaffold alone or the scaffold plus fresh bone marrow. The efficiency of the various combinations was assessed in a large segmental defect model in sheep. The tissue-engineered artificial bone underwent morphogenesis leading to complete recorticalization and the formation of a medullary canal with mature lamellar cortical bone in the most favorable cases. Clinical union never occurred when the defects were left empty or filled with the scaffold alone. In contrast, clinical union was obtained in three out of seven operated limbs when the defects were filled with the tissue-engineered bone.
Article
Full-text available
Previous studies have provided evidence for the existence of adult human bone marrow stromal stem cells (BMSSCs) or mesenchymal stem cells. Using a combination of cell separation techniques, we have isolated an almost homogeneous population of BMSSCs from adult human bone marrow. Lacking phenotypic characteristics of leukocytes and mature stromal elements, BMSSCs are non-cycling and constitutively express telomerase activity in vivo. This mesenchymal stem cell population demonstrates extensive proliferation and retains the capacity for differentiation into bone, cartilage and adipose tissue in vitro. In addition, clonal analysis demonstrated that individual BMSSC colonies exhibit a differential capacity to form new bone in vivo. These data are consistent with the existence of a second population of bone marrow stem cells in addition to those for the hematopoietic system. Our novel selection protocol provides a means to generate purified populations of BMSSCs for use in a range of different tissue engineering and gene therapy strategies.
Article
A new sawing-grinding method is described for the histological evaluation of jaw bones with teeth or bones containing implants (ceramic or metallic). The undecalcified bone is embedded in acrylic resin and sawed at 100 to 150 micrometers. The slices are ground automatically by a special machine to a thickness of 5-10 micrometers. The usually employed staining procedures for hard plastic embedded-tissues may be used. Plaque, fillings, crowns, bridges, implants and soft tissues are preserved in situ. Macroscopic and microscopic detail of good quality is preserved for histological and morphometrical evaluation.
Article
Dentinal repair in the postnatal organism occurs through the activity of specialized cells, odontoblasts, that are thought to be maintained by an as yet undefined precursor population associated with pulp tissue. In this study, we isolated a clonogenic, rapidly proliferative population of cells from adult human dental pulp. These DPSCs were then compared with human bone marrow stromal cells (BMSCs), known precursors of osteoblasts. Although they share a similar immunophenotype in vitro, functional studies showed that DPSCs produced only sporadic, but densely calcified nodules, and did not form adipocytes, whereas BMSCs routinely calcified throughout the adherent cell layer with clusters of lipid-laden adipocytes. When DPSCs were transplanted into immunocompromised mice, they generated a dentin-like structure lined with human odontoblast-like cells that surrounded a pulp-like interstitial tissue. In contrast, BMSCs formed lamellar bone containing osteocytes and surface-lining osteoblasts, surrounding a fibrous vascular tissue with active hematopoiesis and adipocytes. This study isolates postnatal human DPSCs that have the ability to form a dentin/pulp-like complex.
Article
The requirement for new bone to replace or restore the function of traumatised, damaged, or lost bone is a major clinical and socioeconomic need. Bone formation strategies, although attractive, have yet to yield functional and mechanically competent bone. Bone tissue engineering has been heralded as the alternative strategy to regenerate bone. In essence, the discipline aims to combine progenitor or mature cells with biocompatible materials or scaffolds, with or without appropriate growth factors, to initiate repair and regeneration. This brief review outlines the concepts, challenges, and limitations in bone tissue engineering and the potential that could improve the quality of life for many as a result of interdisciplinary collaboration.
Article
Human postnatal bone marrow stromal stem cells (BMSSCs) have a limited life-span and progressively lose their stem cell properties during ex vivo expansion. Here we report that ectopic expression of human telomerase reverse transcriptase (hTERT) in BMSSCs extended their life-span and maintained their osteogenic potential. In xenogenic transplants, hTERT-expressing BMSSCs (BMSSC-Ts) generated more bone tissue, with a mineralized lamellar bone structure and associated marrow, than did control BMSSCs. The enhanced bone-forming ability of BMSSC-Ts was correlated with a higher and sustained expression of the early pre-osteogenic stem cell marker STRO-1, indicating that telomerase expression helped to maintain the osteogenic stem cell pool during ex vivo expansion. These results show that telomerase expression can overcome critical technical barriers to the ex vivo expansion of BMSSCs, and suggest that telomerase therapy may be a useful strategy for bone regeneration and repair.
Article
Mesenchymal stem cell populations have previously been identified in adult bone marrow and dental pulp that are capable of regenerating the bone marrow and dental pulp microenvironments, respectively. Here we show that these stem cell populations reside in the microvasculature of their tissue of origin. Human bone marrow stromal stem cells (BMSSCs) and dental pulp stem cells (DPSCs) were isolated by immunoselection using the antibody, STRO-1, which recognizes an antigen on perivascular cells in bone marrow and dental pulp tissue. Freshly isolated STRO-1 positive BMSSCs and DPSCs were tested for expression of vascular antigens known to be expressed by endothelial cells (von Willebrand factor, CD146), smooth muscle cells, and pericytes (alpha-smooth muscle actin, CD146), and a pericyte-associated antigen (3G5), by immunohistochemistry, fluorescence-activated cell sorting (FACS), and/or immunomagnetic bead selection. Both BMSSCs and DPSCs lacked expression of von Willebrand factor but were found to be positive for alpha-smooth muscle actin and CD146. Furthermore, the majority of DPSCs expressed the pericyte marker, 3G5, while only a minor population of BMSSCs were found to be positive for 3G5. The finding that BMSSCs and DPSCs both display phenotypes consistent with different perivascular cell populations, regardless of their diverse ontogeny and developmental potentials, may have further implications in understanding the factors that regulate the formation of mineralized matrices and other associated connective tissues.
Article
Periodontal diseases that lead to the destruction of periodontal tissues--including periodontal ligament (PDL), cementum, and bone--are a major cause of tooth loss in adults and are a substantial public-health burden worldwide. PDL is a specialised connective tissue that connects cementum and alveolar bone to maintain and support teeth in situ and preserve tissue homoeostasis. We investigated the notion that human PDL contains stem cells that could be used to regenerate periodontal tissue. PDL tissue was obtained from 25 surgically extracted human third molars and used to isolate PDL stem cells (PDLSCs) by single-colony selection and magnetic activated cell sorting. Immunohistochemical staining, RT-PCR, and northern and western blot analyses were used to identify putative stem-cell markers. Human PDLSCs were transplanted into immunocompromised mice (n=12) and rats (n=6) to assess capacity for tissue regeneration and periodontal repair. Findings PDLSCs expressed the mesenchymal stem-cell markers STRO-1 and CD146/MUC18. Under defined culture conditions, PDLSCs differentiated into cementoblast-like cells, adipocytes, and collagen-forming cells. When transplanted into immunocompromised rodents, PDLSCs showed the capacity to generate a cementum/PDL-like structure and contribute to periodontal tissue repair. Our findings suggest that PDL contains stem cells that have the potential to generate cementum/PDL-like tissue in vivo. Transplantation of these cells, which can be obtained from an easily accessible tissue resource and expanded ex vivo, might hold promise as a therapeutic approach for reconstruction of tissues destroyed by periodontal diseases.