Adj 12100

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Australian Dental Journal

The official journal of the Australian Dental Association


Australian Dental Journal 2014; 59:(1 Suppl): 117–130

doi: 10.1111/adj.12100

Stem cells, tissue engineering and periodontal regeneration


J Han,* D Menicanin,* S Gronthos,† PM Bartold*
*Colgate Australian Clinical Dental Research Centre, School of Dentistry, The University of Adelaide, South Australia.
†School of Medical Sciences, The University of Adelaide, South Australia.

ABSTRACT
The aim of this review is to discuss the clinical utility of stem cells in periodontal regeneration by reviewing relevant lit-
erature that assesses the periodontal-regenerative potential of stem cells. We consider and describe the main stem cell
populations that have been utilized with regard to periodontal regeneration, including bone marrow-derived mesenchy-
mal stem cells and the main dental-derived mesenchymal stem cell populations: periodontal ligament stem cells, dental
pulp stem cells, stem cells from human exfoliated deciduous teeth, stem cells from apical papilla and dental follicle pre-
cursor cells. Research into the use of stem cells for tissue regeneration has the potential to significantly influence peri-
odontal treatment strategies in the future.
Keywords: Periodontium, repair, bone grafts, bioactive materials, scaffolds.
Abbreviations and acronyms: ADSC = adipose-derived stromal cell; BMP = bone morphogenetic protein; BMSC = bone marrow stro-
mal stem cell; CFU-F = colony-forming unit fibroblast; DPSC = dental pulp stem cell; EMD = enamel matrix derivative; GFP = green
fluorescent protein; IGF-I = insulin-like growth factor-I; iPS = induced pluripotent stem; ISCT = International Society for Cellular Ther-
apy; MSC = mesenchymal stem cell; PDGF = platelet-derived growth factor; PDL = periodontal ligament; PDLSC = periodontal liga-
ment stem cell; PRP = platelet-rich plasma; SCAP = stem cell from apical papilla; SHED = stem cell from exfoliated deciduous teeth.

economic cost. The ultimate goal of periodontal ther-


INTRODUCTION
apy relies on the achievement of complete restoration
Periodontal disease is a chronic inflammatory condi- of all components of the periodontium to their origi-
tion of the periodontium that is characterized by irre- nal architecture and function. This entails reconstruc-
versible destruction of the tooth attachment and its tion of gingival connective tissue, cementum, alveolar
surrounding bone. The disease state, if left untreated, bone and periodontal ligament (PDL). In addition,
can lead to progressive loss of gingival tissue, peri- formation of Sharpey’s fibres, or the portion of the
odontal ligament and supporting alveolar bone, ulti- PDL embedded in both newly formed cementum and
mately resulting in an aesthetically and functionally alveolar bone, is essential to restore appropriate con-
compromised dentition, including premature tooth nections between the tooth and its supporting tissues.8
loss.1 The pathogenesis of periodontal disease involves Current conventional techniques for the treatment of
a complex interaction between the host’s immune periodontal disease show a limited potential for com-
response to microbial colonization of the periodontal plete periodontal regeneration. An improved under-
attachment, and modifying host factors, including standing of periodontal biology coupled with current
tobacco smoking2 and genetic susceptibility.3 Perio- advances in the development of scaffolding matrices
dontal disease has also been linked to many underly- has introduced novel treatments that use cell and gene
ing systemic disorders such as diabetes,4 therapy to enhance periodontal tissue reconstruction
cardiovascular disease and rheumatoid arthritis.6 Pro-
5
and its biomechanical integration.
gressive periodontitis is seen in most adult human
populations, with a prevalence of either moderate or
The periodontium
severe periodontal disease in the Australian popula-
tion estimated at 22.9%.7 The consequences of The periodontium is a complex organ consisting of
untreated periodontal disease have broad ranging two soft connective tissues (gingival and periodontal
implications on an individual’s quality of life, and ligament) and two hard connective tissues (cementum
thus impact upon the health system and carry a heavy and alveolar bone).9
© 2013 Australian Dental Association 117
J Han et al.

Formation of the periodontium with the dentine of the root.24 Approximately 45% to
50% of cementum is composed of inorganic hydroxy-
Periodontal regeneration aims to recapitulate the cru-
apatite whilst organic collagen and non-collagenous
cial stages of wound healing associated with periodon-
matrix proteins constitute the remaining 50%. Colla-
tal development in order to restore lost tissues to their
gen type III and XII are found alongside collagen type
original form and function.10 Hence, an understand-
I which constitutes up to 90% of the organic compo-
ing of the processes involved in the development of
nent in cementum.25 Additionally, non-collagenous,
the periodontium is essential in order to elucidate the
bone-associated proteins, identified in cementum,
cellular and molecular events that might occur during
include bone sialoprotein, osteopontin, osteocalcin
periodontal regeneration.
and osteonectin.26,27
The tooth is formed as a result of reciprocal inter-
Cementum can be broadly classified as cellular and
actions between the dental epithelium and neural crest
acellular but at least five types have been identified
cell-derived ectomesenchyme.11–15 Dental develop-
and described depending not only on their cellular
ment involves distinct morphological stages, namely
content but also the presence of extrinsic or intrinsic
bud, cap, bell and crown stage. At the bell stage, the
collagen fibres.28 The layer of cementum attached to
tooth germ consists of the enamel organ, dental
the root dentine and covering the cervical portion of
papilla and dental follicle, a fibrocellular layer envel-
the root is acellular and is termed primary cementum.
oping the dental papilla and enamel organ. The sup-
The apical portion of the root is covered by a cellular,
porting tissues of the tooth form from the dental
or secondary cementum, in which cementoblasts,
follicle.16 Development of the periodontal tissues
cementum forming cells, remain trapped in lacunae
begins when cells of the inner and outer dental epithe-
within their own matrix, characteristic of osteocytes
lium proliferate from the cervical loop of the enamel
in bone. These entrapped cells are termed cemento-
organ to form a double layer of cells known as Her-
cytes. Acellular cementum anchors the periodontal lig-
twig’s epithelial root sheath. The root sheath separates
ament fibre bundles to the tooth, while cellular
cells of the dental papilla from the dental follicle.
cementum has an adaptive role, allowing bodily tooth
Cells of the dental papilla differentiate into odonto-
movement within the jaw during orthodontic tooth
blasts to form root dentine, following which cells of
movement.25
the root sheath secrete a fine matrix of proteins onto
the dentine surface, known as the hyaline layer of
Hopewell Smith.17 Subsequent fragmentation of the Periodontal ligament
root sheath allows ectomesenchymal cells of the den-
The periodontal ligament is a highly specialized
tal follicle to attach to this protein matrix and differ-
fibrous connective tissue situated between the cemen-
entiate into cementoblasts.18,19 Apical development of
tum, covering the root of the tooth, and the bone
the root continues with continuing cementum deposi-
that forms the socket wall. It is a highly vascular-
tion. Collagen fibre bundles become embedded in
ized tissue, which reflects the high rate of turnover
newly-formed cementum, known as Sharpey’s fibres.
of its cellular and extracellular constituents. Similar
This process triggers the formation of the periodontal
to all other connective tissues, the periodontal liga-
ligament by fibroblasts also derived from the dental
ment consists of cells and an extracellular compo-
follicle.20,21 Likewise, bundle bone, the portion of the
nent of collagenous fibres and a non-collagenous
alveolar process that lines the tooth socket, is formed
extracellular matrix.16 The constituent cells include
by active osteoblasts originating in the dental follicle.
osteoblasts and osteoclasts (contained within the
Finally, insertion of Sharpey’s fibres into this newly-
ligament but functionally associated with bone),
forming bone completes the development of the
fibroblasts, epithelial cell rests of Malassez, macro-
attachment complex of the periodontium.22 The coor-
phages, undifferentiated mesenchymal cells,29 and
dinated formation of cementum, periodontal ligament
cementoblasts (also contained within the ligament
and alveolar bone, along with the transformation of
but functionally associated with cementum). The
reduced enamel epithelium to sulcular and junctional
extracellular compartment consists of well-defined
epithelium during tooth eruption, give rise to the com-
collagenous and non-collagenous proteins.30 The
plete periodontium.23 A summary of the biochemical
human periodontal ligament spans approximately
composition and function of each of the tissues is
0.2 mm in width, and provides mechanical resistance
presented below.
for the absorption of masticatory forces on the
tooth via the dense masses of collagen fibre bundles.
Cementum Furthermore, the periodontal ligament is mechano-
responsive and shows adaption to increased or
Cementum is an avascular, bone-like connective tissue
decreased mechanical loads by regulating its overall
which lines the tooth root and is firmly interlocked

118 © 2013 Australian Dental Association


Tissue engineered periodontal products

width by as much as 50% without losing its original the local environment to reduce inflammation. Cur-
architecture. The periodontal ligament also provides a rent non-surgical techniques, such as subgingival
proprioceptive function for the neuromuscular control debridement, and surgical procedures, such as open
of mastication.25,31 flap debridement, aim to remove diseased tissue and
clean the root surface to encourage reattachment of
tissues. These techniques typically result in a process
Alveolar bone
of repair (Table 1), leading to the healing of the
The alveolar bone constitutes the alveolar process, wound site by formation of a long junctional epithe-
which is firmly attached to the basal bone of the jaw. lial attachment. This epithelial attachment represents
The alveolar bone comprises inner and outer compo- a non-physiologic epithelial attachment that does not
nents and is perforated by many foramina, which provide a strong connection between root surface and
transmit nerves and vessels. The bone directly lining the adjacent gingival connective tissue.34 This process
the socket, the inner aspect of alveolar bone, is specifi- results in minimal repair of the damaged cementum or
cally referred to as bundle bone. Embedded within alveolar bone and therefore, by definition, cannot be
this bone are the extrinsic collagen fibre bundles of considered true regeneration of the periodontium.8
periodontal ligament (Sharpey’s fibres). The bundle While healing by repair may be effective in preventing
bone generally contains less intrinsic collagen fibrils future progression of disease and therefore any ulti-
than other types of bone. The alveolar bone is sub- mate tooth loss, there are a number of limitations to
jected to remodelling during normal function and dur- the therapeutic outcomes. Firstly, the treatment may
ing increased mechanical loading.25,32 have limited effect in resolving the existing mobility
of the teeth due to lack of regeneration of lost tissues.
Secondly, such treatment frequently results in gingival
Gingiva
recession, which is both unsightly and may result in
The gingiva has the general pattern of stratified predisposition of affected teeth to root caries. Finally,
squamous epithelium with an underlying connective it is postulated that, without restitution of the original
tissue lamina propria which bounds tightly to the anatomy, the site may be more susceptible to recur-
bone. The covering epithelium is keratinized to with- rence of disease in the future.35
stand masticatory and other minor traumatic forces
during normal oral function. Cells from the kerati-
Current treatment regenerative approaches
nized surface layer are continually shed and replaced
from the underlying granular or intermediate layer, A number of different procedures have been described
prickle-cell layer and the basal layer. This represents to promote true and predictable regeneration of the
progressive epithelial maturation and is achieved by periodontium since the 1980s. To describe the latest
proliferation of cells in the basal layer by mitosis, trends, principles of these different treatment
cell differentiation at the supra-basal layers, and cell approaches include the use of graft materials to com-
death at the surface layer. The main cellular compo- pensate for the bone loss incurred as a result of peri-
nent of the oral epithelium is the keratinocyte. Non- odontal disease,36 use of barrier membranes for
keratinocytes also present in the oral epithelium guided tissue regeneration,37,38 and use of bioactive
include Langerhans cells, Merkel’s cells, melanocytes molecules.39–41
and inflammatory cells including lymphocytes. The
connective tissue layer, lamina propria, which under-
lies the oral epithelium contains fibroblasts, immune Table 1. Periodontal wound healing responses by
cells, endothelial cells, blood and lymphatic vessels, repair and regeneration
neural elements, as well as a complex extracellular Repair Regeneration
matrix composed of collagenous and non-collagenous
Control of inflammation Formation of new functional
proteins.30 attachment, including formation
of cementum, periodontal ligament
and alveolar bone
Periodontal wound healing Formation of long
junctional epithelium
The unique anatomy and composition of the peri- Re-attachment of connective
odontium, comprising hard and soft connective tissues, tissue to adjacent root surface
New bone separation from
as well as epithelium, make periodontal wound heal- the root surface by long
ing a complex process.33 The current conventional functional epithelium,
techniques for periodontal repair focus on arresting accompanied by root
resorption, and/or ankylosis
the progression of periodontal disease by reducing the
microbial levels in the periodontium, and modifying Adapted from reference 8.
© 2013 Australian Dental Association 119
J Han et al.

Bone grafts formation. Among those characterized to date are the


platelet-derived growth factor (PDGF) and insulin-like
Various types of bone grafts have been investigated to
growth factor-I (IGF-I) which have shown to enhance
determine their ability to stimulate new bone forma-
regeneration in beagle dogs and monkeys with peri-
tion. These include introduction of alloplastic materi-
odontal disease.53,54 In addition, bone morphogenetic
als (generally synthetic fillers), autografts (grafted
proteins (BMPs) have also shown potential to stimu-
tissue from one site to another in the same individ-
late bone and cementum regeneration.55
ual), allografts (tissue between individuals of the same
The most widely clinically tested of these molecules
species but with different genetic composition) and
is the enamel matrix derivative (EMD), known to reg-
xenografts (grafted materials between different spe-
ulate the epithelial mesenchymal interactions involved
cies). The use of such grafting materials has resulted
in initial tooth formation. The mode of function of
in some gain in clinical attachment levels and radio-
these proteins remains unclear. However, it is known
graphic evidence of bone fill. However, histological
that they are produced by Hertwig’s epithelial sheath,
analyses of these treatments have shown minimal
and play an important role in cementogenesis and the
osteoinductive and cementogenic capacity as most
development of the periodontal attachment appara-
generally become encased in a dense fibrous connec-
tus.41 EmdogainTM (Straumann AG, Basel, Switzer-
tive tissue.42,43
land) is a mixture of enamel matrix proteins primarily
containing amelogenins derived from developing por-
Guided tissue regeneration cine teeth. Furthermore, EmdogainTM contains TGF-b
and BMP growth factors and, as such, the addition of
Based on the hypothesis of Melcher in 1976 that the
these proteins to intrabony defects may promote peri-
first cells to populate a wound will dictate the nature
odontal regeneration by recapitulating the environ-
and quality of tissue that forms there,33 Nyman et al.
ment during initial tooth attachment.39–41 A Cochrane
first demonstrated that by guiding the colonization of
review of 13 clinical trials revealed that while EMD
the periodontal wound healing site by progenitor cells
significantly improved probing attachment levels
from the periodontal ligament and alveolar bone, and
(1.1 mm) and pocket depth reduction (0.9 mm), the
eliminating cells of gingival epithelial and fibroblastic
data could not be interpreted fairly due to a high
lineages from this site, it is feasible to achieve peri-
degree of heterogeneity observed among the included
odontal regeneration in experimental animals and
trials.39 Overall, it was found that EmdogainTM pro-
humans.44,45 This technique, termed guided tissue
duces results similar to those seen with the use of con-
regeneration, has recently become a widely accepted
ventional guided tissue regeneration, and no evidence
clinical procedure, and is currently considered the
of clinically significant differences between the use of
‘gold standard’ upon which to base and compare
EMD and guided tissue regeneration could be
regenerative therapies.46,47 Guided tissue regeneration
found.39 In addition, clinical significance of these pro-
involves utilization of barrier membranes to prevent
cedures, and particularly the benefits that accrue to
unwanted epithelium and gingival connective tissue
patients from these outcomes, remain to be fully dem-
from entering the healing site while recruiting cells
onstrated as all of these procedures are only applica-
from the periodontium to re-populate the defect area.
ble in very specific indications, such as in infrabony
It has, however, been reported that the clinical
defects exhibiting vertical bone loss around single
improvements obtained through the use of guided tis-
teeth, or between furcation lesions. Consequently, in
sue regeneration are, at present, minor and highly var-
many cases where periodontal attachment loss is pres-
iable.48–50 The limited success of this approach has
ent in a regular horizontal pattern throughout the
been attributed to insufficient biocompatibility and
mouth, regenerative procedures currently have little or
poor mechanical properties of the common membrane
no utility.
materials used for their crucial role of wound stabil-
ization and space maintenance. Moreover, there is evi-
dence to suggest that the outcomes of treatment are Periodontal regeneration summary
technique sensitive.51,52
For regeneration to occur, healing events must pro-
gress in an ordered and programmed sequence both
Bioactive materials temporally and spatially, replicating the key events in
periodontal development.56 Currently, it is known
Another approach to induce periodontal regeneration
that appropriate progenitor cells must first migrate to
has involved the direct delivery of bioactive polypep-
the wound site and attach to the denuded root surface
tide growth factors to the root surface in order to
in order to proliferate and mature into the tissue com-
facilitate the cascade of wound healing events
ponents of a functional periodontal attachment
that lead to new cementum and connective tissue

120 © 2013 Australian Dental Association


Tissue engineered periodontal products

apparatus. The success of proliferation, migration and


maturation of these progenitor cells is known to then
depend on the availability of appropriate growth fac-
tors and contact with extracellular matrix, controlling
gene expression.57,58 In the absence of appropriate
cellular, molecular and matrix signals and compo-
nents, healing may be compromised and occur by
repair rather than regeneration. Progenitor cells are of
particular interest in periodontal wound healing and
regeneration as they are most likely the parental cells
of synthetic cells such as osteoblasts, cementoblasts
and fibroblasts, responsible for the restoration of lost
periodontal tissues (Fig. 1). Recent evidence shows Fig. 2 Schematic representation of periodontal tissue engineering. A
successful outcome of periodontal tissue engineering requires an ade-
that subset populations isolated from the periodontal quate supply of appropriate progenitor cells with the capacity to differen-
ligament have characteristics of stem cells. As a conse- tiate into the required mature tissue-forming phenotypes, including
quence, current research trends have been directed osteoblasts, cementoblasts and fibroblasts; the appropriate signalling mol-
ecules to modulate cellular differentiation; and a conductive three-dimen-
towards developing cellular-based techniques for peri- sional extracellular matrix scaffold to support and facilitate these
odontal regeneration. processes. Angiogenic signals, promoting new vascular networks, are
essential to provide the nutritional base for tissue growth and
homeostasis.
Cell-based tissue engineering
In light of the clinical unpredictability of currently associated with conventional regenerative procedures
available treatment modalities to treat all types of as it allows direct placement of growth factors and
periodontal defects, tissue engineering has emerged as progenitor cells into the defect, eliminating the
an alternative approach to alleviate the shortcomings expected lag phase of progenitor cell recruitment to
of conventional therapeutic options, by regenerating the wound site.62 A successful outcome of periodontal
living and functional dental structures. Tissue engi- tissue engineering requires the following essential fac-
neering draws on principles of cell biology, develop- tors: (1) an adequate supply of appropriate progenitor
mental biology and biomaterials science to fabricate cells with the capacity to differentiate into the
new structures to replace lost or damaged tissues.59 required mature tissue-forming phenotypes, including
The concept has been applied to recent applications osteoblasts, cementoblasts and fibroblasts; (2) the
of reconstruction and regeneration of the periodon- appropriate signals to modulate cellular differentiation
tium,56 the dentine-pulp complex60 and orofacial and tissue neogenesis; and (3) a conductive three-
tissues.61 dimensional extracellular matrix scaffold to support
In the context of periodontal therapy, a potential and facilitate these processes.35 Additionally, angio-
tissue engineering approach to periodontal regenera- genic signals, promoting new vascular networks, are
tion involves incorporation of progenitor cells in a essential to provide the nutritional base for tissue
prefabricated three-dimensional construct, which is growth and homeostasis. Furthermore, appropriate
subsequently implanted into the defect site (Fig. 2).56 mechanical loading is required for the development of
This strategy overcomes some of the limitations highly organized, functional periodontal ligament
fibres. Finally, as periodontal defect sites accompany a
microbial load, strategies to control infection and host
Stem cell Progenitor cells Committed cells
response are required for optimum periodontal regen-
Growth factors
eration.63
Pre-
cementoblast Cementoblast One of the most critical factors in tissue engineering
is the choice of scaffold and optimal stem cell popula-
tion to employ.
Pre-
fibroblast Fibroblast
Scaffold-based tissue engineering
For successful tissue engineering to be achieved a
Pre-
osteoblast Osteoblast scaffold must be combined with living cells, and/or
Extra cellular matrix
Cell adhesion molecules biologically active molecules. This will create a ‘tissue
engineering construct’ to be used to promote regenera-
Fig. 1 A schematic diagram of proposed differentiation of adult
mesenchymal stem cells and progenitor cells into periodontal cells. tion of tissues.64,65 These scaffolds need to support
(Reproduced with permission from reference 9.) various important cellular processes and functions,
© 2013 Australian Dental Association 121
J Han et al.

including cell colonization, migration, growth and dif- Furthermore, tissue in-growth does not equate to tis-
ferentiation. Physico-chemical properties, morphology sue maturation and remodelling, in other words a
and degradation kinetics of the scaffold are also defect filled with immature tissue should not be con-
important design features which need to be taken into sidered ‘regenerated’. Hence, many scaffold-based
account. In order for constructs to be clinically suc- strategies have failed in the past as the scaffold degra-
cessful they need to be able to support and stimulate dation was more rapid than tissue remodelling and/or
both the onset and the continuance of tissue maturation. In this context it has been proposed that
in-growth and also allow subsequent remodelling and the onset of degradation should only occur after the
maturation. This can be achieved by providing opti- regenerated tissue within the scaffold has remodelled
mal stiffness together with predetermined external at least once in the natural remodelling cycle.67
and internal geometrical shapes. Tissue engineering
constructs must also provide sufficient initial mechani-
Stem cells
cal strength and stiffness to substitute for the loss of
mechanical function of the diseased, damaged or miss- By definition, a stem cell refers to a clonogenic, rela-
ing tissue. To achieve stable biomechanical conditions tively undifferentiated cell that is capable of self-
and vascularization at the host site, continuous cell renewal and multi-lineage differentiation.68 Therefore,
and tissue remodelling is important. In addition to a stem cell is considered to be capable of propagating
these requirements of mechanics and geometry, a use- and generating daughter stem cells, whilst its progeny
ful construct will: (1) possess a three-dimensional and holds the potential to differentiate and commit to
highly porous interconnected pore network with opti- maturation along multiple lineages, giving rise to a
mized surface properties to allow attachment, migra- range of specialized cell types. A stem cell may either
tion, proliferation and differentiation of cell types undergo prolonged self-renewal or differentiation
needed for tissue regeneration as well as enable satis- depending on intrinsic signals modulated by extrinsic
factory flow transport of nutrients and metabolic factors in the stem cell niche.69 Stem cells have now
waste; and (2) be biocompatible and biodegradable been isolated from a wide variety of tissues and upon
with a controllable rate to complement cell/tissue their characterization, these populations have been
growth and maturation.66 It is now recognized that categorized according to their respective developmen-
for successful tissue formation, remodelling, and mat- tal potential (Fig. 3).
uration at the defect site it is essential to understand Stem cells can be broadly divided into three catego-
and control the scaffold degradation process. In the ries: embryonic stem cells, adult, somatic or postnatal
early days of tissue engineering, it was believed that stem cells and more recently, through genetic manipu-
scaffolds should rapidly degrade and disappear as the lation, induced pluripotent stem cells. Embryonic stem
tissue in-growth was occurring. It is important to rec- cells are pluripotent cells derived from the early mam-
ognize that tissue in-growth, remodelling and matura- malian embryo with the capacity to proliferate exten-
tion differ temporally between various tissues. sively and differentiate into cells with features of all

Germ cells

Sperm Ovum
TOTIPOTENT STEM CELLS
Fertilisation Fertilised Egg (capable of differentiating into all possible cell types
including extraembryonic tissues)

Embryonic Stem Cells


Blastocyst
(derived from inner cell mass of pre-implantation embryo)

Embryonic Germ Cells PLURIPOTENT STEM CELLS


Foetus (derived from primordial germ cells isolated from embryonal (capable of differentiating into all possible cell types
gonad) including extraembryonic tissues)

Adult Embryonic Carcinoma Cells


(derived from primordial germ cells in embryonic gonad but
usually found as components of testicular tumours in adults)

Adult Stem Cells MULTIPOTENT STEM CELLS


(derived from many ectodermal and mesodermal organs in (capable of differentiating into multiple, but a limited
adults) number of cell lineages)

Adults cells undergone nuclear transformation (clonal) TOTIPOTENT

INDUCED PLURIPOTENT STEM CELLS


Adult cells induced to an embryonic stem cell (somatic cells genetically manipulated to a
phenotype pluripotent state, thought to possess comparable
cell potency to embryonic stem cells)

Fig. 3 Derivation of stem cell populations. Stem cells are classified according to their differentiation potency as defined by the site, stage of development
or cell culture induction environment in which the populations are maintained. Cells may be categorized as totipotent, pluripotent, multipotent or induced
pluripotent. (Adapted, with permission, from reference 62.)
122 © 2013 Australian Dental Association
Tissue engineered periodontal products

three embryonic germ layers (mesoderm, endoderm (adipocytes, osteoblasts, chondrocytes, tenocytes, skel-
and ectoderm). These cells have the ability to develop etal myocytes and visceral stromal cells),79,81–85 ecto-
into almost all cell types; however, they lack the dermal (neurons, astrocytes)88 and endodermal
capacity to contribute to extra embryonic tissue and (hepatocytes)89 origins.
therefore cannot develop into a fetal or adult ani- Originally, MSCs were isolated from bone marrow
mal.70–72 Despite their developmental potential, the and stroma of spleen and thymus.86,87 Hence, these
use of embryos to obtain human embryonic stem cell cells have previously been referred to as bone marrow
lines raises serious ethical concerns, which recently stromal stem cells (BMSCs).90 Friedenstein and Owen
prompted efforts to genetically reprogramme somatic demonstrated that BMSCs can be identified by their
cells back to their pluripotent state. These efforts capacity to form clonal colonies when bone marrow
resulted in the generation of induced pluripotent stem aspirates were plated at low density and cultured for
(iPS) cells that are functionally similar to embryonic a period of two weeks.91 Clonal characterization indi-
stem cells. These cells are comparable to embryonic cated that a proportion of cell clones displayed the
stem cells in their morphology, gene expression pro- capacity to differentiate into multiple stromal lineages
files, proliferation and differentiation capacities.73–75 including myelosuppurative stroma, bone, fat and car-
However, genetic manipulations fundamental to gen- tilage, whilst other colonies displayed a restricted
eration of iPS cells may alter their growth and devel- capacity for differentiation and were more representa-
opmental characteristics, which hinders the tive of committed progenitor cells, rather than multi-
predictability of their behaviour and, as such, limits potential BMSCs.89,92–94 The colony forming assay is
their use in tissue-regenerative purposes. Furthermore, a widely accepted technique for establishing BMSC
embryonic stem cells and iPSCs both carry tumouri- cultures from bone marrow aspirates based on their
genic properties, raising a significant safety challenge plastic-adherent properties, allowing for their isolation
in the use of these cells for regenerative therapies.76,77 from non-adherent haemopoietic stem cells. However,
Adult stem cells have been identified in numerous this type of culture system is composed, not only of
tissue niches in the postnatal organism and are BMSCs, but also other adherent accessory cells includ-
thought to function in replenishing cell loss as a con- ing smooth muscle cells, endothelial cells, osteoblasts
sequence of tissue damage and death.78 Since their and adherent macrophages.90 This makes character-
discovery, it has been recognized that the developmen- ization of the biological properties of primitive
tal capabilities of adult stem cells are greater than ini- BMSCs difficult as the contaminating accessory cells
tially thought. In addition to being responsible for the could influence the growth rate and differentiation
maintenance of tissue homeostasis in their host tissue, potential of BMSC-derived colonies. Moreover, subse-
it has also been demonstrated that they have the quent studies which characterized MSC-like popula-
capacity to differentiate into other cellular lineages tions from other tissues including trabecular bone,
beyond their tissue(s) of origin.79,80 Whilst these cells periosteum, articular cartilage, synovium, synovial
exhibit a more restricted proliferation and differentia- fluid, skeletal muscle, adipose tissue,95 tendons,
tion potential compared to embryonic stem cells, they blood,96 blood vessels, umbilical cord vasculature,97
are easily accessible, immunocompatible and are not placental tissue,98,99 fetal tissues100,101 and skin102
associated with ethical concerns. demonstrated that a cell’s ability to adhere to and
grow on plastic was not sufficient to classify them as
MSC. To account for the biologic properties of multi-
Mesenchymal stem cells
potential, clonogenic, plastic-adherent cells derived
Mesenchymal stem cells (MSCs) are adult stem cells from various stromal tissues, the International Society
able to give rise to multiple specialized cell types.79,81–85 for Cellular Therapy (ISCT) has proposed the use of
Due to their extensive distribution in many adult the term ‘mesenchymal stem cell’ and acronym ‘MSC’
tissues, including those of dental origin, MSCs have only where characterized populations satisfy the mini-
become an attractive target for use in periodontal mal criteria as outlined in Table 2.103
regeneration.
Use of mesenchymal stem cells in tissue engineering
Defining properties of mesenchymal stem cells
MSCs are considered suitable candidates for cell-based
MSCs were first described by Friedenstein in 1976 as tissue engineering strategies owing to their extensive
clonogenic, bone marrow-derived, plastic-adherent expansion rate and potential to differentiate into cells
cells which were termed colony-forming unit fibro- of multiple organs and systems. Although MSCs dem-
blasts (CFU-F).86,87 The term now refers to a hetero- onstrate a lower developmental potential and a shorter
geneous mix of progenitors, where subset populations life span than pluripotent embryonic stem cells, MSCs
are capable of differentiating into cells of mesodermal have not been reported to spontaneously form tumours
© 2013 Australian Dental Association 123
J Han et al.

Table 2. Minimal criteria for defining multipotent Embryogenesis Postnatal stem cells
Committed Mature
Progenitors Stromal Cells
mesenchymal stem cells (MSCs) as proposed by the
Myelosupportive
International Society for Cellular Therapy (ISCT) Stroma
Muscle
Minimal criteria for defining multipotent Marrow Stromal Stem
mesenchymal stem cells (MSCs) Mesoderm Cell
Bone

Adipose
Adherence to plastic Cells must be plastic-adherent when
Cartilage
cultured in standard culture conditions
in standard culture flasks. Dentin
A specific surface antigen At least 95% of the MSC population Dental Pulp Stem Cell Fibrous Pulp
(Ag) expression pattern must express immunophenotype
markers CD105, CD73 and CD90, Adipose

while less than 2% of the population Neural crest


should express markers CD45, CD34, Cementum
CD14 or CD11b, CD79a or CD19 Periodontal Ligament
Ligament
and HLA class II. These markers may Stem Cell

also be expressed by cultured Adipose


fibroblasts isolated from different
tissues that have little or no Fig. 4 Proposed stromal hierarchy of cellular differentiation. Precursor
multi-lineage potential. cells residing in the bone marrow stroma, dental pulp and periodontal
Multipotent MSC populations must give rise to at ligament tissues have the capacity to differentiate into lineage-specific
differentiation potential least three cell lineages: osteogenic, cells that give rise to various stromal tissues. (Reproduced, with permis-
chondrogenic and adipogenic under sion, from reference 109.)
standard in vitro differentiation
conditions.
engineering. MSCs derived from multiple tissue
Reproduced, with permission, from reference 99.
sources have been investigated in preclinical animal
studies for the treatment and regeneration of the peri-
odontium.
in vivo, and are not constrained by ethical consider-
ations.104 Additionally, MSCs seem to be not only
Extraoral mesenchymal stem cells in periodontal
hypoimmunogenic and thus suitable for allogeneic
tissue engineering bone marrow-derived
transplantation, but they further exhibit immunosup-
mesenchymal stem cells
pressive properties upon transplantation.105
Considering their potential in regenerative applica- Past research has demonstrated a potential for the
tions, a variety of stem cells have been identified in use of MSCs, derived from tissue sources outside the
most organs and tissues in the body.106–108 Studies on oral cavity, for transplantation to the periodontal
the developmental potential of MSCs have provided complex.114 Auto-transplantation of bone marrow-
the foundation for the proposed stromal hierarchy of derived MSCs has been shown to promote up to
these cells (Fig. 4),109 a concept arising from a series 20% of cementum and alveolar bone regeneration in
of landmark studies by Friedenstein and experimental Class III defects in dogs.115 A cell-label-
Owen.87,91,93,94,110–112 Based on these findings, perio- ling technique in which bone marrow-derived MSCs
dontal ligament derived mesenchymal stem cell-like were engineered to express green fluorescent protein
cells are now being considered as a more appropriate (GFP) showed that, four weeks after transplantation,
cell type for developing novel tissue periodontal tissue the periodontal defects were almost completely
engineering strategies. Nevertheless, aside from fulfill- regenerated and contained GFP positive cemento-
ing the essential factors of optimal cell populations, blasts, osteoblasts, osteocytes and fibroblasts within
signals and scaffolds, required for tissue engineering a the regenerated periodontal tissue. These results sug-
major obstacle that currently limits the clinical use of gest that transplanted bone marrow-derived MSCs
MSCs for tissue regeneration is the heterogeneity of differentiated into cementoblasts, periodontal liga-
the starting cell population, and the inability to estab- ment fibroblasts, and alveolar bone osteoblasts
lish optimal growth and differentiation conditions in vivo.116 In a subsequent small human clinical
in vitro which may result in poor reproducibility of trial, autologous, expanded bone marrow-derived
therapeutic outcomes.113 MSCs mixed with atelocollagen were transplanted
into periodontal osseous defects at the time of peri-
odontal surgery.117 Positive clinical results were
Cells for periodontal regeneration
obtained, including a 4-mm reduction in probing
Both extraoral and intraoral tissues have stem cell depths and respectively, a 4-mm attachment gain.
populations that represent a viable and accessible Furthermore, radiographic assessments showed that
alternative source to harvest and expand multipotent the use of MSCs in conjunction with platelet-rich
colonies for potential use in periodontal tissue plasma (PRP) demonstrated a reduction in intrabony
124 © 2013 Australian Dental Association
Tissue engineered periodontal products

defect depth and resolution of bleeding. Interdental Initially, dental MSCs were isolated from human
papillae, supported by this tissue engineering technol- pulp tissue and, upon their characterization, were
ogy, had also regenerated.117 termed postnatal dental pulp stem cells (DPSCs).123
Thus, from the preclinical trials conducted to date, Subsequently, three more types of dental-MSC-like
bone marrow-derived MSCs have the capacity to pro- populations were isolated and characterized: stem
mote periodontal regeneration through enhanced gen- cells from exfoliated deciduous teeth (SHED)124; peri-
eration of alveolar bone and neovascularization. odontal ligament stem cells (PDLSCs)29; and stem
Notably, transplanted bone marrow-derived MSCs cells from apical papilla (SCAP).125,126 Recent studies
have also been reported to contribute to the formation have also identified more dental-tissue-derived progen-
of new cementum and periodontal ligament. How- itor cell populations, from the dental follicle and gin-
ever, despite their proven capacity to regenerate peri- giva.127,128 The developmental relationship between
odontal tissues, the difficulty associated with the these different mesenchymal stem cell-like populations
ascertainment of these cells for use in the clinical set- has yet to be determined. The different cell popula-
ting has instigated exploration of dental-tissue-derived tions differ in aspects of their growth rate in culture,
MSC-like populations which can be simply obtained gene and protein expression profiles and cell differen-
chairside rather than via an invasive bone marrow tiation capacities, although the extent to which these
aspiration procedure in a secondary clinic. differences can be attributed to tissue of origin, func-
tion or culture conditions remains unclear. Nonethe-
less, the abovementioned intraoral progenitor cells
Adipose-derived stromal cells
have recently been used for tissue engineering studies
Adipose tissue is another extraoral and non-dental in small and large animal models to assess their
derived source of MSCs potentially applicable in peri- potential in preclinical applications.
odontal tissue engineering. Adipose-derived stromal Of particular interest is the capacity of progenitor
cells (ADSCs) mixed with platelet rich plasma have cell populations derived from periodontal ligament as
been shown to promote regeneration of periodontal the presence of multiple cell types within periodontal
ligament-like structures along with alveolar bone in ligament suggests that this tissue contains progenitor
rats.118 These observations suggest that ADSCs may cells that maintain tissue homeostasis and regenera-
be useful in future clinical cell-based therapy for peri- tion of the periodontal tissues. Earlier evidence has
odontal tissue engineering and present a favourable shown that periodontal ligament contains cell popula-
cell candidate due to ease of accessibility to human tions that can differentiate into either cementoblasts
lipoaspirates and low morbidity associated with their or osteoblasts.129,130 These cells can form clonogenic
procurement. colonies with characteristics of postnatal stem cells,
are self-renewable and possess the ability to give rise
to a range of dental and non-dental tissues, including
Intraoral (dental-derived) mesenchymal stem cells for
cementoblast-like cells, adipocytes and collagen-form-
periodontal tissue engineering
ing cells. When transplanted into immunocompro-
Dental-tissue-derived mesenchymal stem cell-like pop- mised rodents, PDLSCs show the capacity to generate
ulations are among many other isolated and charac- a cementum/PDL-like structure and contribute to
terized stem cells residing in specialized tissues. With periodontal tissue repair.29
the exception of enamel, which lacks ameloblasts or
other cellular elements following tooth development,
In vivo differentiation capacity of periodontal
the periodontium and dentine continue to retain some
ligament stem cells
regenerative or reparative capacities. This is thought
to be mediated by the presence of multipotent progen- There has been a considerable amount of research
itor cells that are capable of differentiation into conducted to assess the regenerative capacity of
functional, lineage-specific cells.31,119–121 However, PDLSCs in a range of dental and craniofacial defects
dental-tissue-derived progenitor cells are likely to be in various animal models. It is evident from these
more committed or restricted in their cellular potency studies that implanted PDLSCs generate cementum
when compared to bone marrow-derived MSCs and periodontal ligament-like structures similar to
because dental tissues do not undergo continuous native periodontal complex.
remodelling as do bone tissues. Additionally, dental In a very early study autologous re-implantation of
mesenchyme is termed ‘ectomesenchyme’ due to its extracted dental roots lined with PDL cells in minipigs
earlier interaction with the neural crest. Thus, it is resulted in the formation of connective tissue, resem-
postulated that these ectomesenchyme-derived dental bling PDL and mimicking the orientation of the fibre
stem cells possess characteristics similar to those of bundles, within four weeks of implantation.131 Twelve
neural crest cells (Fig. 4).122 weeks post implantation, root surfaces were covered
© 2013 Australian Dental Association 125
J Han et al.

by organized connective tissue resembling periodontal have also demonstrated potential efficacy and safety
ligament and oriented fibre bundles were attached to of implanting autologous cells and displayed clinical
alveolar bone and root, penetrating the bone and root benefits from the treatment. However, important chal-
surfaces in the same way as Sharpey’s fibres.131 In lenges remain to be addressed before such treatments
subsequent studies reimplanted cultured cells from the are applied as a standard in a clinical setting.
periodontium implanted into experimental furcation
and interdental defects in a variety of animal models
CONCLUSIONS
resulted in formation of new cementum and bone,
and new attachment.130,132–135 Complete and predictable regeneration of periodontal
More recently periodontal ligament cell sheets have tissues lost due to trauma or disease presents a major
emerged as a novel alternative approach for periodon- challenge. Periodontal regeneration requires consider-
tal tissue engineering as the technical handling of ation of many factors that parallel periodontal devel-
implanted cell population bypasses disruption of criti- opment, including the use of optimal progenitor cell
cal cell surface proteins such as ion channels, growth population, signalling molecules and matrix scaffold
factor receptors and cell-to-cell junction proteins.132 in an orderly temporal and spatial sequence. The peri-
Preclinical investigations have noted periodontal odontal ligament is now known to be a rich source of
regeneration involving formation of new bone, peri- MSCs, and while this tissue appears to have high
odontal ligament and cementum after eight weeks, in regenerative potential, it is difficult to harness and uti-
three out of five test sites, when autologous periodon- lize this capacity for clinical utility.
tal ligament cell sheets were implanted into dehiscence To date, extraoral and dental-tissue-derived stem/
and intrabony defects in dogs.136,137 Allogeneic trans- progenitor cells have been used for tissue engineering
plants of periodontal ligament cell sheets in miniature studies in small and large animal models to assess
swine have demonstrated periodontal tissue regenera- their potential in preclinical applications. While there
tion in a manner no different to that observed in may be an overwhelming body of evidence to support
autologous transplants with no evidence of rejection the notion that MSCs can be used for periodontal
of the allogeneic cells.138 regeneration, there are several main objectives that
In a more ambitious study, stem cells from root need to be addressed before the development of effec-
apical papilla (SCAP) together with PDLSCs in a tive cellular-based therapies for regenerative dentistry.
HA/TCP and Gelfoamâ (absorbable gelatine) carrier This requires better understanding of: (a) the mecha-
have been studied in an attempt to generate a func- nisms of self-renewal in order to sufficiently regulate
tional root/periodontal complex capable of supporting adult stem cell growth in vitro to generate required
a porcelain crown in miniature pigs.125 Following cell numbers needed for different applications; (b) the
extraction of the lower incisor, the site was cleaned to regulation of stem cells during differentiation and
ensure removal of all periodontal ligament tissues and maturation into tissue-specific cell types, as well
the implant constructs, coated with the cells/carrier, as during wound healing; (c) the interactions between
were inserted into the extraction socket and sutured. stem cells and the immune system, in particular,
The surface of the constructed implant was surgically regarding use of allogeneic cell populations; and
re-opened at three months post implantation and a (d) mechanisms needed to control and prevent ex
porcelain crown resembling a miniature pig incisor vivo-expanded mesenchymal stem cells from transfor-
was inserted and cemented into a pre-formed site in mation.
the implant. Four weeks post installation of the
crown, CT and histological analysis confirmed regen-
DISCLOSURE
eration of the root/periodontal structure with signifi-
cantly better compression strength than defects which The authors have no conflicts of interest to declare.
did not receive stem cells. This proof-of-concept study
showed the efficacy of using a multi-type stem cell
REFERENCES
approach to mimic a bio-physiological root/periodon-
tal reconstruction.125 1. Pihlstrom BL, Michalowicz BS, Johnson NW. Periodontal
diseases. Lancet 2005;366:1809–1820.
Despite the general consensus observed in preclini-
2. Bergstrom J. Tobacco smoking and chronic destructive perio-
cal animal studies assessing the potential of PDLSCs dontal disease. Odontology 2004;92:1–8.
in periodontal regenerative therapy, only two human 3. Michalowicz BS, Diehl SR, Gunsolley JC, et al. Evidence of a
clinical studies have been conducted to date.139,140 substantial genetic basis for risk of adult periodontitis. J Peri-
The findings from these studies have shown some odontol 2000;71:1699–1707.
encouraging but variable results which provide proof 4. Soskolne WA, Klinger A. The relationship between periodon-
tal diseases and diabetes: an overview. Ann Periodontol
of principle for the use of autologous periodontal liga- 2001;6:91–98.
ment cells for regenerative purposes. These studies
126 © 2013 Australian Dental Association
Tissue engineered periodontal products

5. Scannapieco FA, Bush RB, Paju S. Associations between peri- 27. Matias MA, Li H, Young WG, Bartold PM. Immunohisto-
odontal disease and risk for atherosclerosis, cardiovascular chemical localisation of extracellular matrix proteins in the
disease, and stroke. A systematic review. Ann Periodontol periodontium during cementogenesis in the rat molar. Arch
2003;8:38–53. Oral Biol 2003;48:709–716.
6. Bartold PM, Marshall RI, Haynes DR. Periodontitis and rheu- 28. Schroeder HE. Cementogenesis and radicular cementum. In:
matoid arthritis: a review. J Periodontol 2005;76(Suppl Oral structural biology. Stuttgart: Thieme, 1991:144–147.
11):2066–2074. 29. Seo BM, Miura M, Gronthos S, et al. Investigation of multi-
7. Roberts-Thomson KF, Do L. Gum diseases. In: Slade GD, potent postnatal stem cells from human periodontal ligament.
Spencer AJ, Roberts-Thomson KF, eds. Australia’s dental gen- Lancet 2004;364:149–155.
erations: the National Survey of Adult Oral Health 2004–06. 30. Bartold PM, Narayanan AS. Molecular and cell biology of
Canberra: Australian Institute of Health and Welfare, 2007: healthy and diseased periodontal tissues. Periodontol 2000
118–136. 2006;40:29–49.
8. Narayanan AS, Bartold PM. Biochemistry of periodontal con- 31. Beertsen W, McCulloch CA, Sodek J. The periodontal liga-
nective tissues and their regeneration: a current perspective. ment: a unique, multifunctional connective tissue. Periodontol
Connect Tissue Res 1996;34:191–201. 2000 1997;13:20–40.
9. Ivanovski S. Periodontal regeneration. Aust Dent J 2009;54 32. Saffar JL, Lasfargues JJ, Cherruau M. Alveolar bone and the
(Suppl 1):S118–128. alveolar process: the socket that is never stable. Periodontol
10. MacNeil RL, Somerman MJ. Development and regeneration 2000 1997;13:76–90.
of the periodontium: parallels and contrasts. Periodontol 33. Melcher AH. On the repair potential of periodontal tissues. J
2000 1999;19:8–20. Periodontol 1976;47:256–260.
11. Jernvall J, Thesleff I. Reiterative signaling and patterning during 34. Caton J, Nyman S, Zander H. Histometric evaluation of peri-
mammalian tooth morphogenesis. Mech Dev 2000;92:19–29. odontal surgery. II. Connective tissue attachment levels after
12. Pispa J, Thesleff I. Mechanisms of ectodermal organogenesis. four regenerative procedures. J Clin Periodontol 1980;7:224–
Dev Biol 2003;262:195–205. 231.
13. Thesleff I, Partanen AM, Vainio S. Epithelial-mesenchymal 35. Hughes FJ, Ghuman M, Talal A. Periodontal regeneration: a
interactions in tooth morphogenesis: the roles of extracellular challenge for the tissue engineer? Proc Inst Mech Eng H
matrix, growth factors, and cell surface receptors. J Craniofac 2010;224:1345–1358.
Genet Dev Biol 1991;11:229–237. 36. Aichelmann-Reidy ME, Yukna RA. Bone replacement
14. Thesleff I, Sharpe P. Signalling networks regulating dental grafts. The bone substitutes. Dent Clin North Am 1998;42:
development. Mech Dev 1997;67:111–123. 491–503.
15. Thesleff I, Vaahtokari A, Kettunen P, Aberg T. Epithelial- 37. Gottlow J, Nyman S, Karring T, Lindhe J. New attachment
mesenchymal signaling during tooth development. Connect formation as the result of controlled tissue regeneration. J
Tissue Res 1995;32:9–15. Clin Periodontol 1984;11:494–503.
16. Nanci A. Periodontium. In: Ten Cate’s Oral Histology: Devel- 38. Gottlow J, Nyman S, Lindhe J, Karring T, Wennstr€ om J.
opment, Structure and Function. St. Louis: CV Mosby, New attachment formation in the human periodontium by
2003:240–274. guided tissue regeneration. Case reports. J Clin Periodontol
17. Slavkin HC, Bringas P Jr, Bessem C, et al. Hertwig’s epithelial 1986;13:604–616.
root sheath differentiation and initial cementum and bone 39. Esposito M, Coulthard P, Worthington HV. Enamel matrix
formation during long-term organ culture of mouse mandibu- derivative (Emdogainâ) for periodontal tissue regeneration in
lar first molars using serumless, chemically-defined medium. intrabony defects. Cochrane Database Syst Rev 2009(4):
J Periodontal Res 1989;24:28–40. CD003875.
18. Hammarstrom L, Alatli I, Fong CD. Origins of cementum. 40. Sculean A, Windisch P, Szendr€ oi-Kiss D, et al. Clinical and
Oral Dis 1996;2:63–69. histologic evaluation of an enamel matrix derivative com-
19. MacNeil RL, Thomas HF. Development of the murine peri- bined with a biphasic calcium phosphate for the treatment of
odontium. II. Role of the epithelial root sheath in formation of human intrabony periodontal defects. J Periodontol
the periodontal attachment. J Periodontol 1993;64:285–291. 2008;79:1991–1999.
20. Yamamoto T, Domon T, Takahashi S, Wakita M. Compara- 41. Bosshardt DD. Biological mediators and periodontal regenera-
tive study of the initial genesis of acellular and cellular cemen- tion: a review of enamel matrix proteins at the cellular and
tum in rat molars. Anat Embryol (Berl) 1994;190:521–527. molecular levels. J Clin Periodontol 2008;35(Suppl 8):87–
105.
21. Yamamoto T, Hinrichsen KV. The development of cellular
cementum in rat molars, with special reference to the fiber 42. Bartold PM, Xiao Y, Lyngstaadas SP, Paine ML, Snead ML.
arrangement. Anat Embryol (Berl) 1993;188:537–549. Principles and applications of cell delivery systems for peri-
odontal regeneration. Periodontol 2000 2006;41:123–135.
22. Ten Cate AR. Formation of supporting bone in association
with periodontal ligament organization in the mouse. Arch 43. Garraway R, Young WG, Daley T, Harbrow D, Bartold
Oral Biol 1975;20:137–138. PM. An assessment of the osteoinductive potential of com-
mercial demineralized freeze-dried bone in the murine thigh
23. Ten Cate AR. The role of epithelium in the development, muscle implantation model. J Periodontol 1998;69:1325–
structure and function of the tissues of tooth support. Oral 1336.
Dis 1996;2:55–62.
44. Nyman S, Gottlow J, Karring T, Lindhe J. The regenerative
24. Bosshardt DD, Selvig KA. Dental cementum: the dynamic tis- potential of the periodontal ligament. An experimental study
sue covering of the root. Periodontol 2000 1997;13:41–75. in the monkey. J Clin Periodontol 1982;9:257–265.
25. Nanci A, Bosshardt DD. Structure of periodontal tissues in 45. Nyman S, Lindhe J, Karring T, Rylander H. New attachment
health and disease. Periodontol 2000 2006;40:11–28. following surgical treatment of human periodontal disease.
26. Bosshardt DD, Zalzal S, McKee MD, Nanci A. Developmen- J Clin Periodontol 1982;9:290–296.
tal appearance and distribution of bone sialoprotein and os- 46. Karring T, Nyman S, Gottlow J, Laurell L. Development of
teopontin in human and rat cementum. Anat Rec 1998;250: the biological concept of guided tissue regeneration–animal
13–33. and human studies. Periodontol 2000 1993;1:26–35.

© 2013 Australian Dental Association 127


J Han et al.

47. Nakae H, Narayanan AS, Raines E, Page RC. Isolation and 67. Woodruff MA, Hutmacher DW. The return of a forgotten
partial characterization of mitogenic factors from cementum. polymer–Polycaprolactone in the 21st century. Progress in
Biochemistry 1991;30:7047–7052. Polymer Science 2010;35:1217–1256.
48. Bratthall G, S€
oderholm G, Neiderud AM, Kullendorff B, Ed- 68. Smith A. A glossary for stem-cell biology. Nature
wardsson S, Attstr€om R. Guided tissue regeneration in the 2006;441:1060–1060.
treatment of human infrabony defects. Clinical, radiographi- 69. Morrison SJ, Shah NM, Anderson DJ. Regulatory mecha-
cal and microbiological results: a pilot study. J Clin Periodon- nisms in stem cell biology. Cell 1997;88:287–298.
tol 1998;25(11 Pt 1):908–914.
70. Evans MJ, Kaufman MH. Establishment in culture of pluri-
49. Pontoriero R, Lindhe J. Guided tissue regeneration in the potential cells from mouse embryos. Nature 1981;292:154–
treatment of degree III furcation defects in maxillary molars. 156.
J Clin Periodontol 1995;22:810–812.
71. Martin GR. Isolation of a pluripotent cell line from early
50. Wallace SC, Gellin RG, Miller MC, Mishkin DJ. Guided tis- mouse embryos cultured in medium conditioned by teratocar-
sue regeneration with and without decalcified freeze-dried cinoma stem cells. Proc Natl Acad Sci U S A 1981;78:7634–
bone in mandibular Class II furcation invasions. J Periodontol 7638.
1994;65:244–254.
72. Thomson JA, Itskovitz-Eldor J, Shapiro SS, et al. Embryonic
51. Tonetti MS, Cortellini P, Suvan JE, et al. Generalizability of stem cell lines derived from human blastocysts. Science
the added benefits of guided tissue regeneration in the treat- 1998;282:1145–1147.
ment of deep intrabony defects. Evaluation in a multi-center
randomized controlled clinical trial. J Periodontol 1998; 73. Takahashi K, Tanabe K, Ohnuki M, et al. Induction of plu-
69:1183–1192. ripotent stem cells from adult human fibroblasts by defined
factors. Cell 2007;131:861–872.
52. Tonetti MS, Prato GP, Cortellini P. Factors affecting the heal-
ing response of intrabony defects following guided tissue 74. Takahashi K, Yamanaka S. Induction of pluripotent stem
regeneration and access flap surgery. J Clin Periodontol 1996; cells from mouse embryonic and adult fibroblast cultures by
23:548–556. defined factors. Cell 2006;126:663–676.
53. Lynch SE, de Castilla GR, Williams RC, et al. The effects of 75. Yu J, Vodyanik MA, Smuga-Otto K, et al. Induced pluripo-
short-term application of a combination of platelet-derived tent stem cell lines derived from human somatic cells. Science
and insulin-like growth factors on periodontal wound healing. 2007;318:1917–1920.
J Periodontol 1991;62:458–467. 76. Bongso A, Fong CY, Gauthaman K. Taking stem cells to the
54. Rutherford RB, Ryan ME, Kennedy JE, Tucker MM, Cha- clinic: major challenges. J Cell Biochem 2008;105:1352–
rette MF. Platelet-derived growth factor and dexamethasone 1360.
combined with a collagen matrix induce regeneration of the 77. Lee H, Park J, Forget BG, Gaines P. Induced pluripotent stem
periodontium in monkeys. J Clin Periodontol 1993;20:537– cells in regenerative medicine: an argument for continued
544. research on human embryonic stem cells. Regen Med
55. Ripamonti U, Reddi AH. Periodontal regeneration: potential 2009;4:759–769.
role of bone morphogenetic proteins. J Periodontal Res 78. Blau HM, Brazelton TR, Weimann JM. The evolving concept
1994;29:225–235. of a stem cell: entity or function? Cell 2001;105:829–841.
56. Bartold PM, McCulloch CA, Narayanan AS, Pitaru S. Tissue 79. Pittenger MF, Mackay AM, Beck SC, et al. Multilineage
engineering: a new paradigm for periodontal regeneration potential of adult human mesenchymal stem cells. Science
based on molecular and cell biology. Periodontol 2000 1999;284:143–147.
2000;24:2532–2569.
80. Bianco P, Robey PG. Stem cells in tissue engineering. Nature
57. Aukhil I. Biology of wound healing. Periodontol 2000 2001;414:118–121.
2000;22:44–50.
81. Gronthos S, Zannettino AC, Hay SJ, et al. Molecular and cel-
58. Ivanovski S, Gronthos S, Shi S, Bartold PM. Stem cells in the lular characterisation of highly purified stromal stem cells
periodontal ligament. Oral Dis 2006;12:358–363. derived from human bone marrow. J Cell Sci 2003;116(Pt
59. Srisuwan T, Tilkorn DJ, Wilson JL, et al. Molecular aspects 9):1827–1835.
of tissue engineering in the dental field. Periodontol 2000 82. Horwitz EM, Prockop DJ, Fitzpatrick LA, et al. Transplant-
2006;41:88–108. ability and therapeutic effects of bone marrow-derived mesen-
60. Nakashima M, Akamine A. The application of tissue engi- chymal cells in children with osteogenesis imperfecta. Nat
neering to regeneration of pulp and dentin in endodontics. Med 1999;5:309–313.
J Endod 2005;31:711–718. 83. Jiang Y, Jahagirdar BN, Reinhardt RL, et al. Pluripotency of
61. Kaigler D, Mooney D. Tissue engineering’s impact on den- mesenchymal stem cells derived from adult marrow. Nature
tistry. J Dent Educ 2001;65:456–462. 2002;418:41–49.
62. Lin NH, Gronthos S, Bartold PM. Stem cells and periodontal 84. Pereira RF, Halford KW, O’Hara MD, et al. Cultured adher-
regeneration. Aust Dent J 2008;53:108–121. ent cells from marrow can serve as long-lasting precursor cells
for bone, cartilage, and lung in irradiated mice. Proc Natl
63. Taba M Jr, Jin Q, Sugai JV, Giannobile WV. Current con- Acad Sci U S A 1995;92:4857–4861.
cepts in periodontal bioengineering. Orthod Craniofac Res
2005;8:292–302. 85. Smith JR, Pochampally R, Perry A, Hsu SC, Prockop DJ. Iso-
lation of a highly clonogenic and multipotential subfraction
64. Bartold PM, Xiao Y, Lyngstaadas SP, Paine ML, Snead ML. of adult stem cells from bone marrow stroma. Stem Cells
Principles and applications of cell delivery systems for peri- 2004;22:823–831.
odontal regeneration. Periodontology 2000 2006;41:123–
135. 86. Friedenstein AJ, Gorskaja JF, Kulagina NN. Fibroblast pre-
cursors in normal and irradiated mouse hematopoietic organs.
65. Slavkin HC, Bartold PM. Challenges and potential in tissue Exp Hematol 1976;4:267–274.
engineering. Periodontol 2000 2006;41:41.
87. Friedenstein AJ, Piatetzky S II, Petrakova KV. Osteogenesis in
66. Hutmacher DW, Cool S. Concepts of scaffold-based tissue transplants of bone marrow cells. J Embryol Exp Morphol
engineering–the rationale to use solid free-form fabrication 1966;16:381–390.
techniques. J Cell Mol Med 2007;1:654–669.

128 © 2013 Australian Dental Association


Tissue engineered periodontal products

88. Woodbury D, Schwarz EJ, Prockop DJ, Black IB. Adult rat 108. Kolf CM, Cho E, Tuan RS. Mesenchymal stromal cells. Biol-
and human bone marrow stromal cells differentiate into neu- ogy of adult mesenchymal stem cells: regulation of niche,
rons. J Neurosci Res 2000;61:364–370. self-renewal and differentiation. Arthritis Res Ther 2007;
89. Petersen BE, Bowen WC, Patrene KD, et al. Bone marrow as 9:204.
a potential source of hepatic oval cells. Science 1999; 109. Friedenstein AJ. Stromal mechanisms of bone marrow: clon-
284:1168–1170. ing in vitro and retransplantation in vivo. Haematol Blood
90. Caplan AI. Mesenchymal stem cells. J Orthop Res 1991; Transfus 1980;25:19–29.
9:641–650. 110. Friedenstein AJ. Marrow stromal fibroblasts. Calcif Tissue Int
91. Owen ME, Cave J, Joyner CJ. Clonal analysis in vitro of oste- 1995;56(Suppl 1):S17.
ogenic differentiation of marrow CFU-F. J Cell Sci 1987;87 111. Owen M. Marrow stromal stem cells. J Cell Sci Suppl
(Pt 5):731–738. 1988;10:63–76.
92. Owen M. The origin of bone cells in the postnatal organism. 112. Wagner W, Wein F, Seckinger A, et al. Comparative charac-
Arthritis Rheum 1980;23:1073–1080. teristics of mesenchymal stem cells from human bone mar-
93. Owen M, Friedenstein AJ. Stromal stem cells: marrow- row, adipose tissue, and umbilical cord blood. Exp Hematol
derived osteogenic precursors. Ciba Found Symp 1988; 2005;33:1402–1416.
136:42–60. 113. Menicanin D, Bartold PM, Gronthos S. Stem cells in dental
94. Friedenstein AJ, Latzinik NV, Gorskaya YuF, Luria EA, structure regeneration. J Stomatol Invest 2007;1:3–13.
Moskvina IL. Bone marrow stromal colony formation 114. Li H, Yan F, Lei L, Li Y, Xiao Y. Application of autologous
requires stimulation by haemopoietic cells. Bone and Mineral cryopreserved bone marrow mesenchymal stem cells for peri-
1992;18:199–213. odontal regeneration in dogs. Cells Tissues Organs
95. Zannettino AC, Paton S, Arthur A, et al. Multipotential 2009;190:94–101.
human adipose-derived stromal stem cells exhibit a perivascu- 115. Kawaguchi H, Hirachi A, Hasegawa N, et al. Enhancement
lar phenotype in vitro and in vivo. J Cell Physiol of periodontal tissue regeneration by transplantation of bone
2008;214:413–421. marrow mesenchymal stem cells. J Periodontol 2004;
96. Kuznetsov SA, Mankani MH, Gronthos S, Satomura K, Bi- 75:1281–1287.
anco P, Robey PG. Circulating skeletal stem cells. J Cell Biol 116. Hasegawa N, Kawaguchi H, Hirachi A, et al. Behavior of
2001;153:1133–1140. transplanted bone marrow-derived mesenchymal stem cells in
97. Erices A, Conget P, Minguell JJ. Mesenchymal progenitor periodontal defects. J Periodontol 2006;77:1003–1007.
cells in human umbilical cord blood. Br J Haematol 117. Yamada Y, Ueda M, Hibi H, Baba S. A novel approach to
2000;109:235–242. periodontal tissue regeneration with mesenchymal stem cells
98. Portmann-Lanz CB, Schoeberlein A, Huber A, et al. Placental and platelet-rich plasma using tissue engineering technology:
mesenchymal stem cells as potential autologous graft for pre- a clinical case report. Int J Periodontics Restorative Dent
and perinatal neuroregeneration. Am J Obstet Gynecol 2006;26:363–369.
2006;194:664–673. 118. Tobita M, Uysal AC, Ogawa R, Hyakusoku H, Mizuno H.
99. Soncini M, Vertua E, Gibelli L, et al. Isolation and character- Periodontal tissue regeneration with adipose-derived stem
ization of mesenchymal cells from human fetal membranes. cells. Tissue Eng Part A 2008;14:945–953.
J Tissue Eng Regen Med 2007;1:296–305. 119. Boyko GA, Melcher AH, Brunette DM. Formation of new
100. In ‘t Anker PS, Noort WA, Scherjon SA, et al. Mesenchymal periodontal ligament by periodontal ligament cells implanted
stem cells in human second-trimester bone marrow, liver, in vivo after culture in vitro. A preliminary study of trans-
lung, and spleen exhibit a similar immunophenotype but a planted roots in the dog. J Periodontal Res 1981;16:73–88.
heterogeneous multilineage differentiation potential. Haema- 120. Gould TR, Melcher AH, Brunette DM. Migration and divi-
tologica 2003;88:845–852. sion of progenitor cell populations in periodontal ligament
101. In ‘t Anker PS, Scherjon SA, Kleijburg-van der Keur C, after wounding. J Periodontal Res 1980;15:20–42.
et al. Isolation of mesenchymal stem cells of fetal or mater- 121. Liu HW, Yacobi R, Savion N, Narayanan AS, Pitaru S. A col-
nal origin from human placenta. Stem Cells 2004;22:1338– lagenous cementum-derived attachment protein is a marker for
1345. progenitors of the mineralized tissue-forming cell lineage of the
102. Young HE, Steele TA, Bray RA, et al. Human reserve plurip- periodontal ligament. J Bone Miner Res 1997;12:1691–1699.
otent mesenchymal stem cells are present in the connective 122. Huang GT, Gronthos S, Shi S. Mesenchymal stem cells
tissues of skeletal muscle and dermis derived from fetal, derived from dental tissues vs. those from other sources: their
adult, and geriatric donors. Anat Rec 2001;264:51–62. biology and role in regenerative medicine. J Dent Res
103. Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria 2009;88:792–806.
for defining multipotent mesenchymal stromal cells. The 123. Gronthos S, Mankani M, Brahim J, Robey PG, Shi S. Postna-
International Society for Cellular Therapy position statement. tal human dental pulp stem cells (DPSCs) in vitro and
Cytotherapy 2006;8:315–317. in vivo. Proc Natl Acad Sci U S A 2000;97:13625–13630.
104. Gronthos S, Chen S, Wang CY, Robey PG, Shi S. Telomerase 124. Miura M, Gronthos S, Zhao M, et al. SHED: stem cells from
accelerates osteogenesis of bone marrow stromal stem cells human exfoliated deciduous teeth. Proc Natl Acad Sci U S A
by upregulation of CBFA1, osterix, and osteocalcin. J Bone 2003;100:5807–5812.
Miner Res 2003;18:716–722. 125. Sonoyama W, Liu Y, Fang D, et al. Mesenchymal stem cell-
105. Wada N, Menicanin D, Shi S, Bartold PM, Gronthos S. mediated functional tooth regeneration in swine. PLoS One
Immunomodulatory properties of human periodontal liga- 2006;1:e79.
ment stem cells. J Cell Physiol 2009;219:667–676. 126. Sonoyama W, Liu Y, Yamaza T, et al. Characterization of
106. Baksh D, Song L, Tuan RS. Adult mesenchymal stem cells: the apical papilla and its residing stem cells from human
characterization, differentiation, and application in cell and immature permanent teeth: a pilot study. J Endod
gene therapy. J Cell Mol Med 2004;8:301–316. 2008;34:166–171.
107. Porada CD, Zanjani ED, Almeida-Porad G. Adult mesenchy- 127. Morsczeck C, G€ otz W, Schierholz J, et al. Isolation of precur-
mal stem cells: a pluripotent population with multiple appli- sor cells (PCs) from human dental follicle of wisdom teeth.
cations. Curr Stem Cell Res Ther 2006;1:365–369. Matrix Biol 2005;24:155–165.

© 2013 Australian Dental Association 129


J Han et al.

128. Zhang QZ, Nguyen AL, Yu WH, Le AD. Human oral 136. Akizuki T, Oda S, Komaki M, et al. Application of periodon-
mucosa and gingiva: a unique reservoir for mesenchymal stem tal ligament cell sheet for periodontal regeneration: a pilot
cells. J Dent Res 2012;91:1011–1018. study in beagle dogs. J Periodontal Res 2005;40:245–251.
129. McCulloch CA, Bordin S. Role of fibroblast subpopulations 137. Tsumanuma Y, Iwata T, Washio K, et al. Comparison of different
in periodontal physiology and pathology. J Periodontal Res tissue-derived stem cell sheets for periodontal regeneration in a
1991;26(3 Pt 1):144–154. canine 1-wall defect model. Biomaterials 2011;32:5819–5825.
130. Isaka J, Ohazama A, Kobayashi M, et al. Participation of 138. Ding G, Liu Y, Wang W, et al. Allogeneic periodontal liga-
periodontal ligament cells with regeneration of alveolar bone. ment stem cell therapy for periodontitis in swine. Stem Cells
J Periodontol 2001;72:314–323. 2010;28:1829–1838.
131. Lang H, Sch€ uler N, Arnhold S, Nolden R, Mertens T. 139. Feng F, Akiyama K, Liu Y, et al. Utility of PDL progenitors
Formation of differentiated tissues in vivo by periodontal for in vivo tissue regeneration: a report of 3 cases. Oral Dis
cell populations cultured in vitro. J Dent Res 1995;74: 2010;16:20–28.
1219–1225. 140. Gault P, Black A, Romette JL, et al. Tissue-engineered liga-
132. Lang H, Sch€uler N, Nolden R. Attachment formation follow- ment: implant constructs for tooth replacement. J Clin Perio-
ing replantation of cultured cells into periodontal defects – dontol 2010;37:750–758.
a study in minipigs. J Dent Res 1998;77:393–405.
133. Liu Y, Zheng Y, Ding G, et al. Periodontal ligament stem Address for correspondence:
cell-mediated treatment for periodontitis in miniature swine. Professor PM Bartold
Stem Cells 2008;26:1065–1073.
Colgate Australian Clinical Dental Research Centre
134. Kim SH, Kim KH, Seo BM, et al. Alveolar bone regenera-
tion by transplantation of periodontal ligament stem
School of Dentistry
cells and bone marrow stem cells in a canine peri-implant The University of Adelaide
defect model: a pilot study. J Periodontol 2009;80: Frome Road
1815–1823.
Adelaide SA 5005
135. Park JY, Jeon SH, Choung PH. Efficacy of periodontal stem Australia
cell transplantation in the treatment of advanced periodonti-
tis. Cell Transplant 2011;20:271–285. Email: mark.bartold@adelaide.edu.au

130 © 2013 Australian Dental Association

You might also like