You are on page 1of 13

Periodontology 2000, Vol.

51, 2009, 239–251  2009 John Wiley & Sons A/S


Printed in Singapore. All rights reserved PERIODONTOLOGY 2000

Stem cells and future


periodontal regeneration
N I -H U N G L I N , S T A N G R O N T H O S & P. M A R K B A R T O L D

Periodontitis is an inflammatory disease that causes entiate into a number of specialized daughter cells to
pathological alterations in the tooth-supporting tis- perform specific function(s) (95, 115). A stem cell can
sues, potentially leading to tooth loss. National sur- divide asymmetrically, in which case one of the two
veys have shown that the majority of adults in the daughter cells retains the stem cell characteristics
population suffer from moderate periodontitis, with while the other is destined for specialization under
up to 15% of the population being affected by severe specific conditions (83). A pluripotent stem cell can
generalized periodontitis at some stage in their lives differentiate into all cell types of the body, whereas a
(2, 17). The significant burden of periodontal disease multipotent stem cell can differentiate along multi-
and its impact on general health and patient quality lineages into many different cell types (95). The po-
of life point to the need for more effective manage- tential for self-renewal vs. differentiation is governed
ment of this condition (113, 143). Several procedures by extracellular signals coupled to intracellular
have been attempted to achieve periodontal regen- signaling cascades (140). Information that specifies
eration, including root surface conditioning, bone the fate of stem cells is encoded by the presence or
graft placement, guided tissue regeneration and absence of signals, and also by the combinations,
growth factor application. However, current regen- localization, level and timing of these signals.
erative procedures that are used either alone or in There are two main types of stem cells – embryonic
combination have limitations in attaining complete stem cells and adult stem cells – which are classified
and predicable regeneration, especially in advanced according to their origin and differentiation potential
periodontal defects (108, 109, 123, 139, 146). Recent (Table 1). Human embryonic stem cells, derived from
advances in stem cell biology and regenerative the inner cell mass of blastocysts, are pluripotent
medicine have presented opportunities for tissue stem cells capable of differentiating into cells of all
engineering as well as gene-based approaches in three germ layers of the adult body (131). Human
periodontal therapy (60, 63, 70, 73, 138). The follow- embryonic stem cells have been derived primarily
ing discussion reviews our current understanding of from spare blastocysts created by in vitro fertiliza-
stem cells and their potential application in regen- tion. Human embryonic stem cell lines are unique in
erative periodontal therapy. Major progress in stem that they can be maintained in an undifferentiated
cell biology in the dental context is highlighted and state in vitro for an indefinite period of time, while
the challenges in translating stem cell research into retaining their ability to differentiate into all types of
clinical practice are identified. specialized cells in the body (57, 107, 131). These cells
have been demonstrated to produce approximately
200 different types of cell within the adult body but,
Definition of stem cells and their because of ethical concerns, have not been tested for
types their ability to participate in human embryonic
development or to contribute to germ lines in vivo. It
Stem cells are the foundation cells for every organ is estimated that at least 225 human embryonic stem
and tissue in the body, including the periodontium cells lines, including some with genetic disorders,
(129, 130). A stem cell has two defining characteris- have been generated in different laboratories world-
tics: (i) the ability for indefinite self-renewal to give wide (47, 51, 100). The use of human embryonic stem
rise to more stem cells; and (ii) the ability to differ- cells for clinical therapy is a relatively new endeavor

239
Lin et al.

Table 1. Different stem cell populations and their differentiation potential


Development stage Type of stem cell Differentiation potential
Blastocyst Embryonic stem cells Pluripotent
Derived from the inner cell mass of the
pre-implantation embryo
Fetus Embryonic germ cells Pluripotent
Derived from primordial germ cells iso-
lated from the embryonal gonad
Adult Embryonal carcinoma cells Pluripotent
Derived from primordial germ cells in
embryonic gonad and usually found as
components of testicular tumors in adults
Adult stem cells Multipotent
Derived from ectodermal and mesoder-
mal organs of adults
Adult cells that have undergone nuclear Totipotent
transformation
Adult cells that can be induced to an Inducible pluripotent
embryonic stem cell phenotype

and, currently, this development has been hampered 125), have drawn intense debate regarding the source
by ethical concerns (145). of human oocytes ⁄ embryos and the use of oocyte
In 2006, cells with properties similar to those of progenitors in the adult ovary (26). Furthermore,
embryonic stem cells, termed induced pluripotent somatic cell nuclear transfer has yet to be success-
stem cells, were created from specialized somatic fully demonstrated in humans, and the pluripotent
cells via the forced over-expression of four key factors cells resulting from the fusion of somatic cells with
(126, 127). The authors reported that when plasmids embryonic stem cells are often tetraploid with limited
containing Oct3 ⁄ 4, Sox2, cMyc and Klf4 genes were practical application. By contrast, the technology
introduced into murine fetal fibroblasts, the cells used to generate induced pluripotent stem cells can
were reprogrammed to an embryonic-like state. create patient-specific and disease-specific cell lines
These murine-induced pluripotent stem cells exhibi- for research purposes with fewer ethical concerns,
ted the morphology and growth properties of murine but this method of reprogramming is still inefficient
embryonic stem cells although they had different and many fundamental questions remain unan-
gene-expression and DNA-methylation profiles swered. Thus, research on human pluripotent cells
(Table 1). Follow-up reports with improved repro- derived using different methods should be continued
gramming techniques have yielded more embryonic to identify the best method for regenerative therapy
stem cell-like cells in mice, with the reprogrammed (56).
cells contributing to every cell type in the body (77, Adult, or tissue-specific, stem cells are found in the
97, 141). Human counterparts of murine-induced majority of fetal and adult tissues. They have been
pluripotent stem cells have subsequently been gen- derived from tissues that continually replenish
erated using various combinations of Oct3 ⁄ 4, Sox2, themselves [e.g. peripheral blood (3, 116), dermis
cMyc, Klf4, Nanog, LIN28, hTERT and SV40 large-T (134, 153) and gastrointestinal epithelium (12)] as
factors (75, 99, 126, 155). Although the mechanism by well as from tissues with lower regenerative capacity
which each of these factors restores pluripotency is [e.g. brain (117)]. Adult stem cells are thought to
not clearly understood, human-induced pluripotent function in long-term tissue maintenance and ⁄ or
stem cells attract immense interest from stem cell repair by replacing cells that are either injured or lost
researchers because of the lack of ethical issues (71). They are generally multipotent stem cells that
involving the use of human oocytes or embryos. can form a limited number of cell types corre-
Earlier techniques to restore pluripotency to human sponding with their tissues of origin, although some
somatic cells, involving somatic cell nuclear transfer studies have suggested that they are more versatile
into enucleated oocytes (144) or fusion of somatic and can develop into many other cell types than
cell nuclei with human embryonic stem cells (25, previously expected (15, 68, 80). The most common

240
Stem cells and future periodontal regeneration

source of adult stem cells is the bone marrow, which Bone marrow mesenchymal stem cells are the most
contains hematopoietic stem cells (132) and bone widely studied mesenchymal stem cells because they
marrow stromal cells or mesenchymal stromal ⁄ stem are easily accessible in quantities appropriate for
cells (29, 52). Hematopoietic stem cells were the clinical applications (19, 20, 46). These cells are clo-
first stem cells to be successfully used in therapies, nogenic and have demonstrated the ability to form
particularly in the treatment of blood malignancies bone and cartilage in vivo (101, 102). Because of their
and immunodeficiency syndromes, but they are differentiation potential, bone marrow mesenchymal
not capable of giving rise to supporting connective stem cells have been used in various phases of clinical
tissues (65). Mesenchymal stem cells, by contrast, are application, particularly in orthopedics (21, 53). Mes-
a cell type of interest with the therapeutic potential enchymal stem cells have also been shown to form
to treat a range of musculoskeletal abnormalities, cementum, periodontal ligament and alveolar bone
cardiac diseases and immune abnormalities (65). in vivo after implantation into periodontal defects in
This review will focus on the properties and function beagle dogs (50, 63), suggesting that bone marrow may
of mesenchymal stem cells as potential candidates be a useful source of mesenchymal stem cells for
for periodontal regeneration (50, 63). periodontal regeneration. Apart from the bone mar-
row, mesenchymal stem cell-like cells have also been
identified in many other tissues, including adipose
Human mesenchymal stem cells tissue, muscle, peripheral blood, fetal pancreas and
liver (6, 18, 54, 67, 157). Adipose tissue-derived mes-
Mesenchymal stem cells were initially identified in enchymal stem cells exhibit stable growth kinetics
rodent marrow as colony-forming unit fibroblasts in vitro and display a multilineage differentiation
capable of forming bone, cartilage and fat, as well as capacity (including osteogenesis, chondrogenesis and
reconstituting the hematopoietic microenvironment adipogenesis) which is similar to that of bone marrow
(37, 38). Colony-forming unit fibroblasts have sub- mesenchymal stem cells (42, 59, 74, 159). As adipose
sequently been detected in many species, including tissue can be obtained by using less-invasive methods
humans (19, 20, 46, 101), and the formation of col- and in larger quantities than bone marrow cells, the
ony-forming unit fibroblasts is considered to be a use of adipose-derived mesenchymal stem cells as an
distinguishing feature of mesenchymal stem cells. alternative to bone marrow mesenchymal stem cells
Mesenchymal stem cells have also been characterized is very appealing. Indeed, at least one study has
both morphologically and immunophenotypically demonstrated that eight weeks after implantation
using different cell-surface markers. Mesenchymal of adipose-derived stem cells into rat periodontal de-
stem cells occur as either large and flat cells or as fects, new periodontal ligament-like and alveolar
elongated and fibroblastic cells, but the functional bone-like structures were formed, implying that
significance of this diverse morphology is unclear (9). adipose-derived stem cells could promote periodontal
Phenotypically, mesenchymal stem cells can express regeneration in vivo (133). The abundance of human
the characteristics of endothelial, perivascular, neu- lipoaspirates, coupled with low host morbidity in their
ral, bone or muscle cells, and also express a range of procurement, makes them a unique stem cell reservoir
surface markers (including CD49a ⁄ CD29, CD44, for regenerative periodontal therapy.
STRO-1, CD90, CD105, CD106, CD146, CD140b,
CD166 and CD271). This suggests a common link
between different cell types because most of these Stem cells in dental and
markers are expressed by all mesenchymal stem cells periodontal tissues
(45, 46, 101, 104). However, clonal studies have
highlighted the heterogeneous nature of these cells, In addition to nondental stem cells, the potential of
given that different mesenchymal stem cell colonies dental mesenchymal stem cell-like cells for peri-
demonstrate different proliferative and developmen- odontal regeneration and repair has been extensively
tal capacities both in vitro and in vivo (46, 67, 85, studied in recent years. The ability of mesenchymal
101). Hence, characterization of candidate cells has stem cells to differentiate into multiple specialized
involved the combined evaluation of their surface cell types in many adult tissues (including dental
maker expression as well as of their proliferative tissues) has made them a promising cell source for
ability (to form colonies) and developmental poten- use in periodontal regeneration.
tials (to differentiate into osteoblasts, adipocytes and The first human dental stem cells were isolated
chondrocytes) (11, 101, 103). from dental pulp tissue of extracted third-molar teeth

241
Lin et al.

and were characterized relative to bone marrow have been shown to give rise to dentin tissue in
mesenchymal stem cells (43). These dental pulp stem immunocompromised mice, and to generate a
cells were found to be highly proliferative, clonogenic root ⁄ periodontal complex capable of supporting a
cells capable of differentiating into odontoblast-like porcelain crown after transplantation with perio-
cells and forming dentin ⁄ pulp-like complex when dontal ligament stem cells in minipigs (119, 120).
implanted into immunocompromised mice (41, 43). These findings provide support for the use of com-
Subsequently, human mesenchymal stem cells were bined mesenchymal stem cell populations in root
isolated from exfoliated deciduous teeth and were and periodontal regeneration.
noted to induce bone and dentin formation in vivo Mesenchymal progenitor cells have also been iso-
(82). Periodontal ligament stem cells were first iso- lated from the dental follicle of human third-molar
lated in 2004 and have been shown to give rise to teeth (84). These fibroblast-like, colony-forming and
adherent clonogenic clusters resembling fibroblasts plastic-adherent cells express stem cell markers
(110). They are capable of developing into adipocytes, (STRO-1 and nestin) and can be maintained in culture
osteoblast-like cells and cementoblast-like cells for at least 15 passages. STRO-1-positive dental fol-
in vitro as well as producing cementum-like and licular progenitors have been shown to differentiate
periodontal ligament-like tissues in vivo (39, 110, into cementoblasts in vitro (64, 148) and to form
112). Recent studies have also shown their ability to cementum in vivo (48). The finding that immortalized
differentiate into neuronal precursors (24, 128). dental follicle cells can generate periodontal liga-
Periodontal ligament stem cells also express an array ment-like tissue after in vivo implantation (151) im-
of cementoblast and osteoblast markers as well as the plies that dental follicular progenitor cells may be a
STRO-1 and CD146 antigens, which are found on useful research tool for studying periodontal ligament
dental pulp stem cells and bone marrow mesenchy- formation and for regenerative periodontal therapies.
mal stem cells (43, 135). These findings indicate that While the use of both dental and nondental mes-
periodontal ligament stem cells have properties enchymal stem cells for tooth formation and perio-
similar to those of dental pulp stem cells and bone dontal regeneration are promising, mesenchymal cell
marrow mesenchymal stem cells, and this may rep- populations from different tissues display distinct
resent another mesenchymal stem cell-like popula- biological properties (27). Even if they carry common
tion (Fig. 1). genetic markers, they are likely to be conditioned by
In 2006, stem cells from the apical papilla of hu- their specific microenvironment and to be commit-
man teeth were isolated (1, 120). These cells form ted towards a specific differentiation pathway (87,
adherent clonogenic clusters and, similar to other 88). This notion is supported by the observation that
mesenchymal stem cell populations, are capable of bone marrow mesenchymal stem cells display a
differentiating into adipocytes and odontoblasts ⁄ lower odontogenic potential than dental mesenchy-
osteoblasts in vitro. Stem cells from the apical papilla mal stem cells (156). In this context, it remains to be
determined which source of dental mesenchymal
stem cells will be most suitable for regenerative
Mesenchymal stem cell collection periodontal therapy. The use of dental follicular cells
is likely to be limited by the availability of tooth
germs, and nondental stem cells may require the
Tooth
Pulp transfer of genes to enhance their odontogenic
Bone marrow Periodontal potential. Although the best stem cell source is yet to
ligament
be identified, the prospect of using these cells for
regeneration represents a step forward in the devel-
opment of more predictable biologically based
Magnetically activated cell sorting therapies for the periodontium.
(MACS) or Dynal bead selection
using STRO-1 antibody

Potential uses of stem cells in


+ –
periodontal regeneration

Fig. 1. Schematic illustration of the isolation of mesen-


Stem cells have been used extensively in many
chymal stem cells from bone marrow, dental pulp and medical disciplines for the repair and ⁄ or regenera-
periodontal ligament. tion of defective tissues and organs such as bone (53),

242
Stem cells and future periodontal regeneration

cartilage (86), heart (122) and spinal cord (81). In by reiteration of tooth development based on epi-
dentistry, the identification of mesenchymal stem thelial–mesenchymal tissue recombination or by
cell-like populations from both dental and nondental seeding of cells on biomaterial scaffolds (32, 63, 89–
tissues has presented exciting possibilities for the 92, 98, 120, 133). The tissue-recombination technique
application of tissue engineering as well as gene- aims to replicate key reciprocal interactions between
based therapies. These techniques have the potential the dental epithelium and the ectomesenchyme
to lead to the development of novel strategies for during odontogenesis to regenerate the periodon-
regenerative periodontal therapy. tium. For example, following the combination of
embryonic epithelial and mesenchymal cells derived
from mouse fetuses, the re-associated tooth germs
Periodontal tissue engineering
result in tooth-like structures including roots, peri-
Tissue engineering is a contemporary area of science odontal ligament and alveolar bone in vivo (55, 91).
based on the principles of cell biology, developmen- Furthermore, the combination of oral epithelium
tal biology and biomaterials science to develop new with nondental-derived mesenchyme (e.g. embryonic
procedures and biomaterials to replace lost or dam- stem cells, neural stem cells and adult bone-marrow-
aged tissues (69, 93, 114). The main requirements for derived cells), results in the formation of both tooth
producing an engineered tissue are appropriate pro- crown and bone in vivo (96). Several studies have
genitor cells, signaling molecules, an extracellular reported that engineered tooth primordia give rise to
matrix or carrier construct and an adequate blood proper tooth development after being transferred
supply (114, 121). Although tissue engineering into the adult mandible (32, 91, 96), indicating that
encompasses many different strategies, including cell cultured stem cells can replace lost or damaged
injection, tissue culturing, porous and injectable dental structures following transplantation into the
scaffolds, and three-dimensional printing (7, 114), adult oral cavity. However, the periodontal structures
this field is most frequently linked to the paradigm regenerated using tissue-recombination techniques
of cell delivery within biocompatible scaffolds (70, are not formed in isolation from other dental tissues
138). In this context, a potential tissue-engineering and this may pose problems for implantation into
approach to periodontal regeneration involves the periodontal defects. In addition, there is currently
incorporation of progenitor cells and instructive no suitable substitute for the embryonic epithelial
messages in a prefabricated three-dimensional con- compartment of the engineered tooth germ, and the
struct and subsequent implantation of the construct use of human embryonic tissues for periodontal
into the defect site (7) (Fig. 2). This approach may engineering may limit the practical application of this
overcome a major shortcoming associated with approach.
conventional regenerative procedures because the The technique of cell transplantation, within
recruitment of progenitor cells and growth factors is three-dimensional matrices, to regenerate the perio-
negated and healing can be enhanced. dontium has been extensively studied. Nondental
Tissue-engineering strategies have been applied to stem cells (such as bone marrow mesenchymal stem
reconstruct damaged periodontal apparatus, either cells and adipose-derived stem cells) have been

Fig. 2. Schematic representation of the concept of tissue that this is an illustration only and the radiographs do not
engineering for periodontal regeneration. A periodontal represent an actual clinical outcome; they are used to
defect may be filled with a biodegradable scaffold con- illustrate a potential outcome based on the concepts of
taining specific molecular instructional messages and tissue engineering.
stem cells to result in full regeneration of the defect. Note

243
Lin et al.

investigated as cell sources for periodontal regener- entiated into a variety of autologous cell types
ation. Autologous bone marrow mesenchymal stem (including dental stem cells and dental epithelium)
cells and adipose-derived stem cells can regenerate for in vivo administration.
alveolar bone and periodontal ligament-like struc-
tures after transplantation in vivo, and this provides
Stem cell-mediated gene therapy
support for the use of these cells in regenerative
periodontal therapy (63, 72, 133). Studies have also Gene therapy relies on genetic engineering involving
used dental stem cells to achieve periodontal regen- molecular techniques to introduce, suppress or
eration both in vitro (136, 137) and in vivo (32, 44, 66, manipulate specific genes, thereby directing an
120, 152). In these studies, progenitor cells were individualÕs own cells to produce a therapeutic agent
seeded directly onto biomaterial scaffolds (e.g. poly- (4). In the context of periodontal regeneration, gene
glycolic acid, calcium phosphate material, collagen therapy seeks to optimize the delivery of agents, such
sponges) followed by transplantation into periodon- as growth factors, to periodontal defects so that the
tal defects in animal models (such as mice, rats, dogs, limitations associated with the efficacy of topical
pigs and sheep). Seeding of tooth bud cells (which application of these factors (e.g. a short duration of
presumably include some progenitor cells) on bio- action) can be overcome (106). Gene-delivery tech-
degradable scaffolds resulted in the formation of new niques have been used in periodontal ligament and
periodontal ligament and bone tissues after implan- alveolar bone regeneration in rats (60, 92), and
tation into the omenta of adult rat hosts and at the embryonic stem cells have been shown to express
site of previously lost teeth (32, 152). A novel study tooth-initiation genes in mice (96, 149). Two poten-
has reported the combination of swine stem cells tial strategies for delivering therapeutic transgenes
from the apical papilla with periodontal ligament into human recipients are: (i) the direct infusion of
stem cells to regenerate root and periodontal struc- the gene of interest using viral or nonviral vectors
ture in minipigs (117). The swine stem cells from the in vivo; and (ii) the introduction of the gene into
apical papilla were seeded into a root-shaped scaffold delivery cells (often a stem cell) outside the body
that was then wrapped with gelfoam containing ex vivo followed by transfer of the delivery cells back
porcine periodontal ligament stem cells. After a into the body (160). The inherent proliferative and
three-month healing period, periodontal ligament pluripotent capabilities of stem cells may offer life-
tissue was noted to form around this bio-root struc- long opportunities for treatment by repairing,
ture and appeared to have a natural relationship with replacing or regenerating the damaged tissues.
the surrounding bone. Regenerated root structure The use of adenoviral vectors to enable the over-
containing new cementum and periodontal ligament expression of growth-promoting molecules, such as
has been reported to occur by culturing swine dental platelet-derived growth factor and bone morphoge-
bud cells on a cylinder scaffold and then grafting it netic protein-7, has been investigated for its potential
back into the alveolar socket (66). These promising in periodontal regeneration (5, 60, 61). Sustained re-
results reported in animal models using dental stem lease of bone morphogenetic protein-7 and platelet-
cells are paving the way for human regenerative derived growth factor by transformed cells implanted
periodontal therapy. into experimental periodontal defects results in en-
Overall, these findings demonstrate the feasibility hanced regeneration of bone and cementum (33, 60).
and potential of using dental and nondental stem The use of such technology with dental stem cells
cells for functional periodontal and tooth regenera- may offer an alternative to conventional methods as a
tion. An ideal source of stem cells for periodontal result of their ability to provide a renewable source of
regeneration should be nonimmunogenic, highly growth factor release for regeneration. STRO-1-
proliferative, easy to harvest, resilient to prolonged selected rat dental pulp stem cells transduced with
processing and have the ability to differentiate into bone morphogenetic protein-2 demonstrate en-
desired cell types (40). Recent findings confirming hanced odontogenic differentiation compared with
that human adult dermal fibroblasts can be repro- nontransduced cells (147). However, much work is
grammed to pluripotency (75, 99, 126, 155) may en- still needed to optimize the number of cells that are
able researchers to overcome immune rejection and virally transduced and to maximize the duration and
to produce large quantities of cells for regenerative extent of gene expression. Further research is also
periodontal therapy. Human-induced pluripotent needed to address the potential risks of viral recom-
stem cells can be derived from a dental source (e.g. bination and immune responses towards viral anti-
gingival fibroblasts of the patient) and then differ- gens (36, 118), which may ultimately determine the

244
Stem cells and future periodontal regeneration

success of gene-transfer techniques in periodontal safely and consistently. Furthermore, as cell-culture


regeneration. Many challenges remain in bringing medium often requires xenogenic products (such as
stem cell research to clinical practice in humans, and fetal bovine serum or mouse feeder layers), cell
it is important to overcome these challenges to fulfill cultures may not be completely free of pathogens
our goal of treating periodontal diseases with stem and infectious risks are a concern (79). The estab-
cell-based therapy. lishment of an optimal culture condition free of
potential cross-contaminations is relevant in pro-
ducing clinical-grade human stem cell lines and for
Challenges in bringing stem performing basic research involving the regulation of
their self-renewal and lineage determination. Sec-
cell-based research to practice ond, timing is an inherent constraint in cell therapy
and tissue engineering. Some autologous construct-
Biological challenges
based approaches may involve weeks to months of
Despite biological evidence showing that regenera- ex vivo processing (35). Although the use of stem
tion can occur in humans, complete and predictable cells can often minimize the processing time com-
regeneration still remains an elusive clinical goal pared with somatic cells, possible karyotypic insta-
(especially in advanced periodontal defects). The bility and gene mutations of the cells after prolonged
isolation and characterization of stem cells from culture can also limit their usefulness (78). Third, the
periodontal tissues have provided a good starting search for the ideal biocompatible scaffolding
point for understanding the role of progenitor cells in material(s) and delivery system is a significant
periodontal healing. However, we have incomplete technical pursuit. The ideal matrix scaffold should
understanding of the way that roots develop, and mimic native extracellular matrix, support cell
little is known about the signaling mechanisms that attachment, allow controlled release of bioactive
occur during this process (124, 150). The molecular factors, be conducive to tissue ingrowth and facili-
pathways that underlie stem cell self-renewal and tate laboratory handling (8, 10, 76, 142). There is
differentiation are also largely unknown (16). For evidence to suggest that cultured human periodontal
periodontal regeneration to occur, we need to repli- ligament stem cells in a suitable scaffold implanted
cate the key cellular events that parallel periodontal into surgically created periodontal defects can result
development and to understand the specific cell in the formation of a periodontal ligament-like
types, the inductive factors and the cellular processes structure (111). However, further refinement of the
involved in formation of the periodontium (7). Given methodology to propagate and incorporate these
that the fate of stem cells is influenced by their cells into a carrier scaffold is needed, and this should
interaction with the microenvironment (including limit the ex vivo processing time and encourage the
soluble and immobilized factors, extracellular matrix integration and function of the implanted cells with
and signals from neighboring cells), understanding the host cells (62, 158).
the key components regulating the properties of stem
cells may elucidate ways to expand stem cells prop-
Clinical challenges
erly and control their differentiation precisely (34).
Numerous studies on stem cells to date have Clinical challenges in stem cell-based periodontal
emerged from work on cell culture or animal models. therapy relate to immune rejection after administra-
While some species (e.g. primate) are more suited tion, oncogenic properties of stem cells and func-
than others (e.g. mouse) for understanding stem cell tional integration of transplanted tissues into the host
behavior in humans, these findings may not always (23). It is important to understand how the immune
be directly extrapolated to humans. system will respond to stem cells or their derivatives
upon transplantation. Generally, the immunogenicity
of a human cell depends on its expression of class I
Technical challenges
and II major histocompatibility antigens, which allow
The technical challenges in stem cell therapy are the body to distinguish its own cells from foreign cells
associated with cell manipulations, scaffold materi- (14). Human embryonic stem cells express a low level
als and delivery systems. First, culture conditions are of class I major histocompatibility antigens, but this
not sufficiently developed to mimic the cell micro- expression is up-regulated with differentiation (30). A
environment in vivo and to ensure that both cell potential solution to this problem lies in the use of
proliferation and differentiation can be performed autologous stem cells (from cell ⁄ tissue banks) to

245
Lin et al.

overcome immune rejection (14, 58). Furthermore, within a matrix scaffold, together with the introduc-
the production of patient-specific pluripotent stem tion of various signaling molecules in an orderly
cells (or induced pluripotent stem cells) from adult temporal and spatial sequence. To date, a number of
somatic cells is now feasible and the differentiation of studies have reported that stem cells, in conjunction
autologous induced pluripotent stem cells into cell with different physical matrices and growth factors,
types desired for transplantation is being explored have the capacity to regenerate periodontal tissues
(49). Recent findings relating to the immuno- in vivo. Notwithstanding these significant advances
suppressive effects of mesenchymal stem cells both there are still numerous biological, technical and
in vitro and in vivo have also raised the possibility of clinical hurdles to be overcome. In particular, a
using allogenic stem cells without the need for donor thorough understanding of the underlying pro-
and recipient cross-matching (22, 94, 105). cesses in periodontal development, as well as the
The challenge relating to genomic stability and the mechanisms behind stem cell self-renewal and
risk of tumorigenesis following stem cell transplan- differentiation, will be required. Refinement of cur-
tation are major safety considerations because rent laboratory techniques to facilitate handling of
reliable methods to eliminate undifferentiated stem cells and their translation to a clinical setting
embryonic stem cells from culture are yet to be will be equally critical in advancing the field. Studies
established (13), and current studies lack long-term on both embryonic cells and adult stem cells should
follow-up to draw firm conclusions (154). It is likely continue as part of a collective effort to expand our
that the more specific and extensive the therapeutic knowledge on how cells function and what fails in
application, the longer the stem cells may have to the disease process. It is this combined and solid
remain in vitro. During this extended period in cul- knowledge base that will underpin future treatment
ture, there is a greater likelihood for genetic or epi- modalities and ultimately make stem cell-based
genetic changes in stem cells (31, 78). Improvement tissue engineering and gene therapy a realistic alter-
in the understanding of such changes in vitro will native in periodontal regeneration.
better enable researchers to address the risk of
tumorigenesis in stem cell therapy.
Finally, it is unclear whether human stem-cell Acknowledgments
derivatives can integrate into the recipient tissue and
fulfill the specific functions of lost or injured tissues The authorsÕ work cited in this review has been fun-
(28). It will be necessary to demonstrate that stem ded variously by the National Health and Medical
cells develop into stable cells and display the char- Research Council of Australia (NHMRC), the Austra-
acteristics and functions of normal host cells fol- lian Dental Research Fund and the Australian Peri-
lowing their transplantation. It is hoped that, as our odontology Research Foundation.
knowledge on progenitor cells, growth factors and
delivery systems improves, we will make stem cell-
based therapy a safe and effective approach in References
periodontal regeneration.
1. Abe S, Yamaguchi S, Amagasa T. Multilineage cells from
apical pulp of human tooth with immature apex. Oral Sci
Int 2007: 4: 455–458.
Conclusion 2. Albandar JM. Epidemiology and risk factors of periodontal
diseases. Dent Clin North Am 2005: 49: 517–532, v-vi.
Restoration of tissues destroyed by periodontitis to 3. Amos TA, Gordon MY. Sources of human hematopoietic
their original form and function has been a long- stem cells for transplantation – a review. Cell Transplant
standing goal of periodontal therapy. However, our 1995: 4: 547–569.
4. Anderson WF. Human gene therapy. Nature 1998: 392:
current available regenerative therapies are crude
25–30.
and of poor clinical predictability. There is need for 5. Anusaksathien O, Webb SA, Jin QM, Giannobile WV.
novel regenerative technologies to be developed Platelet-derived growth factor gene delivery stimulates ex
based on contemporary understanding. In order for vivo gingival repair. Tissue Eng 2003: 9: 745–756.
this to become a reality it will be necessary for us to 6. Baroffio A, Hamann M, Bernheim L, Bochaton-Piallat ML,
Gabbiani G, Bader CR. Identification of self-renewing
obtain a complete understanding of periodontal
myoblasts in the progeny of single human muscle satellite
development and the progenitor cells involved in this cells. Differentiation 1996: 60: 47–57.
process. Subsequent tissue-engineering approaches 7. Bartold PM, McCulloch CA, Narayanan AS, Pitaru S.
may then be developed using these progenitor cells Tissue engineering: a new paradigm for periodontal

246
Stem cells and future periodontal regeneration

regeneration based on molecular and cell biology. 26. Darnovsky M, Fogel SB. Oocyte donation for stem cell
Periodontol 2000 2000: 24: 253–269. research. Science 2007: 316: 368–370.
8. Bartold PM, Xiao Y, Lyngstaadas SP, Paine ML, Snead ML. 27. De Bari C, DellÕaccio F. Cell therapy: a challenge in
Principles and applications of cell delivery systems for modern medicine. Biomed Mater Eng 2008: 18: S11–17.
periodontal regeneration. Periodontol 2000 2006: 41: 123– 28. Deans RJ, Moseley AB. Mesenchymal stem cells: biology
135. and potential clinical uses. Exp Hematol 2000: 28: 875–884.
9. Beyer Nardi N, da Silva Meirelles L. Mesenchymal stem 29. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I,
cells: isolation, in vitro expansion and characterisation. Marini F, Krause D, Deans R, Keating A, Prockop D,
In: Alison MR, Wobus AM, Bocheler KR (eds). Stem cells. Horwitz E. Minimal criteria for defining multipotent
Berlin: Springer, 2006: 249–282. mesenchymal stromal cells. The International Society for
10. Bhatnagar RS, Qian JJ, Wedrychowska A, Sadeghi M, Wu Cellular Therapy position statement. Cytotherapy 2006: 8:
YM, Smith N. Design of biomimetic habitats for tissue 315–317.
engineering with P-15, a synthetic peptide analogue of 30. Draper JS, Pigott C, Thomson JA, Andrews PW. Surface
collagen. Tissue Eng 1999: 5: 53–65. antigens of human embryonic stem cells: changes upon
11. Bianco P, Riminucci M, Gronthos S, Robey PG. Bone differentiation in culture. J Anat 2002: 200: 249–258.
marrow stromal stem cells: nature, biology, and potential 31. Draper JS, Smith K, Gokhale P, Moore HD, Maltby E,
applications. Stem Cells 2001: 19: 180–192. Johnson J, Meisner L, Zwaka TP, Thomson JA, Andrews
12. Bjerknes M, Cheng H. Multipotential stem cells in adult PW. Recurrent gain of chromosomes 17q and 12 in cul-
mouse gastric epithelium. Am J Physiol Gastrointest Liver tured human embryonic stem cells. Nat Biotechnol 2004:
Physiol 2002: 283: G767–777. 22: 53–54.
13. Bongso A, Fong CY, Gauthaman K. Taking stem cells to 32. Duailibi SE, Duailibi MT, Zhang W, Asrican R, Vacanti JP,
the clinic: Major challenges. J Cell Biochem 2008: 105: Yelick PC. Bioengineered dental tissues grown in the rat
1352–1360. jaw. J Dent Res 2008: 87: 745–750.
14. Bradley JA, Bolton EM, Pedersen RA. Stem cell medicine 33. Dunn CA, Jin Q, Taba M Jr, Franceschi RT, Bruce
encounters the immune system. Nat Rev Immunol 2002: Rutherford R, Giannobile WV. BMP gene delivery for
2: 859–871. alveolar bone engineering at dental implant defects. Mol
15. Brazelton TR, Rossi FM, Keshet GI, Blau HM. From mar- Ther 2005: 11: 294–299.
row to brain: expression of neuronal phenotypes in adult 34. Eshghi S, Schaffer DV. Engineering microenvironments to
mice. Science 2000: 290: 1775–1779. control stem cell fate and function (September 11, 2008),
16. Brivanlou AH, Gage FH, Jaenisch R, Jessell T, Melton D, StemBook, ed. The Stem Cell Research Community, StemBook,
Rossant J. Stem cells. Setting standards for human doi/10.3824/stembook.1.5.1, http://www.stembook.org.
embryonic stem cells. Science 2003: 300: 913–916. 35. Fauza DO. Tissue engineering: current state of clinical
17. Burt B. Position paper: epidemiology of periodontal dis- application. Curr Opin Pediatr 2003: 15: 267–271.
eases. J Periodontol 2005: 76: 1406–1419. 36. Ferber D. Gene therapy. Safer and virus-free? Science
18. Campagnoli C, Roberts IA, Kumar S, Bennett PR, 2001: 294: 1638–1642.
Bellantuono I, Fisk NM. Identification of mesenchymal 37. Friedenstein AJ, Chailakhyan RK, Latsinik NV, Panasyuk
stem ⁄ progenitor cells in human first-trimester fetal AF, Keiliss-Borok IV. Stromal cells responsible for trans-
blood, liver, and bone marrow. Blood 2001: 98: 2396–2402. ferring the microenvironment of the hemopoietic tissues.
19. Caplan AI. Mesenchymal stem cells. J Orthop Res 1991: 9: Cloning in vitro and retransplantation in vivo. Trans-
641–650. plantation 1974: 17: 331–340.
20. Castro-Malaspina H, Gay RE, Resnick G, Kapoor N, Meyer 38. Friedenstein AJ, Deriglasova UF, Kulagina NN, Panasuk
P, Chiarieri D, McKenzie S, Broxmeyer HE, Moore MAS. AF, Rudakowa SF, Luria EA, Ruadkow IA. Precursors for
Characterization of human bone marrow fibroblast col- fibroblasts in different populations of hematopoietic cells
ony-forming cells (CFU-F) and their progeny. Blood 1980: as detected by the in vitro colony assay method. Exp
56: 289–301. Hematol 1974: 2: 83–92.
21. Chamberlain JR, Schwarze U, Wang PR, Hirata RK, 39. Gay IC, Chen S, MacDougall M. Isolation and character-
Hankenson KD, Pace JM, Underwood RA, Song KM, ization of multipotent human periodontal ligament stem
Sussman M, Byers PH, Russell DW. Gene targeting in stem cells. Orthod Craniofac Res 2007: 10: 149–160.
cells from individuals with osteogenesis imperfecta. 40. Griffith LG, Naughton G. Tissue engineering – current
Science 2004: 303: 1198–1201. challenges and expanding opportunities. Science 2002:
22. Chen X, Armstrong MA, Li G. Mesenchymal stem cells in 295: 1009–1014.
immunoregulation. Immunol Cell Biol 2006: 84: 413–421. 41. Gronthos S, Brahim J, Li W, Fisher LW, Cherman N, Boyde
23. Choumerianou DM, Dimitriou H, Kalmanti M. Stem cells: A, DenBesten P, Robey PG, Shi S. Stem cell properties of
promises versus limitations. Tissue Eng Part B Rev 2008: human dental pulp stem cells. J Dent Res 2002: 81: 531–
14: 53–60. 535.
24. Coura GS, Garcez RC, de Aguiar CB, Alvarez-Silva M, 42. Gronthos S, Franklin DM, Leddy HA, Robey PG, Storms
Magini RS, Trentin AG. Human periodontal ligament: a RW, Gimble JM. Surface protein characterization of hu-
niche of neural crest stem cells. J Periodont Res 2008: 43: man adipose tissue-derived stromal cells. J Cell Physiol
531–536. 2001: 189: 54–63.
25. Cowan CA, Atienza J, Melton DA, Eggan K. Nuclear 43. Gronthos S, Mankani M, Brahim J, Robey PG, Shi S. Post-
reprogramming of somatic cells after fusion with human natal human dental pulp stem cells (DPSCs) in vitro and
embryonic stem cells. Science 2005: 309: 1369–1373. in vivo. Proc Natl Acad Sci U S A 2000: 97: 13625–13630.

247
Lin et al.

44. Gronthos S, Mrozik K, Shi S, Bartold PM. Ovine perio- 59. Izadpanah R, Trygg C, Patel B, Kriedt C, Dufour J, Gimble
dontal ligament stem cells: isolation, characterization, JM, Bunnell BA. Biologic properties of mesenchymal stem
and differentiation potential. Calcif Tissue Int 2006: 79: cells derived from bone marrow and adipose tissue. J Cell
310–317. Biochem 2006: 99: 1285–1297.
45. Gronthos S, Simmons PJ. The growth factor requirements 60. Jin Q, Anusaksathien O, Webb SA, Printz MA, Giannobile
of STRO-1-positive human bone marrow stromal pre- WV. Engineering of tooth-supporting structures by deliv-
cursors under serum-deprived conditions in vitro. Blood ery of PDGF gene therapy vectors. Mol Ther 2004: 9: 519–
1995: 85: 929–940. 526.
46. Gronthos S, Zannettino AC, Hay SJ, Shi S, Graves SE, 61. Jin QM, Anusaksathien O, Webb SA, Rutherford RB,
Kortesidis A, Simmons PJ. Molecular and cellular charac- Giannobile WV. Gene therapy of bone morphogenetic
terisation of highly purified stromal stem cells derived protein for periodontal tissue engineering. J Periodontol
from human bone marrow. J Cell Sci 2003: 116: 1827–1835. 2003: 74: 202–213.
47. Guhr A, Kurtz A, Friedgen K, Loser P. Current state of 62. Jin QM, Zhao M, Webb SA, Berry JE, Somerman MJ,
human embryonic stem cell research: an overview of cell Giannobile WV. Cementum engineering with three-
lines and their use in experimental work. Stem Cells 2006: dimensional polymer scaffolds. J Biomed Mater Res A
24: 2187–2191. 2003: 67: 54–60.
48. Handa K, Saito M, Tsunoda A, Yamauchi M, Hattori S, 63. Kawaguchi H, Hirachi A, Hasegawa N, Iwata T,
Sato S, Toyoda M, Teranaka T, Narayanan AS. Progenitor Hamaguchi H, Shiba H, Takata T, Kato Y, Kurihara H.
cells from dental follicle are able to form cementum Enhancement of periodontal tissue regeneration by
matrix in vivo. Connect Tissue Res 2002: 43: 406–408. transplantation of bone marrow mesenchymal stem cells.
49. Hanna J, Wernig M, Markoulaki S, Sun CW, Meissner A, J Periodontol 2004: 75: 1281–1287.
Cassady JP, Beard C, Brambrink T, Wu LC, Townes TM, 64. Kemoun P, Laurencin-Dalicieux S, Rue J, Farges JC,
Jaenisch R. Treatment of sickle cell anemia mouse model Gennero I, Conte-Auriol F, Briand-Mesange F, Gade-
with iPS cells generated from autologous skin. Science lorge M, Arzate H, Narayanan AS, Brunel G, Salles JP.
2007: 318: 1920–1923. Human dental follicle cells acquire cementoblast
50. Hasegawa N, Kawaguchi H, Hirachi A, Takeda K, Mizuno features under stimulation by BMP-2 ⁄ -7 and enamel
N, Nishimura M, Koike C, Tsuji K, Iba H, Kato Y, Kurihara matrix derivatives (EMD) in vitro. Cell Tissue Res 2007:
H. Behavior of transplanted bone marrow-derived mes- 329: 283–294.
enchymal stem cells in periodontal defects. J Periodontol 65. Korbling M, Estrov Z. Adult stem cells for tissue repair – a
2006: 77: 1003–1007. new therapeutic concept? N Engl J Med 2003: 349: 570–
51. Hoffman LM, Carpenter MK. Characterization and culture 582.
of human embryonic stem cells. Nat Biotechnol 2005: 23: 66. Kuo TF, Huang AT, Chang HH, Lin FH, Chen ST, Chen RS,
699–708. Chou CH, Lin HC, Chiang H, Chen MH. Regeneration of
52. Horwitz EM, Le Blanc K, Dominici M, Mueller I, Slaper- dentin–pulp complex with cementum and periodontal
Cortenbach I, Marini FC, Deans RJ, Krause DS, Keating A. ligament formation using dental bud cells in gelatin-
Clarification of the nomenclature for MSC: The Interna- chondroitin-hyaluronan tri-copolymer scaffold in swine.
tional Society for Cellular Therapy position statement. J Biomed Mater Res A 2008: 86: 1062–1068.
Cytotherapy 2005: 7: 393–395. 67. Kuznetsov SA, Krebsbach PH, Satomura K, Kerr J,
53. Horwitz EM, Gordon PL, Koo WK, Marx JC, Neel MD, Riminucci M, Benayahu D, Robey PG. Single-colony
McNall RY, Muul L, Hofmann T. Isolated allogeneic bone derived strains of human marrow stromal fibroblasts
marrow-derived mesenchymal cells engraft and stimulate form bone after transplantation in vivo. J Bone Miner Res
growth in children with osteogenesis imperfecta: Impli- 1997: 12: 1335–1347.
cations for cell therapy of bone. Proc Natl Acad Sci USA 68. Lagasse E, Connors H, Al-Dhalimy M, Reitsma M, Dohse
2002: 99: 8932–8937. M, Osborne L, Wang X, Finegold M, Weissman IL, Grompe
54. Hu Y, Liao L, Wang Q, Ma L, Ma G, Jiang X, Zhao RC. M. Purified hematopoietic stem cells can differentiate into
Isolation and identification of mesenchymal stem cells hepatocytes in vivo. Nat Med 2000: 6: 1229–1234.
from human fetal pancreas. J Lab Clin Med 2003: 141: 69. Langer R, Tirrell DA. Designing materials for biology and
342–349. medicine. Nature 2004: 428: 487–492.
55. Hu B, Nadiri A, Kuchler-Bopp S, Perrin-Schmitt F, Peters 70. Langer R, Vacanti JP. Tissue engineering. Science 1993:
H, Lesot H. Tissue engineering of tooth crown, root, and 260: 920–926.
periodontium. Tissue Eng 2006: 12: 2069–2075. 71. Leblond CP. Classification of cell populations on the basis
56. Hyun I, Hochedlinger K, Jaenisch R, Yamanaka S. New of their proliferative behavior. Natl Cancer Inst Monogr
advances in iPS cell research do not obviate the need for 1964: 14: 119–150.
human embryonic stem cells. Cell Stem Cell 2007: 1: 367– 72. Li Y, Jin F, Du Y, Ma Z, Li F, Wu G, Shi J, Zhu X, Yu J, Jin Y.
368. Cementum and periodontal ligament-like tissue forma-
57. Itskovitz-Eldor J, Schuldiner M, Karsenti D, Eden A, tion induced using bioengineered dentin. Tissue Eng Part
Yanuka O, Amit M, Soreq H, Benvenisty N. Differentiation A 2008: 14: 1731–1742.
of human embryonic stem cells into embryoid bodies 73. Lin NH, Gronthos S, Bartold PM. Stem cells and peri-
compromising the three embryonic germ layers. Mol Med odontal regeneration. Aust Dent J 2008: 53: 108–121.
2000: 6: 88–95. 74. Liu TM, Martina M, Hutmacher DW, Hui JH, Lee EH, Lim
58. Ivanovski S, Gronthos S, Shi S, Bartold PM. Stem cells in B. Identification of common pathways mediating differ-
the periodontal ligament. Oral Dis 2006: 12: 358–363. entiation of bone marrow- and adipose tissue-derived

248
Stem cells and future periodontal regeneration

human mesenchymal stem cells into three mesenchymal 90. Nakahara T, Nakamura T, Kobayashi E, Kuremoto K,
lineages. Stem Cells 2007: 25: 750–760. Matsuno T, Tabata Y, Eto K, Shimizu Y. In situ tissue
75. Lowry WE, Richter L, Yachechko R, Pyle AD, Tchieu J, engineering of periodontal tissues by seeding with
Sridharan R, Clark AT, Plath K. Generation of human in- periodontal ligament-derived cells. Tissue Eng 2004: 10:
duced pluripotent stem cells from dermal fibroblasts. Proc 537–544.
Natl Acad Sci U S A 2008: 105: 2883–2888. 91. Nakao K, Morita R, Saji Y, Ishida K, Tomita Y, Ogawa M,
76. Lutolf MP, Hubbell JA. Synthetic biomaterials as Saitoh M, Tomooka Y, Tsuji T. The development of a
instructive extracellular microenvironments for morpho- bioengineered organ germ method. Nat Methods 2007: 4:
genesis in tissue engineering. Nat Biotechnol 2005: 23: 227–230.
47–55. 92. Nakashima M, Reddi AH. The application of bone mor-
77. Maherali N, Sridharan R, Xie W, Utikal J, Eminli S, Arnold phogenetic proteins to dental tissue engineering.
K, Stadtfeld M, Yachechko R, Tchieu J, Jaenisch R, Plath K, Nat Biotechnol 2003: 21: 1025–1032.
Hochedlinger K. Directly reprogrammed fibroblasts show 93. Narem R, Sambanis A. Tissue engineering: from biology to
global epigenetic remodeling and widespread tissue biological structures. Tissue Eng 1995: 1: 3–13.
contribution. Cell Stem Cell 2007: 1: 55–70. 94. Nasef A, Mathieu N, Chapel A, Frick J, Francois S,
78. Maitra A, Arking DE, Shivapurkar N, Ikeda M, Stastny V, Mazurier C, Boutarfa A, Bouchet S, Gorin NC, Thierry D,
Kassauei K, Sui G, Cutler DJ, Liu Y, Brimble SN, Noaksson Fouillard L. Immunosuppressive effects of mesenchymal
K, Hyllner J, Schulz TC, Zeng X, Freed WJ, Crook J, stem cells: involvement of HLA-G. Transplantation 2007:
Abraham S, Colman A, Sartipy P, Matsui S, Carpenter M, 84: 231–237.
Gazdar AF, Rao M, Chakravarti A. Genomic alterations in 95. National Institutes of Health (NIH). Stem cells: scientific
cultured human embryonic stem cells. Nat Genet 2005: progress and future research directions. Bethesda: NIH,
37: 1099–1103. 2001.
79. Martin MJ, Muotri A, Gage F, Varki A. Human embryonic 96. Ohazama A, Modino SA, Miletich I, Sharpe PT. Stem-cell-
stem cells express an immunogenic nonhuman sialic acid. based tissue engineering of murine teeth. J Dent Res 2004:
Nat Med 2005: 11: 228–232. 83: 518–522.
80. Mezey E, Chandross KJ, Harta G, Maki RA, McKercher SR. 97. Okita K, Ichisaka T, Yamanaka S. Generation of germline-
Turning blood into brain: cells bearing neuronal antigens competent induced pluripotent stem cells. Nature 2007:
generated in vivo from bone marrow. Science 2000: 290: 448: 313–317.
1779–1782. 98. Owen GR, Jackson J, Chehroudi B, Burt H, Brunette DM.
81. Mezey E, Key S, Vogelsang G, Szalayova I, Lange GD, A PLGA membrane controlling cell behaviour for
Crain B. Transplanted bone marrow generates new neu- promoting tissue regeneration. Biomaterials 2005: 26:
rons in human brains. Proc Natl Acad Sci USA 2003: 100: 7447–7456.
1364–1369. 99. Park IH, Zhao R, West JA, Yabuuchi A, Huo H, Ince TA,
82. Miura M, Gronthos S, Zhao M, Lu B, Fisher LW, Robey PG, Lerou PH, Lensch MW, Daley GQ. Reprogramming of
Shi S. SHED: stem cells from human exfoliated deciduous human somatic cells to pluripotency with defined factors.
teeth. Proc Natl Acad Sci U S A 2003: 100: 5807–5812. Nature 2008: 451: 141–146.
83. Morrison SJ, Kimble J. Asymmetric and symmetric stem- 100. Pickering SJ, Minger SL, Patel M, Taylor H, Black C, Burns
cell divisions in development and cancer. Nature 2006: CJ, Ekonomou A, Braude PR. Generation of a human
441: 1068–1074. embryonic stem cell line encoding the cystic fibrosis
84. Morsczeck C, Gotz W, Schierholz J, Zeilhofer F, Kuhn U, mutation deltaF508, using preimplantation genetic diag-
Mohl C, Sippel C, Hoffmann KH. Isolation of precursor nosis. Reprod Biomed Online 2005: 10: 390–397.
cells (PCs) from human dental follicle of wisdom teeth. 101. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas
Matrix Biol 2005: 24: 155–165. R, Mosca JD, Moorman MA, Simonetti DW, Craig S,
85. Muraglia A, Cancedda R, Quarto R. Clonal mesenchymal Marshak DR. Multilineage potential of adult human
progenitors from human bone marrow differentiate in mesenchymal stem cells. Science 1999: 284: 143–147.
vitro according to a hierarchical model. J Cell Sci 2000: 7: 102. Pountos I, Jones E, Tzioupis C, McGonagle D, Giannoudis
1161–1166. PV. Growing bone and cartilage. The role of mesenchymal
86. Murphy JM, Fink DJ, Hunziker EB, Barry FP. Stem cell stem cells. J Bone Joint Surg Br 2006: 88: 421–426.
therapy in a caprine model of osteoarthritis. Arthritis 103. Prockop DJ. Marrow stromal cells as stem cells for
Rheum 2003: 48: 3464–3474. nonhematopoietic tissues. Science 1997: 276: 71–74.
87. Musina RA, Bekchanova ES, Sukhikh GT. Comparison of 104. Prockop DJ, Sekiya I, Colter DC. Isolation and character-
mesenchymal stem cells obtained from different human ization of rapidly self-renewing stem cells from cultures of
tissues. Bull Exp Biol Med 2005: 139: 504–509. human marrow stromal cells. Cytotherapy 2001: 3: 393–396.
88. Musina RA, Bekchanova ES, Belyavskii AV, Sukhikh GT. 105. Puissant B, Barreau C, Bourin P, Clavel C, Corre J, Bous-
Differentiation potential of mesenchymal stem cells of quet C, Taureau C, Cousin B, Abbal M, Laharrague P,
different origin. Bull Exp Biol Med 2006: 141: 147–151. Penicaud L, Casteilla L, Blancher A. Immunomodulatory
89. Nakahara T, Nakamura T, Kobayashi E, Inoue M, Shigeno effect of human adipose tissue-derived adult stem cells:
K, Tabata Y, Eto K, Shimizu Y. Novel approach to regen- comparison with bone marrow mesenchymal stem cells.
eration of periodontal tissues based on in situ tissue Br J Haematol 2005: 129: 118–129.
engineering: effects of controlled release of basic fibro- 106. Ramseier CA, Abramson ZR, Jin Q, Giannobile WV. Gene
blast growth factor from a sandwich membrane. Tissue therapeutics for periodontal regenerative medicine. Dent
Eng 2003: 9: 153–162. Clin North Am 2006: 50: 245–263.

249
Lin et al.

107. Reubinoff BE, Pera MF, Fong CY, Trounson A, Bongso A. 124. Steele-Perkins G, Butz KG, Lyons GE, Zeichner-David M,
Embryonic stem cell lines from human blastocysts: so- Kim HJ, Cho MI, Gronostajski RM. Essential role for
matic differentiation in vitro. Nat Biotechnol 2000: 18: NFI-C ⁄ CTF transcription-replication factor in tooth root
399–404. development. Mol Cell Biol 2003: 23: 1075–1084.
108. Ripamonti U, Reddi AH. Tissue engineering, morpho- 125. Tada M, Takahama Y, Abe K, Nakatsuji N, Tada T. Nuclear
genesis, and regeneration of the periodontal tissues by reprogramming of somatic cells by in vitro hybridization
bone morphogenetic proteins. Crit Rev Oral Biol Med with ES cells. Curr Biol 2001: 11: 1553–1558.
1997: 8: 154–163. 126. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T,
109. Sander L, Karring T. Healing of periodontal lesions in Tomoda K, Yamanaka S. Induction of pluripotent stem
monkeys following the guided tissue regeneration proce- cells from adult human fibroblasts by defined factors. Cell
dure. A histological study. J Clin Periodontol 1995: 22: 2007: 131: 861–872.
332–337. 127. Takahashi K, Yamanaka S. Induction of pluripotent stem
110. Seo BM, Miura M, Gronthos S, Bartold PM, Batouli S, cells from mouse embryonic and adult fibroblast cultures
Brahim J, Young M, Robey PG, Wang CY, Shi S. Investi- by defined factors. Cell 2006: 126: 663–676.
gation of multipotent postnatal stem cells from human 128. Techawattanawisal W, Nakahama K, Komaki M, Abe M,
periodontal ligament. Lancet 2004: 364: 149–155. Takagi Y, Morita I. Isolation of multipotent stem cells
111. Seo BM, Miura M, Sonoyama W, Coppe C, Stanyon R, Shi from adult rat periodontal ligament by neurosphere-
S. Recovery of stem cells from cryopreserved periodontal forming culture system. Biochem Biophys Res Commun
ligament. J Dent Res 2005: 84: 907–912. 2007: 357: 917–923.
112. Shi S, Bartold PM, Miura M, Seo BM, Robey PG, Gronthos 129. Ten Cate AR. The development of the periodontium – a
S. The efficacy of mesenchymal stem cells to regenerate largely ectomesenchymally derived unit. Periodontol 2000
and repair dental structures. Orthod Craniofac Res 2005: 8: 1997: 13: 9–19.
191–199. 130. Thesleff I, Tummers M. Stem cells and tissue engineering:
113. Slade GD, Spencer AJ, Roberts-Thomson KF. AustraliaÕs prospects for regenerating tissues in dental practice. Med
dental generations: the National Survey of Adult Oral Princ Pract 2003: 12(Suppl. 1): 43–50.
Health 2004–06. Canberra: Australian Institute of Health 131. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA,
and Welfare, 2007. Swiergiel JJ, Marshall VS, Jones JM. Embryonic stem cell
114. Slavkin HC, Bartold PM. Challenges and potential in tis- lines derived from human blastocysts. Science 1998: 282:
sue engineering. Periodontol 2000 2006: 41: 9–15. 1145–1147.
115. Smith A. A glossary for stem-cell biology. Nature 2006: 132. Till JE, McCulloch EA. A direct measurement of the radi-
441: 1060. ation sensitivity of normal mouse bone marrow cells.
116. Smith C, Storms B. Hematopoietic stem cells. Clin Orthop Radiat Res 1961: 14: 213–222.
Relat Res 2000: S91–97. 133. Tobita M, Uysal AC, Ogawa R, Hyakusoku H, Mizuno H.
117. Snyder EY, Deitcher DL, Walsh C, Arnold-Aldea S, Periodontal tissue regeneration with adipose-derived
Hartwieg EA, Cepko CL. Multipotent neural cell lines can stem cells. Tissue Eng Part A 2008: 14: 945–953.
engraft and participate in development of mouse cere- 134. Toma JG, Akhavan M, Fernandes KJ, Barnabe-Heider F,
bellum. Cell 1992: 68: 33–51. Sadikot A, Kaplan DR, Miller FD. Isolation of multipotent
118. Somia N, Verma IM. Gene therapy: trials and tribulations. adult stem cells from the dermis of mammalian skin. Nat
Nat Rev Genet 2000: 1: 91–99. Cell Biol 2001: 3: 778–784.
119. Sonoyama W, Liu Y, Yamaza T, Tuan RS, Wang S, Shi S, 135. Trubiani O, Di Primio R, Traini T, Pizzicannella J, Scarano
Huang GT. Characterization of the apical papilla and its A, Piattelli A, Caputi S. Morphological and cytofluori-
residing stem cells from human immature permanent metric analysis of adult mesenchymal stem cells
teeth: a pilot study. J Endod 2008: 34: 166–171. expanded ex vivo from periodontal ligament. Int J Im-
120. Sonoyama W, Liu Y, Fang D, Yamaza T, Seo BM, Zhang C, munopathol Pharmacol 2005: 18: 213–221.
Liu H, Gronthos S, Wang CY, Shi S, Wang S. Mesenchymal 136. Trubiani O, Orsini G, Zini N, Di Iorio D, Piccirilli M,
stem cell-mediated functional tooth regeneration in Piattelli A, Caputi S. Regenerative potential of human
Swine. PLoS ONE 2006: 1: e79. periodontal ligament derived stem cells on three-dimen-
121. Srisuwan T, Tilkorn DJ, Wilson JL, Morrison WA, Messer sional biomaterials: A morphological report. J Biomed
HM, Thompson EW, Abberton KM. Molecular aspects of Mater Res A 2008: 87: 986–993.
tissue engineering in the dental field. Periodontol 2000 137. Trubiani O, Scarano A, Orsini G, Di Iorio D, DÕArcangelo
2006: 41: 88–108. C, Piccirilli M, Sigismondo M, Caputi S. The performance
122. Stamm C, Westphal B, Kleine HD, Petzsch M, Kittner C, of human periodontal ligament mesenchymal stem cells
Klinge H, Schumichen C, Nienaber CA, Freund M, on xenogenic biomaterials. Int J Immunopathol Phar-
Steinhoff G. Autologous bone-marrow stem-cell trans- macol 2007: 20: 87–91.
plantation for myocardial regeneration. Lancet 2003: 361: 138. Vacanti JP, Langer R. Tissue engineering: the design and
45–46. fabrication of living replacement devices for surgical
123. Stavropoulos A, Kostopoulos L, Nyengaard JR, Karring T. reconstruction and transplantation. Lancet 1999:
Deproteinized bovine bone (Bio-Oss) and bioactive 354(Suppl 1): SI32–34.
glass (Biogran) arrest bone formation when used as 139. Venezia E, Goldstein M, Boyan BD, Schwartz Z. The use of
an adjunct to guided tissue regeneration (GTR): an enamel matrix derivative in the treatment of periodontal
experimental study in the rat. J Clin Periodontol 2003: 30: defects: a literature review and meta-analysis. Crit Rev
636–643. Oral Biol Med 2004: 15: 382–402.

250
Stem cells and future periodontal regeneration

140. Watt FM, Hogan BL. Out of Eden: stem cells and their Establishment of immortalized dental follicle cells for
niches. Science 2000: 287: 1427–1430. generating periodontal ligament in vivo. Cell Tissue Res
141. Wernig M, Meissner A, Foreman R, Brambrink T, Ku M, 2007: 327: 301–311.
Hochedlinger K, Bernstein BE, Jaenisch R. In vitro repro- 152. Young CS, Abukawa H, Asrican R, Ravens M, Troulis MJ,
gramming of fibroblasts into a pluripotent ES-cell-like Kaban LB, Vacanti JP, Yelick PC. Tissue-engineered hybrid
state. Nature 2007: 448: 318–324. tooth and bone. Tissue Eng 2005: 11: 1599–1610.
142. Whang K, Healy KE, Elenz DR, Nam EK, Tsai DC, Thomas 153. Young HE, Steele TA, Bray RA, Hudson J, Floyd JA,
CH, Nuber GW, Glorieux FH, Travers R, Sprague SM. Engi- Hawkins K, Thomas K, Austin T, Edwards C, Cuzzourt J,
neering bone regeneration with bioabsorbable scaffolds Duenzl M, Lucas PA, Black AC Jr. Human reserve plurip-
with novel microarchitecture. Tissue Eng 1999: 5: 35–51. otent mesenchymal stem cells are present in the
143. Williams RC, Barnett AH, Claffey N, Davis M, Gadsby R, connective tissues of skeletal muscle and dermis derived
Kellett M, Lip GY, Thackray S. The potential impact of from fetal, adult, and geriatric donors. Anat Rec 2001: 264:
periodontal disease on general health: a consensus view. 51–62.
Curr Med Res Opin 2008: 24: 1635–1643. 154. Yu J, Thomson JA. Embryonic stem cells. In: National
144. Wilmut I, Schnieke AE, McWhir J, Kind AJ, Campbell KH. Institutes of Health (NIH) (ed). Regenerative medicine
Viable offspring derived from fetal and adult mammalian 2006. Bethesda: Department of Health and Human Ser-
cells. Nature 1997: 385: 810–813. vices, 2006: 1–12.
145. Wood A. Ethics and embryonic stem cell research. Stem 155. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J,
Cell Rev 2005: 1: 317–324. Frane JL, Tian S, Nie J, Jonsdottir GA, Ruotti V, Stewart R,
146. Xiao Y, Parry DA, Li H, Arnold R, Jackson WJ, Bartold PM. Slukvin II, Thomson JA. Induced pluripotent stem cell
Expression of extracellular matrix macromolecules lines derived from human somatic cells. Science 2007: 318:
around demineralized freeze-dried bone allografts. 1917–1920.
J Periodontol 1996: 67: 1233–1244. 156. Yu J, Wang Y, Deng Z, Tang L, Li Y, Shi J, Jin Y. Odonto-
147. Yang X, van der Kraan PM, van den Dolder J, Walboomers genic capability: bone marrow stromal stem cells versus
XF, Bian Z, Fan M, Jansen JA. STRO-1 selected rat dental dental pulp stem cells. Biol Cell 2007: 99: 465–474.
pulp stem cells transfected with adenoviral-mediated 157. Zannettino AC, Paton S, Arthur A, Khor F, Itescu S,
human bone morphogenetic protein 2 gene show en- Gimble JM, Gronthos S. Multipotential human adipose-
hanced odontogenic differentiation. Tissue Eng 2007: 13: derived stromal stem cells exhibit a perivascular
2803–2812. phenotype in vitro and in vivo. J Cell Physiol 2008: 214:
148. Yao S, Pan F, Prpic V, Wise GE. Differentiation of stem 413–421.
cells in the dental follicle. J Dent Res 2008: 87: 767–771. 158. Zhao M, Jin Q, Berry JE, Nociti FH Jr, Giannobile WV,
149. Yen AH, Sharpe PT. Regeneration of teeth using stem cell- Somerman MJ. Cementoblast delivery for periodontal
based tissue engineering. Expert Opin Biol Ther 2006: 6: 9– tissue engineering. J Periodontol 2004: 75: 154–161.
16. 159. Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ,
150. Yokohama-Tamaki T, Ohshima H, Fujiwara N, Takada Y, Benhaim P, Lorenz HP, Hedrick MH. Multilineage cells
Ichimori Y, Wakisaka S, Ohuchi H, Harada H. Cessation of from human adipose tissue: implications for cell-based
Fgf10 signaling, resulting in a defective dental epithelial therapies. Tissue Eng 2001: 7: 211–228.
stem cell compartment, leads to the transition from crown 160. Zwaka TP. Use of genetically modified stem cells in
to root formation. Development 2006: 133: 1359–1366. experimental gene therapies. In: National Institutes of
151. Yokoi T, Saito M, Kiyono T, Iseki S, Kosaka K, Nishida E, Health (NIH) (ed). Regenerative medicine 2006. Bethesda:
Tsubakimoto T, Harada H, Eto K, Noguchi T, Teranaka T. Department of Health and Human Services, 2006:45–52.

251

You might also like