You are on page 1of 6

Pulp Regeneration—Translational Opportunities

Pulp Stem Cells: Implication in Reparative Dentin Formation


Sasha Dimitrova-Nakov, DDS, PhD, Anne Baudry, PhD, Yassine Harichane, DDS, PhD,
Odile Kellermann, Dr es Sciences Naturelles, and
Michel Goldberg, DDS, PhD, Dr es Sciences Naturelles

Abstract
Many dental pulp stem cells are neural crest derivatives
essential for lifelong maintenance of tooth functions and
homeostasis as well as tooth repair. These cells may be
P ostnatal dental pulp contains heterogeneous cell populations responsible for its
maintenance, defense, and capacity of repair. The resident cells include stromal fi-
broblasts also named pulpoblasts by Baume (1), odonto-osteoprogenitors, neuronal
directly implicated in the healing process or indirectly and vascular cells, and inflammatory and immune system cells (2). The pulp responds
involved in cell-to-cell diffusion of paracrine messages to a variety of pathological injuries by a deposition of reparative dentin by pulp ‘‘pro-
to resident (pulpoblasts) or nonresident cells (migrating genitors’’ (2, 3). However, the origin, localization, and precise identity of odontogenic
mesenchymal cells). The identity of the pulp progenitors stem cells remain largely unknown. Identifying stem cells mobilized in response to pulp
and the mechanisms sustaining their regenerative ca- injury is a prerequisite to design alternative strategies for pulp healing and regeneration
pacity remain largely unknown. Taking advantage of and/or for endodontic treatment (3, 4).
the A4 cell line, a multipotent stem cell derived from Mitotic divisions and apoptosis constantly renew the pulp cell population, in
the molar pulp of mouse embryo, we investigated the contrast with odontoblasts and cells of the Hoehl’s layer, which are postmitotic cells.
capacity of these pulp-derived precursors to induce Pulpoblasts are implicated in the synthesis and secretion of an extracellular matrix
in vivo the formation of a reparative dentin-like struc- (ECM). Enzymatic cleavages followed by the reuptake of ECM remnants are prerequisite
ture upon implantation within the pulp of a rodent to promote dentin and/or pulp mineralization.
incisor or a first maxillary molar after surgical exposure. A few pulp cells are stem cells, probably less than 1% of the total cell population.
One month after the pulp injury alone, a nonmineralized According to Kenmotsu et al (5), approximately 0.40% of the pulp cells may be stem
fibrous matrix filled the mesial part of the coronal pulp cells (also referred to as the side population) when they are found in young rats,
chamber. Upon A4 cell implantation, a mineralized os- whereas only 0.11% is found in adult rats.
teodentin was formed in the implantation site without Variations in cell number and the respective distribution and functions have been
affecting the structure and vitality of the residual pulp recorded between the central part of the pulp and the periphery and also between the
in the central and distal parts of the pulp chamber. These radicular and coronal pulp (2). This article highlights the pulp complexity and how it is
results show that dental pulp stem cells can induce difficult to select genuine stem cell–based and cell-free approaches for pulp therapies.
the formation of reparative dentin and therefore consti-
tute a useful tool for pulp therapies. Finally, reparative Dental Pulp Stem Cells
dentin was also built up when A4 progenitors were per- Postnatal dental pulp stem cells (DPSCs) were firstly isolated from human teeth
formed by alginate beads, suggesting that alginate is a (6). They are largely neural crest–derived cells expressing genes that are also present
suitable carrier for cell implantation in teeth. (J Endod in embryonic stem cells but lacking expression of mesodermal genes. It is difficult to
2014;40:S13–S18) have a clear idea of the outcome of stem cells because of the fact that their origin
has not yet been elucidated although cell membrane markers and receptors specifically
Key Words identify them. Depending on the culture medium, they become odontoblasts, osteo-
Cell niches, dentin repair, osteodentin, pulpoblasts, pulp blasts, chondrocytes, adipocytes, neurons, and smooth muscles. Implanted with
stem cells Hap/tricalcium phosphate, the clones differentiate into odontoblast-like cells produc-
ing a dentin-like structure, which are widely used to produce reparative dentin. In the
presence of bone morphogenetic protein 2 and 4 (BMP2 and BMP4), DPSCs differen-
tiate into dentin-forming odontoblasts. This hypothesis is strengthened by the fact that
From the INSERM UMR-S 747, Equipe 5: Cellules souches,
Signalisation et Prions, Paris, France; Universite Paris Descartes, the BMP antagonist Noggin inhibits the capacity of DPSCs to differentiate into odonto-
Sorbonne, Paris, France; and Biomedicale des Saint Peres, Paris, genic cells as suggested by the lack of expression of dentin sialophosphoproteins.
France. Populations of DPSCs exhibit generic mesenchymal stem cell (MSC)-like proper-
This paper is based on a presentation from the International ties; display the ability to form colonies; and express in vitro osteoblastic, adipogenic,
Association for Dental Research (IADR) Pulp Biology and
Regeneration Group Satellite Meeting, which was held March
chondrogenic, and even neuronal markers (6–9). DPSCs share many similarities with
24–26, 2013 in San Francisco, California. MSCs of the bone marrow (BMSCs), which are the most studied stromal stem cell
Address requests for reprints to Dr Michel Goldberg, Bio- populations. More than 4,000 human genes are expressed either by BMSCs or
medicale des Saints Peres and INSERM UMR-S 747, 45 rue DPSCs (9). Dental stem cell populations also express different panels of stem cell sur-
des Saints Peres, 75006 Paris, France. E-mail address: michel. face markers used to characterize hematopoietic stem cells of the bone marrow
goldberg@parisdescartes.fr
0099-2399/$ - see front matter (BMSCs) (10). However, it is important to note that DPSCs and BMSCs do not have
Copyright ª 2014 American Association of Endodontists. the same embryonic origin. Cells derived from human or animal dental pulps have
http://dx.doi.org/10.1016/j.joen.2014.01.011 not been able to support hematopoiesis in transplantation assays (10, 11). DPSCs
are thought to contribute to reparative dentin formation, and it appears that they

JOE — Volume 40, Number 4S, April 2014 Pulp Stem Cells S13
Pulp Regeneration—Translational Opportunities
may correspond to heterogeneous populations of precursor cells or
represent distinct differentiation stages along the odontoblastic lineage.
Pulp stem cells are implicated in pulp repair (8). Nevertheless, the
formal demonstration that pulpal resident stem cells are actually the
reparative dentin-forming cells recruited in response to injury is still
lacking. The hypothesis that a subset of stem cells carried by the vascu-
lature replenishes the pulp after a lesion cannot be totally excluded.
Moreover, the responsiveness of the pulp provides a dynamic system
for tissue repair that may imply migration of stem cells from their resting
places to the site of injury. Undifferentiated mesenchymal/mesectoder-
mal cells present in the stroma and perivascular cells, such as Rouget’s
pericytes, have been proposed as potential progenitors mediating pulp
repair after destruction of the odontoblasts and the Hoehl’s subodonto-
blastic cell layer (12, 13). Advances in the identification of stem cell
markers are still needed to visualize stem/precursor cells in situ.

Different Dental Stem Cells


In addition to DPSCs, others stem cells have been obtained from
human dental tissues (deciduous teeth and the apical part of the papilla)
(6, 11, 14) and periodontal tissues (gingiva, cementum, alveolar bone,
and periodontal ligament) (15). Stem cells were also isolated from the
dental follicle. Other cellular candidates have been identified and impli-
cated as new tools in the formation of mineralized tissues. BMSCs gave
rise to osteoblast-like cells that generate a bone-like structure. DPSCs
are producing structures resembling dentin, whereas BMSCs create Figure 1. The multipotent pulp-derived A4 cell line cultured in monolayer for
bone condensation at some distance to capillaries (14). 15 days in an odonto-/osteogenic medium (bGP/AA/Dex) forms a mineralized
Dental stem cells are considered as a population of MSC-like cells. matrix as shown by the von Kossa staining. In absence of an inducer, the A4
Therefore, the markers that have been used for identifying MSCs were cells never differentiate spontaneously.
also instrumental for isolating dental stem cells. Subpopulations of DPSCs
or stem cells from human exfoliated deciduous teeth (SHED) expressing
c-kit/CD34/cell surface antigen expressed by stromal elements in human MSCs and Cell Migration
bone marrow (STRO-1) are considered as multipotent stem cells MSCs are found in bone marrow, cord blood, and dental pulp.
although c-kit and CD34 have also been shown to be markers for hemato- They differentiate into osteoblasts, chondroblasts, adipoblasts, fibro-
poietic cell lineages (11–19). Therefore, dental stem cells and non– blasts, myofibroblasts, and neuroblasts. The cells migrate from bone
dental stem cells sharing a high proliferation rate, multidifferentiation marrow, transit through the vasculature, and arrive at the affected tis-
ability, and easy accessibility are candidates for the development of sues. Chemotaxis implicates enzymes, phosphorylases, and proteins
tooth regeneration and/or tooth engineering (6, 8, 10). inducing cell polarization and directional movement. These cells take
origin in the bone marrow, and they are found in blood vessels. This
implies that they may penetrate through the open apex and colonize
Can Nonresident Migrating Cells the dental pulp.
Become Stem Cells? Some years ago, a limited number of stem cells were identified, and
In addition, 2 other groups of cells may potentially be stem cell marked differences were detected between embryonic and adult stem
candidates. These possibilities have been experimentally addressed. cells. Differences in gene expression, transcription factors, and growth
factors account for the dissimilarities noticed between embryonic and
adult lines. Adult stem cells alone are considered in this review within
The Origin and Role(s) of Nonresident Progenitor the frame of a therapeutic armamentarium. The number, origin, and
and Hematopoietic Cells in Pulp Repair stage of differentiation have increased, and selecting acceptable stem cells
Investigating the potential roles of nonresident progenitors and among a heterogeneous cell population becomes quite a hard task.
hematopoietic cells in the contribution of stem cells found in the dental
pulp, nonresident progenitors were suggested to contribute to repara-
tive dentinogenesis. Parabiosis was established between transgenic Genomic Stability
(freen fluorescent protein + [GFP+]) and wild-type (GFP ) mice to Immortalized human stem cells have been obtained from dental
ensure cross-circulation between the 2 mice. Pulp exposure and pulp. Genomic stability from transformed residing cells of ectomesen-
capping of the first maxillary molar were performed. GFP-positive cells chymal origin has the potential to differentiate into odontoblast-like
were detected in close association with reparative dentin formed at the cells. It is actually required that the cells and their derivatives maintain
site of pulp exposure of GFP-negative mice (20). their genomic stability. The presence of mosaicism and the accumula-
This observation shows the participation of migrating nonresident tion of karyotypic abnormalities have been reported within cultured
progenitor and hematopoietic cells in the formation of reparative cell subpopulations. Gradually, many of the cultured cells (70%)
dentin. Used for the cell migration the blood circulation and moving exhibit karyotypic abnormalities after some passages. The heteroge-
throughout the mesenchymal tissues, nonresident progenitor neous spectrum of abnormalities indicates a high frequency of chromo-
GFP + cells were clearly implicated in the reparative process. somal mutations that continuously arise upon extended

S14 Dimitrova-Nakov et al. JOE — Volume 40, Number 4S, April 2014
Pulp Regeneration—Translational Opportunities

Figure 2. The A4 clone behaves as a multipotent mesenchymal progenitor. Depending on the nature of the inducers, A4 cells are able to undergo odontogenic,
osteogenic, chondrogenic, or adipogenic differentiation in a mutually exclusive manner. Two other clonal pulpal cell lines, C5 and H8, appear as monopotent
progenitors, with a differentiation potential restricted to the odontoblastic program.

culture. Consequently, there is a need for careful analysis of the Hallmarks of a niche include the stem cell itself, stromal-
cytogenetic stability before undertaking any clinical therapies (21). supporting cells that interact directly with the stem cells via secreted
To obtain stable cell lines, immortalized dental papilla derived factors, and cell surface molecules (29). The ECM provides a struc-
cell lines were used. The MO6-G3 mouse-derived cell line (22), the tural support to the niche and allows the diffusion of mechanical and
cloned 3T6 cell (23), odontoblast-like cells immortalized by telome- chemical signals. In teeth, the location and the elements of the pre-
rase (24), and SV-40 immortalized cell lines (25–27) allowed us to sumptive niche providing the physiological environment for stem cell
obtain the lines, which are required for further studies. This is the self-renewal and odontogenic differentiation are unknown. It is
only way to study the signaling cascade leading to the differentiation assumed that multiple niches exist in teeth, but specific markers al-
of odontoblastic-like pulp cells. Even if it is not known yet to which lowing the accurate identification of stem cells within the pulp are
extent the immortalization procedure of cells or mice influences the lacking.
genes and protein expression, it seems impossible to circumvent this Many authors hypothesized a perivascular origin for stem cells.
strategic step. MSCs are associated with perivascular niches and they coexpress
many markers in common with pericytes. Some pericytes differentiate
Stem Cell Niches into specialized tooth mesenchyme-derived cells.
Some data suggest that pericytes could differentiate into osteo-
Stem cell behavior is regulated by a local microenvironment
blast-like cells, and odontogenic stem cells may reside in a perivascular
referred as to ‘‘the stem cell niche,’’ which is characterized by 3
niche (18). In this context, it is interesting to mention that many he-
essential properties (14, 28):
matopoietic stem cells and neuronal stem cells are localized close to
1. A niche provides an anatomic space where the stem cell number is the vascular network. Besides, alterations in ECM components and ma-
regulated. trix stiffness related to damage or aging may also provide mechanical
2. It is the place where a stem cell is instructed to control the mainte- signals that could have a profound impact on stem cell activity (29).
nance, quiescence, self-renewal, and recruitment toward differenti- Thus, it appears that the distribution of tissue stem cells is not random,
ation, fate determination, and long-term regenerative capacity. and, within the dental pulp, there are potentially several distinct niches
3. The niche will influence cell motility. of stem/progenitor cells (30). This implies that signals ensure their

JOE — Volume 40, Number 4S, April 2014 Pulp Stem Cells S15
Pulp Regeneration—Translational Opportunities

Figure 3. After 10 days, implantation of A4 cells in the surgically exposed dental pulp of the mouse incisor promotes the massive formation of reparative osteo-
dentin. The control group (nonimplanted with cells) exhibits no pulp repair.

survival and prevent their differentiation while maintaining their Implantation into the Mouse Incisor
responsiveness after pulp damage. Whether an endogenous pool of In the first in vivo pilot study, the A4 cells were implanted in the
stem cells associated with supportive stromal cells is mobilized at the mouse mandibular incisor, and this led to the formation of a mineral-
site of injury and/or whether attraction of migrating stem cell is neces- ized osteodentin within the dental pulp (Fig. 3). Because mouse incisor
sary to repopulate a niche and expand precursor cells at the appropriate is a continuously growing tooth, it was necessary to choose a model
site for dentin repair is unknown. Summarizing the conclusions of some closer to the human tooth physiology.
reports, it seems obvious that some dental pulp side populations are
implicated in the stimulation of angiogenesis, neurogenesis, and pulp
regeneration (12) although PCNA immunostaining and the alpha- Implantation into the Rat Molar
actin labeling characteristic of microvascularization never overlap in The small size of the mouse molars and teeth of limited growth
the dental pulp (7, 12). make experiment procedures difficult. In contrast, the rat molar pro-
vides an easier experimental approach involving the preparation of
tooth lesions (Fig. 4).
We performed surgical pulp exposure with or without cell implan-
The A4 Cell Line Induces the Formation tation in the first maxillary molar of 9-week-old Sprague-Dawley rats old
of Reparative Dentin In Vivo following institutionally approved protocols for animal experimentation
Dental stem cell research is still confronted with the lack of research. After anesthesia, a cavity was drilled on the mesial aspect of
precise knowledge related to the location and identity of the cells the tooth using a tungsten dental bur (ISO 006; Dentsply France SAS,
participating in reparative dentin formation upon tooth injury. In or- Montigny le Bretonneux, France) with a low-speed handpiece. Pulp
der to characterize the dental pulp progenitors and study the molec- perforation was accomplished by pressure with the tip of a steel probe.
ular and cellular mechanisms governing their differentiation, our This method avoids rolling the pulp around the dental burr. About 105
laboratory has obtained clonal cell lines from tooth germs of day A4 cells were collected in a tube and centrifuged to form a cell pellet
18 mouse embryos transgenic for an adenovirus-SV40 recombinant subsequently implanted into the pulp of 14 rat molars (31, 32).
plasmid (pK4) (25). Clonal cell lines derived from the dental pulp Two, 14, and 30 days after surgery, the rats were euthanized. Block
provide valuable tools to answer several fundamental questions such sections including the hemimaxillaries were dissected, fixed in a 4%
as the localization of the stem cells niches. Combining in vitro and paraformaldehyde solution, EDTA demineralized, and included in
in vivo experimental approaches, the answers to these questions paraffin. Seven-micrometer-thick sections were collected on glass
lead to a better understanding of the potential of stem cells for tooth slides and observed after Masson trichrome staining.
repair (26, 27).
Whether stem cell phenotypes result from genuine multipotent
cells or from the coexistence of distinct progenitors is still an open
The Sham Group
question. Three clonal pulp precursor cell lines (A4, C5, and H8) A surgical pulp exposure was performed in the first rat molars.
were induced to differentiate toward the odonto-/osteogenic, chondro- Two days after the injury, the pulp tissue appeared normal (Fig. 4A
genic, or adipogenic program (Figs. 1 and 2). Implanted into mandib- and B). However, inflammatory cells were recruited near the implanta-
ular incisors or calvaria of adult mice, the A4 clone behaves as a tion site (Fig. 4C). One month after the surgery, the mesial pulp cham-
multipotent cell. In contrast, the C5 and H8 clones displayed a more ber was fibrotic but not mineralized (Fig. 4B).
restricted potential. Altogether, isolation of these clonal lines shows
the coexistence of multipotential and restricted-lineage progenitors in The Experimental A4 Cell Implantation Group
the mouse pulp, unraveling the specificities of the different types of After 1 month, the mesial part of the pulp chamber was massively
pulp progenitors (27). To explore for the first time the potential of filled by mineralized osteodentin seen under a calciotraumatic line
pulp-derived ‘‘stem’’ cells to improve dental repair upon injury, we (Fig. 4D). Thus, A4 cells have the capacity after implantation in a rat
developed several in situ approaches. molar and in the absence of any carrier or biomolecule to promote

S16 Dimitrova-Nakov et al. JOE — Volume 40, Number 4S, April 2014
Pulp Regeneration—Translational Opportunities

Figure 4. After a sham pulp exposure in the first maxillary rat molar, inflammatory processes are limited after (A) 2 days and (B) 28 days. (C) In the control
(nonimplanted with cells), a nonmineralized fibrous reaction is seen in the mesial part of the coronal pulp. (D) In contrast, A4 implantation promotes the massive
formation of reparative dentin in the mesial part of the coronal pulp beneath the calciotraumatic lines (arrows). Implantation of alginate beads loaded by A4 cells
induces the formation of a dentin barrier (db and asterisk) located between the mesial and central part of the pulp chamber. (E and F) A fibrous matrix (fm) is
seen in the mesial pulp chamber.

efficient dentin repair (26, 27). It is of note that the inflammatory dental therapies. The present results indicate that the direct implanta-
response normally observed upon pulp injury is not enhanced by the tion of mouse progenitor cells in the dental pulp of a rat molar leads
implantation of mouse-derived pulpal cells in a rat molar. to the formation of reparative osteodentin.
It is important to determine (1) whether precursor cells reintro-
duced in a pulpal ‘‘natural’’ environment differentiate into osteoodon-
Implantation of Alginate Beads Loaded togenic cells or whether the implanted cells recruit resident pulp stem
with or without A4 Cells cells toward osteoodontogenic differentiation and indirectly promote
In order to improve the implantation protocol (ie, precisely visu- the formation of the dentinal bridge. Future prospects will determine
alize the implantation site and control the number of cells implanted whether the implanted progenitor cells are directly involved in the for-
within the pulp), we used as carrier alginate beads. Alginate, a linear mation of the reparative dentin or whether they induce the recruitment
copolymer of anionic polysaccharide, was selected as a suitable and and differentiation of host progenitor cells.
potentially nontoxic biomaterial because its pH is neutral during and In conclusion, our preclinical experimental approach paves the
after gelation. Moreover, this irreversible hydrocolloid is biocompat- way for the development of cellular therapies after pulp injury. The
ible and biodegradable (33). Alginate beads alone or loaded with long-term goal should provide new clinical strategies to restore
cells were implanted in exposed pulp. After 1 month, the group im- the functionality of an injured tooth by using pulp stem cells.
planted with the alginate beads alone was comparable with the sham
group. Conversely, 1 month after the implantation of alginate beads
loaded with 3.103 A4 cells, we observed the formation of reparative Acknowledgments
osteodentin appearing as a dentin plug located in the isthmus located The author denies any conflicts of interest related to this study.
between the mesial and central parts of the pulp chambers (Fig. 4E
and F). These preliminary data using alginate beads as a cell carrier
suggest that this biomaterial is suitable for cell implantation inducing References
an efficient pulp repair. Hence, we get the first evidence that implan- 1. Baume LJ. Biology of pulp and dentin: a historic, terminologicotaxonomic, histolog-
tation of pulpal precursor cells within the pulp promotes the forma- ic-biochemical, embryonic and clinical survey. In: Myers HM, ed. Monographs in
tion of a robust dentin barrier separating the residual pulp from the Oral Science. Basel: S Karger AG; 1980:1–220.
reparative area. 2. Goldberg M, Smith A. Cells and extracellular matrices of dentin and pulp. A
biological basis for repair and tissue engineering. Crit Rev Oral Biol Med 2004;
15:1327.
3. Tziafas D, Smith AJ, Lesot H. Designing new treatment strategies in vital pulp therapy.
Conclusions and Perspectives J Dent 2000;28:77–92.
The stem cells appear as tools to get a better understanding of the 4. Balic A, Mina M. Characterization of progenitor cells in pulps of murine incisors.
cellular mechanisms of pulp repair. They display innovating potential in J Dent Res 2010;89:1287–92.

JOE — Volume 40, Number 4S, April 2014 Pulp Stem Cells S17
Pulp Regeneration—Translational Opportunities
5. Kenmotsu M, Matsuzaka K, Kokubu E, et al. Analysis of side population cells derived 20. Frozoni M, Zaia AA, Line SR, et al. Analysis of the contribution of nonresident pro-
from dental pulp tissue. Int Endod J 2010;43:1132–42. genitor cells and hematopoietic cells to reparative dentinogenesis using parabiosis
6. Gronthos S, Mankani M, Brahim J, et al. Postnatal human dental pulp stem cells model in mice. J Endod 2012;38:1214–9.
(DPSCs) in vitro and in vivo. Proc Natl Acad Sci U S A 2000;97:13625–30. 21. Duailibi MT, Kulikowski LD, Duailibi SE, et al. Cytogenetic instability of dental pulp
7. Batouli S, Miura M, Brahim J, et al. Comparison of stem-cell mediated osteogenesis stem cell lines. J Mol Histol 2012;43:89–94.
and dentinogenesis. J Dent Res 2003;82:976–81. 22. MacDougall M, Thiemann F, Ta H, et al. Temperature-sensitive simian virus 40 large
8. Huang GT. Dental pulp and dentin tissue engineering and regeneration—advance- T antigen immortalization of murine cell cultures: establishment of clonal odonto-
ment and challenge. Front Biosci (Elite Ed) 2012;3:788–800. blast cell line. Connect Tissue Res 1995;33:97–103.
9. Shi S, Robey PG, Gronthos S. Comparison of human dental pulp and bone marrow 23. Hanks CT, Sun ZL, Fang DN, et al. Cloned 3T6 cell line from CD-1 mouse fetal molar
stromal stem cells by cDNA microarray analysis. Bone 2001;29:532–9. dental papillae. Connect Tissue Res 1998;37:233–49.
10. Huang GT, Gronthos S, Shi S. Mesenchymal stem cells derived from dental tissues vs. 24. Hao J, Narayanan K, Ramachandran A, et al. Odontoblast cells immortalized by telo-
those of other sources: their biology and role in regenerative medicine. J Dent Res merase produce mineralized dentin-like tissue both in vitro and in vivo. J Biol
2009;88:792–806. Chem 2002;277:19976–81.
11. Gronthos S, Brahim J, Li W, et al. Stem cell properties of human dental pulp stem 25. Priam F, Ronco V, Locker M, et al. New cellular models for tracking the odontoblast
cells. J Dent Res 2002;81:531–5. phenotype. Arch Oral Biol 2005;50:271–7.
12. Ishizaka R, Hayashi Y, Iohara K, et al. Stimulation of angiogenesis, neurogenesis and 26. Harichane Y, Hirata A, Dimitrova-Nakov S, et al. Pulpal progenitors and dentin
regeneration by side population cells from dental pulp. Biomaterials 2013;34: repair. Adv Dent Res 2011;23:307–12.
1888–97. 27. Lacerda-Pinheiro S, Dimitrova-Nakov S, Harichane Y, et al. Concomitant multipotent
13. Fitzgerald M, Chiego DJ Jr, Heys DR. Autoradiographic analysis of odontoblast replace- and unipotent dental pulp progenitors and their respective contribution to miner-
ment following pulp exposure in primate teeth. Arch Oral Biol 1990;35:707–15. alised tissue formation. Europ Cells Materials 2012;23:371–86.
14. Ema H, Suda T. Two anatomically distinct niches regulate stem cell activity. Blood 28. Huang GT, Sonoyama W, Liu Y, et al. The hidden treasure in apical papilla: the po-
2012;120:2174–81. tential role in pulp/dentin regeneration and BioRoot engineering. J Endod 2008;34:
15. Miura M, Gronthos S, Zhao M, et al. SHED: stem cells from human exfoliated decid- 645–51.
uous teeth. Proc Natl Acad Sci U S A 2003;100:5807–12. 29. Engler AJ, Sen S, Sweeney HL, et al. Matrix elasticity directs stem cell lineage spec-
16. Seo B, Miura M, Gronthos S, et al. Investigation of multipotent postnatal stem cells ification. Cell 2006;126:677–89.
from human periodontal ligament. Lancet 2004;364:149–55. 30. Feng J, Mantesso A, De Bari C, et al. Dual origin of mesenchymal stem Cells contrib-
17. Janebodin K, Horst OV, Ieronimakis N, et al. Isolation and characterization of neural uting to organ growth and repair. Proc Natl Acad Sci U S A 2011;108:6503–8.
crest-derived stem cells from dental Pulp of Neonatal Mice. PLoS One 2011;6:e27526. 31. Ohshima H. Ultrastructural changes in odontoblasts and pulp capillaries following
18. Shi S, Gronthos S. Perivascular niche of postnatal mesenchymal stem cells in human cavity preparation in rat molars. Arch Histol Cytol 1990;53:423–8.
bone marrow and dental pulp. J Bone Miner Res 2003;18:696–704. 32. Decup F, Six N, Palmier B, et al. Bone sialoprotein-induced reparative dentinogen-
19. Sonoyama W, Liu Y, Yamaza T, et al. Characterization of apical papilla and its esis in the pulp of rat’s molar. Clin Oral Investig 2000;4:110–9.
residing stem cells from human immature permanent teeth—a pilot study. 33. Kl€ock G, Pfeffermann A, Ryser C, et al. Biocompatibility of mannuronic acid-rich al-
J Endod 2008;34:166–71. ginates. Biomaterials 1997;18:707–13.

S18 Dimitrova-Nakov et al. JOE — Volume 40, Number 4S, April 2014

You might also like