ArticlePDF Available

Abstract and Figures

All over the world a large number of people suffer from tooth diseases like dental caries, tooth abscess, and plaques. Tooth loss or damage, which occurs frequently in our society are generally repaired by applying several conventional methods, such as root-canal treatment, direct pulp capping and dental implants. These methods are quite painful, create damage to the surrounding tooth tissues and also may at times have adverse side-effects. The limitations of the conventional methods can be overcome by applying the concept of tooth tissue engineering. Tooth tissue engineering is the application of biosciences and engineering to regenerate a biofunctional tooth, which can be used to replace the missing tooth or repair the damaged tooth. Tissue engineering involves three key elements - cell, scaffold and growth factors, which interact with each other to regenerate a specific tissue. The success of tissue engineering depends on the proper selection of these three key elements and understanding the interactions among them. To bring us close to the realization of a tissue-engineered tooth, immense progress is going on in understanding how tooth is first developed, and there is a good advancement in tooth regeneration. In this review, “tooth tissue engineering” will be discussed, along with the recent advancements and challenges in bring a biofunctional tooth from laboratory out into clinical use.
Content may be subject to copyright.
Tooth Tissue Engineering: Potential and Pitfalls
Sweta Gupta1,a, Chhavi Sharma1,b, Amit K. Dinda2,c, Amiya K. Ray1,d,
Narayan C. Mishra1,e*
1 Department of Paper Technology, Indian Institute of Technology Roorkee, Saharanpur Campus,
Saharanpur-247001, U.P, India
2 Department of Pathology, All India Institute of Medical Sciences, New Delhi - 110029, India
a swetag009@gmail.com, b chhavisharma19@gmail.com, c amit_dinda@yahoo.com,
d amiyakumarray@gmail.com, e mishrawise@googlemail.com,
Keywords: Tooth; scaffold; tissue engineering; biomaterials; growth factors; stem cell.
Abstract. All over the world a large number of people suffer from tooth diseases like dental caries,
tooth abscess, and plaques. Tooth loss or damage, which occurs frequently in our society are
generally repaired by applying several conventional methods, such as root-canal treatment, direct
pulp capping and dental implants. These methods are quite painful, create damage to the
surrounding tooth tissues and also may at times have adverse side-effects. The limitations of the
conventional methods can be overcome by applying the concept of tooth tissue engineering. Tooth
tissue engineering is the application of biosciences and engineering to regenerate a biofunctional
tooth, which can be used to replace the missing tooth or repair the damaged tooth. Tissue
engineering involves three key elements - cell, scaffold and growth factors, which interact with each
other to regenerate a specific tissue. The success of tissue engineering depends on the proper
selection of these three key elements and understanding the interactions among them. To bring us
close to the realization of a tissue-engineered tooth, immense progress is going on in understanding
how tooth is first developed, and there is a good advancement in tooth regeneration. In this review,
“tooth tissue engineering” will be discussed, along with the recent advancements and challenges in
bring a biofunctional tooth from laboratory out into clinical use.
Introduction
Tooth is a small calcified structure, found in the entrance of the alimentary canal of both
invertebrates and vertebrates. It aids in grasping and processing of food, providing defense from
any attack, giving shape to the face and aiding clear speech. Presence of teeth also helps to prevent
dementia as mastication stimulates the brain [1]. Tooth plays an important role in overall health
and activity. Tooth loss or damage is quite frequent. All over the world, a large amount of
population suffers from tooth diseases like dental caries (cavities, holes in the teeth), tooth abscess,
and plaques. 20% of all inherited disorders are accounted for by tooth congenital abnormalities. In
the western world, 85% of adults have had some dental treatment and 7% have lost one or more
teeth by the age of 17 [2]. Tooth loss is a problem of great magnitude since approximately 35% of
the world population is edentulous by the age of 65. World Oral Health Report 2003, identified
dental caries as a continuing global problem, with an estimated five billion people worldwide,
having experienced the disease. Treatment costs are estimated as accounting for some 510% of
the healthcare budgets in industrialized countries [3]. Tooth imposes an incredible amount of
importance in our life although it is not essentially required for our survival. Missing tooth
adversely affects mastication, articulation, facial esthetics and psychological health, and it can result
in difficulty in movement of the remaining teeth and a lack of self-confidence. Though these are
not life-threatening, but the quality of daily life is adversely affected. It leads to physical and
mental suffering that can markedly lower an individual's quality of life. Loss or damage of tooth is
mainly because of dental wear lesions (abrasion, attrition, and erosion), bacterial infections,
chemicals, trauma, fractures and genetic defects. The major acquired pathological conditions that
affect tooth, concern the erosion of non-living elements (enamel and dentin), damage to living
tissues (pulp and periodontium) and the loss of whole tooth.
To repair the loss or damage of tooth, several conventional methods are applied, which include
extrinsic dental interventions such as tooth transplantation (allotransplantation, autotransplantation),
root-canal treatment, direct pulp capping, artificial dentures and dental implants (metal like titanium
or ceramics) [4]. But these methods are quite painful, create damage to the surrounding tooth
tissues and also sometimes have adverse side-effects because of several reasons, like
incompatibility between dental tissues and non-biological substitutes, insufficiency of host bone in
the jaw to accommodate the implant, poor mechanical properties of the periodontium and lack of
immunological properties of the crevicular sulcus [5, 6]. In addition, these approaches fail quite
frequently and have considerable costs. Even if the outcome is good, pathological repeats are
common [5, 6]. Therefore, there is a need for a better, biologically-oriented therapeutic approach,
which is “tooth tissue engineering” or “regeneration of biofunctional tooth”. Tooth tissue
engineering is the application of our knowledge of biosciences and engineering to regenerate a
biofunctional tooth, which can be used to replace the missing tooth by a bio-functional one or repair
the damaged tooth.
In this review, a concise part of the tooth anatomy, morphology and its development is
discussed. It is then followed by discussing the tissue engineering approaches to repair tooth and
describes the three key elements of tooth tissue engineering, i.e. dental stem cell sources, scaffold
biomaterials and the growth factors required for tooth tissue regeneration. Also discussed is recent
endeavours in tooth tissue engineering as well as the prospective outcome of the current
advancement in this line of research. Finally, the article will highlight significant challenges and
hurdles in bringing tooth tissue engineering out from laboratory to clinical use.
Tooth A Brief Overview of Basic Anatomy, Morphology and Tooth Development
Knowledge of tooth anatomy is necessary for understanding the pathological process and the
possible therapies. Due to different dietary habits and ecological adaptations, the tooth of
vertebrates acquires numerous anatomical forms and shapes such as incisors (cutting teeth), canines
(tearing teeth), premolars and molars (grinding teeth). These different shapes of teeth help in
protecting oral mucosa physiologically and aid to reduce stress forces on the tooth and the alveolar
bone. A whole tooth is divided into two parts - the crown and the root (Figure 1). The line of
junction between the crown and the root is called the cementum-enamel junction (cej - neck). The
crown emerges from the alveolar bone and is covered by enamel. The root is inserted into the bone
and is coated by cementum.
Although the morphology of tooth varies depending on species and location, but they are similar
in structure being organized into 3 parts - enamel organ (enamel), dental papilla (dentin and dental
pulp) and dental follicle (cementum, alveolar bone and periodontal ligament) (Figure 1). The hard
part of the tooth is composed of three different calcified tissues, enamel, dentin and cementum,
where enamel is the hardest of all these three. The dental pulp is the soft part and fills the centre of
the tooth. Tooth structural composition provides durability for a lifetime despite the enormous
abrasive forces, large temperature changes, varying pH and a moist and corrosive environment to
which teeth are continuously exposed [7]. The structural composition, physical properties and
different functions of tooth has been summarized in Table 1.
The morphological stages of tooth development consist of early bud stage, cap or proliferative
stage and bell stage, where tooth histo-differentiation and morpho-differentiation occurs, as
illustrated in Figure 2. Tooth development is characterized by highly orchestrated mutual inductive
interactions between two major cell types: stomodeal ectoderm (epithelial) cells and cranial, neural
crest-derived ectomesenchyme cells, which result in differentiation and spatial organization of cells
to form tooth-organs [13]. Stomodeal ectoderm cells form ameloblasts, which contributes to
prepare enamel tissues, whereas ectomesenchyme cells form odontoblasts, pulp cells,
cementoblasts, osteoblast and fibroblast (Table 2). Dental mesenchyme is termed
‘ectomesenchyme’ due to its earlier interaction with the neural crest [14]. These two types of cells,
stomodeal ectoderm cells and ectomesenchyme cells are always required for the growth of tooth
and the source of these cells is an important issue in tooth regeneration.
Figure 1. Anatomy of the tooth.
Figure 2. Schematic illustration of the main stages of tooth development: Oral epithelium
(ectoderm) forms a thickened epithelial band, dental lamina (a) budding of this epithelium towards
the underlying cranial neural crest-derived ectomesenchyme starts at the sites of future primary
teeth. Morphogenesis proceeds through the bud (b) and cap stages (c) to the bell stage (d) where
differentiation of odontoblasts (mesenchymal) and ameloblasts (epithelial) takes place. Finally
developing tooth starts erupting into the mouth (e).
Table 1. Structural composition, physical properties and functions of tooth tissues.
Tooth Tissue
Physical
Properties
Component
Protein
Cell constituents
Functions
Ref.
Enamel
Translucent,
vary in colour
from yellowish
to greyish
white, non-
living tissue
96%
inorganic
matter*,
1% organic
matter, and
3%water
Amelogenin
(90%),
ameloblastin,
enamelin and
tuftelin
Formed by dental
epithelial cells
(ameloblasts)
Endure crushing
pressure, produces a
cushioning effect of the
tooth's different
structures, and protects
the tooth from the wear
and tear of chewing.
[8]
Dentin
Light yellow,
calcified living
tissue, more
radiolucent
than enamel
and very
porous
70%
inorganic
matter*,
20% organic
matter, and
10%water
Dentin
sialoprotein,
dentin
phosphoprotein
and
dentin matrix
protein I
Formed by
odontoblasts that
line the walls of the
pulp cavity.
Living tissue transmits
pain stimuli by the way
of dentinal fibres.
[9]
Dental pulp
Hollow space
within the
dentin
Coronal pulp,
and Radicular
pulp
_
Composed of dental
mesenchymal cells
including putative
dental stem cells
(DSCs), fibroblasts,
macrophages and T
lymphocytes
Furnishes nourishment
to the dentin; provides
sensation to the tooth,
and responds to
irritation, either by
forming reparative
secondary dentin or by
becoming inflamed.
[10]
Cementum
Mineralized
tissue, light
yellow in
colour, covers
the roots of the
tooth in a thin
layer
45%
inorganic
matter*, 55%
organic
matter
Type I collagen
proteins
Formed by
precementoblast
Serves as an attachment
surface to anchor the
tooth to the bony walls
of the tooth sockets in
the periodontium and
protect the root.
[11]
Periodontium
Connective
tissues that
attach the
cementum to
the alveolar
bone via the
Sharpey's
fibres.
_
_
Osteoblasts and
osteoclasts present
on the alveolar
bone, fibroblasts,
epithelial cell rests
of Malassez, and
macrophages.
Support, protect and
provide nourishment to
the tooth, enable
sensory perception and
cushion mechanical
forces during
mastication as it has
shock absorbing
capacity.
[12]
* Inorganic matter - Calcium and Phosphorus as in hydroxyapatite
Table 2. Summary of derivation of tooth tissues.
Origin
Tooth Tissues formed
Cells involved
Ectoderm
Enamel
Ameloblast
Condensed
inner mass of
ectomesenchyme
Dentine
Dental pulp
Odontoblast (outer cells
of dental papilla)
Central cells of dental
papilla
Remaining
ectomesenchyme
surrounding outside the
enamel
Cementum
Alveolar bone
Periodontal Ligament
Cementoblast
Osteoblast
Fibroblast
Tissue Engineering Approach to Repair Tooth
Langer and Vacanti defined tissue engineering as an “interdisciplinary field that applies the
principles of engineering and life sciences toward the development of biological substitutes that
restore, maintain, or improve tissue function” [15]. Tooth tissue engineering may be described as
the regeneration of tooth by the application of our knowledge of biosciences and engineering. For
successful tooth tissue engineering, three prerequisites are needed: (1) sufficient numbers of cells
with odontogenic capacity; (2) an appropriate scaffold, a 3D porous architecture, to seed the cells;
and (3) factors to stimulate odontogenic differentiation in vivo. Despite of tooth's structural
complexity, the advancement of biomedical engineering techniques has given rise to two currently
employed approaches for tooth regeneration (Figure 3). The first approach is to regenerate tooth by
seeding stem cells in scaffolding biomaterials. The cell-seeded scaffold guides and supports tooth
formation. This technique has shown promising results in regeneration of the periodontium. The
second approach attempts to reproduce or mimic the developmental or “organotype” approach that
facilitates the development of tooth from epithelial and mesenchymal (stem) cells, either from tooth
germs or other sources. This approach uses embryonic tissues (dental epithelium and dental
mesenchyme) harvested from a mouse foetus and direct the formation of organized tooth through
epithelial-mesenchymal interactions in organ culture. It requires an understanding of the principles
that regulate early tooth development in the embryo [16]. The first method is more convincing
since its key elements are more controllable.
With the emerging concepts of tissue engineering, biological tooth may become an alternative
for replacing missing tooth. For biological replacement of tooth tissues or whole tooth, both in vivo
and in vitro, new strategies have been designed [17]. A major move ahead would be to reconstruct
tooth in patients without adequate bone support [18]. To grow replacement tooth in the individual’s
own mouth, would be a highly attractive therapy to permanently replace tooth in situ. Dentistry has
taken the same approach to tooth decay-filling cavities for decades, but new techniques for
rebuilding tooth from inside out could transform the profession over the next decade [19]. An
ambitious dream of numerous dentists is to be able to substitute the artificial material with a
biological, cell-based one that is able to form a genuine replica of the damaged tooth part or the
entire lost tooth. It should lead to a permanent solution to the damaged tooth without the need for
supplementary therapies, thus making it a cost-effective treatment in the long term.
Stem Cells used in Tooth Tissue Engineering
Stem cells have a self-renewing capacity that can regenerate any tissue for life time, and because of
this key property the stem cells are being used in tissue regeneration. Tooth organ contain two
types of stem cells Embryonic Stem (ES) cells and Adult Stem cells. Pluripotent ES cells are
derived from the inner cell mass of mammalian blastocysts and can be maintained indefinitely in
culture. When injected back into blastocysts, these cells contribute to all tissues, including germ
cells [20]. Thus, ES cells are a renewable source of cells that can be stimulated to differentiate into
precursors of any cell type. Adult dental stem contains several populations of stem cells, which
have been differentiated into two categories on the basis of their origin, that is whether they are
derived from mesodermal layer (dental ectomesenchymal stem cells) or ectodermal layer (dental
epithelial stem cells) of the embryo (Box 1). The postnatal epithelial and ectomesenchymal dental
stem cells present in immature tooth buds have demonstrated the ability to generate bioengineered
and anatomically correct but miniature-sized tooth crowns containing enamel, dentin, pulp and
alveolar bone [21].
Adult dental stem cells can be isolated at various stages of the organogenesis. Stem cells from
apical papilla (SCAP) and dental follicle precursor cells (DFPC) are present in tooth where
developmental program continues, whereas periodontal ligament stem cells (PDLF), dental pulp
stem cells (DPSC), and stem cells from human exfoliated deciduous tooth (SHED) represent the
cells present in tooth (Figure 4) which have already completed their development [22]. Dental stem
cells can also be isolated according to their anatomical locations, colony-forming ability, expression
of stem cell markers, and regeneration of pulp/dentin structures in vivo. Tooth tissue engineering
using stem cells is based on their isolation, association and culture as recombinants in vitro or ex
vivo conditions to assess tooth morphogenesis and cell differentiation into tooth specific cells that
will form enamel, dentin, cementum and alveolar bone. Dental stem cells are multipotent stem cells
having the future potential to differentiate into a variety of other cell types (Table 3) including
cardio myocytes to repair damaged cardiac tissue following a heart attack [23], neuronal to generate
nerve and brain tissue [24], myocytes to repair muscle [25], osteocytes to generate bone [26],
chondrocytes to generate cartilage and adipocytes to generate fat.
Dental tissues are specialized tissues that do not undergo continuous remodelling as shown in
bony tissues. Therefore, stem/progenitor cells derived from these tissues may be more committed
or restricted in their differentiation potency in comparison with bone marrow mesenchymal stem
cells (BMMSCs). The use of dental stem cells has been increased significantly over the past 15
years. Stem cells of dental origin can be used for various functions in dentistry, e.g., intended for
development of new tooth, management of periodontal diseases, for cleft palate treatment,
dentofacial orthopaedics corrective methods and geno-dental interrelations [35, 36].
Figure 3. Two current ways of tooth tissue engineering (a) Dissociated tooth germs seeded onto a
tooth-shaped scaffold, produced small complex tooth-like structures; (b) Epithelial and
mesenchymal (stem) cells, either from tooth germs or other sources, grown in organ culture and
form organized tooth through epithelial-mesenchymal interactions.
Box 1. Dental stem cells for tooth tissue engineering.
Figure 4. Derivation of Dental derived Stem cells. (a) Dental Follicle Progenitor cells (DFPCs)
isolated at early stage before tooth eruption. (b) Dental Pulp Stem cells (DPSC), Periodontal
Ligament Stem cells (PDLSC), and Apical Papilla Stem cells (APSC) isolated from permanent
tooth.
Table 3. Different stem cells with their differing degrees of proliferative potential and the
differentiation potency [27, 21].
Cell Type
Proliferative
potential
Differentiation Potency
Ref.
In vitro Analysis
(Multipotentiality)
In vivo Analysis (Ectopic
tissue formation)
DPSCs
++++
Osteo/Dentinogenic, Adipogenic,
Chondrogenic, Myogenic,
Neurogenic
Dentin-pulp-like complex,
Odontoblast-like cells, bone-
like tissue
[28]
SHED
++++++++
Dentinogenic, Adipogenic,
Chondrogenic, Myogenic,
Neurogenic, Osteoconductive
Dentin-pulp-like complex,
Odontoblast-like cells, No
dentin-pulp complex
formation, Bone formation
[29]
SCAP
++++
Dentinogenic, Adipogenic,
Chondrogenic, Myogenic,
Neurogenic
Dentin-pulp-like complex,
Odontoblast-like cells
[30]
PDLSCs
ND
Osteo/Cementogenic, Adipogenic,
Chondrogenic, Myogenic,
Neurogenic
PDL like formation,
Cementum like formation
[31]
DFPCs
ND
Cementogenic, Odontogenic,
Adipogenic, Chondrogenic,
Myogenic, Neurogenic
PDL like formation,
Cementum matrix formation
[32]
BMSC
++++++++
Osteogenic, Chondrogenic,Adipogenic,Neurogenic, Myogenic.
[33]
Human Embryonic
Stem Cells
+++
All three germ layer representatives
[34]
Scaffold Characteristics and Different Biomaterials Used for Tooth Tissue Engineering
Like cell source and type, scaffold is also one of the most important elements in tissue regeneration.
Scaffold provides a physicochemical and biological three-dimensional microenvironment for cell
growth, adhesion, migration and differentiation. Its complex structure and properties guide the cell
to differentiate into specific tissue. To mimic the physiological functions of the natural
extracellular matrix of the cells, scaffold must have to bear some unique characteristics. To address
the biomimetic requirements, a tooth scaffold should have the following characteristics [37, 38, 39].
1. relatively easy to handle;
2. allow for the incorporation of cells and act as a substrate for matrix deposition;
3. provide temporary mechanical support to the affected area;
4. contain a porous architecture to allow for vascularization and tissue-in-growth;
5. allow for the free diffusion of cells and growth factors;
6. permit the establishment of a vascular bed to ensure survival of the implanted cells;
7. should be effective for transport of nutrients, oxygen, and waste;
8. support and promote cell differentiation in the synthetic scaffold;
9. enhance cellular activity towards scaffold-host tissue integration;
10. degrade in a controlled manner to facilitate load transfer to developing tooth;
11. produce non-toxic degradation products;
12. do not incite an active chronic inflammatory response;
13. be capable of sterilization without loss of bioactivity;
14. deliver bioactive molecules or drugs in controlled manner to accelerate healing and prevent
pathology;
15. be ultimately biodegraded and replaced by regenerative tissue, retaining the feature of the
final tissue structure.
Biomaterials used for fabricating scaffold, should be biodegradable and biocompatible. The rate
of biodegradation should be equal to the rate of tissue formation; so that when the tissue is fully
developed the biomaterial will get completely degraded or absorbed in the body. For the scaffold
fabrication, various types of biomaterials, including natural and synthetic polymers, have been
employed (Table 4). Much of the body’s native extracellular matrix is composed of natural
materials like collagen, elastin, fibronectin and laminin. Natural materials, mostly natural
polymers, are cost effective, eco-friendly, have good biodegradability but they have some
disadvantages (Table 4). Some synthetic materials have also been used for tooth tissue engineering
due to flexibility in tailoring the physical, mechanical and chemical properties, and easy
processability of scaffold into desired shape and size. But they too have some disadvantages (Table
4). Therefore, nowadays composite scaffolds (combination of two or more polymers) are used to
overcome the disadvantages associated with the natural and synthetic polymers. Various composite
polymers used for tooth tissue engineering are listed in Table 4.
Table 4. Different scaffold biomaterials employed for tooth tissue engineering along with their
advantages and disadvantages
Scaffold biomaterials
Advantages
Disadvantages
Natural
materials
Bone sialoprotein [40]
Collagen [41]
Fibrin gel [41]
Chitosan [42]
Silk [43]
Platelet-rich plasma [44]
Mineral trioxide aggregate [45]
Cost effective, eco-friendly,
good biodegradability, low
toxicity, low manufacture costs,
low disposal costs and
renewability, have the
properties of biological
signalling, cell adhesion, cell
responsive degradation
Undergoing rapid
degradation, possibility of
losing their biological
properties during scaffold
fabrication processes, the
risk of immunorejection
and disease transmission.
Synthetic
materials
Polylactic acid [46]
Polyglycolic acid [46]
Polycaprolactone [46]
Polylactic-co-glycolic acid [47]
β-Tricalcium phosphate [48]
Hydroxyapatite [49]
Flexibility in tailoring the
physical, mechanical and
chemical properties, and easy
processability of scaffold into
desired shape and size
Adverse tissue reactions
caused by acidic
degradation products, lack
of cellular adhesion and
interaction.
Composite
materials
Glycidyl methacrylated
dextran/gelatin hybrid hydrogel [50]
Polyethyl methacrylate-co-
hydroxyethyl acrylate [48]
Gelatin-chondroitin-hyaluronan tri-
copolymer [51]
Polycaprolactone-gelatin-
hydroxyapatite tricopolymer [52]
More tailorable properties with
the possibility of controlling
biocompatibility, mechanical
properties, tunable degradation
kinetics and physical structures
to suit various applications.
Composite materials
generally don’t have any
disadvantage as they are
the combination of two or
more polymers to impart
various desired properties.
Growth Factors (Morphogens) for Tooth Regeneration
Tooth cannot be considered as a single organ, but somewhere between 3-16 organs, because each
tooth in the upper and lower jaw quadrants is different, making a possible eight different shapes per
jaw. In order to engineer a bio-tooth, an understanding of the molecular principle of tissue
developments and morphogenesis is a prerequisite factor. The promotion and maintenance of
morphogenesis is achieved by a variety of morphogens or growth factors. Growth factors are
naturally occurring proteins capable of stimulating cellular proliferation and differentiation. They
act as signalling molecules that coax the cells to differentiate into specific phenotype. These
signalling molecules help in tooth development and patterning. They also control stem cell activity
by increasing the rate of proliferation, inducing differentiation of the cells into another tissue type,
or stimulating stem cells to synthesize and secrete mineralized matrix. A variety of growth factors
have been successfully used for dentin-pulp complex regeneration, including Transforming Growth
Factors (TGFs) [53], Bone morphogenetic proteins (BMPs) [54], Platelet-derived growth factor
(PDGF) [55], Insulin-like growth factor (IGF) [56], Fibroblast Growth Factors (FGF), Epidermal
Growth Factors (EGF) and phosphates, as discussed in Table 5. According to the origin of stem
cells or cell source, various approaches have been used for tooth tissue regeneration, which are
outlined in Table 6.
Table 5. Signalling molecules, their origin, members, and functions in tooth tissue engineering.
Family
Growth
factor
Member
Primary Origin
Receptors
Functions
Ref.
TGF-β
super-
family
BMP
BMP2- BMP8
Dentine matrix, activated
TH1 cells (T-Helper) and
Natural Killer cells
Transmembrane
serine/threonine
kinase of type I
and type II BMP
receptors
Determine cusps number and position, and therefore the tooth patterning,
required for the transition of tooth development from the bud to the cap
stage, promotes proliferation, differentiation of mesenchymal cells, e.g.,
PDL fibroblasts, follicle cells, fibroblasts, toward a cell with capacity to
form mineral matrix, e.g., osteoblasts and cementoblasts.
[57]
FGF family
FGF
FGF 2,FGF 3,
FGF 4, FGF 8,
FGF 9,FGF 10
A wide range of cells
Notch pathway
Function at distinct steps of odontogenesis, from tooth initiation to the
formation of the last tooth cusp, restrict tooth forming sites by inducing
the expression of Pax9, Pitx1, and Pitx2. FGF10 is able to stimulate cell
proliferation only in the dental epithelium but not in the mesenchyme,
FGF4 is activated by the Wnt signalling pathway, promotes proliferation
and attachment of endothelial and PDL cells.
[58]
EGF family
EGF
EGF, TGF-α
Submaxillary glands
Tyrosine kinase
receptors
Stimulated cell proliferation in tooth explants but at the same time
inhibited tooth morphogenesis and dental cell differentiation.
[59]
IGF family
IGF
IGF-1, IGF-2
IGF-1-Liver
IGF-2 variety of cells
Tyrosine kinase
receptors
Promotes cell growth, migration, proliferation, differentiation and induce
the synthesis of enamel specific proteins like amelogenin and
ameloblastin.
[60]
PDGF
family
PDGF
PDGFA,
PDGFB,
PDGF C,
PDGF D
Platelets
Tyrosine kinase
receptors
PDGF A regulates the size and stage of tooth development.
[61]
Hedgehog
(Hh) family
Sonic
hedgehog
(Shh)
Shh
Notochord (Central nervous
system)
Patched receptor
Shh signalling is required for the development of early tooth germ
.When Hh signal is decreased below certain degree, the maxillary
incisors are missing.
[62]
Wnt
(Wingless
integrated)
gene family
WNT
Wnt1, Wnt2,
Wnt3,Wnt3a,
Wnt4,Wnt5a,
Wnt 10a,
Wnt 10b
secreted by specific cells to
regulate cellular programs in
the surrounding tissue
Frizzled family of
receptor and low-
density
lipoprotein (LDL)
receptor, known
as LRP5/6.
Play role in determination of the regions of the future tooth formation
and those that will form the remaining of the oral ectoderm.
Detailed function for each Wnt member in tooth development is not
clear at present.
[63]
Transferrin
_
_
Serum protein
_
Necessary growth factor for early tooth morphogenesis, required for the
development of bud- and early cap-staged tooth.
[59]
Emdogain;
Amelogen-
-ins
_
_
Tooth germ
_
Regeneration of periodontal tissues by periodontal cell attachment and
growth, contributes in cementum formation.
[64]
Parathyroid
hormone
(PTH)
_
_
Parathyroid gland
_
Epithelial-mesenchymal interactions during the formation of teeth, helps
in tooth eruption.
[65]
Phosphates
_
_
-
_
Cells within the periodontium are highly sensitive to changes in
phosphate/pyrophosphate levels, it may directly control expression of
genes associated with maturation of cementoblasts and osteoblasts.
[66]
Table 5. Various approaches used for regeneration of dental tissues, according to the source of cells and biomaterials used [67].
Approach
Cell Source
Technique
Biomaterial
Relevance
Ref.
Periodontal
regeneration
Canine (beagle)
Harvest cells, seed onto collagen
sponge, implant against
Periodontium
Collagen sponge (70%Type 1,
30% Type 2)
Potential of in situ tissue engineering using
autologous cells for the regeneration of
periodontal tissues.
[68]
periodontal ligament Stem
cells of miniature pig
Harvest cells, seed onto collagen
sponge, implant against
Periodontium
Hydroxyapatite/ tricalcium
phosphate scaffold
Feasibility of using stem cell-mediated tissue
engineering to treat periodontal diseases.
[69]
Endo-dontal
regeneration
Human stem cells from
exfoliated deciduous tooth
Seed cells onto scaffold and place
in prepared canals of human tooth
D,D-L,L-polylactic acid scaffold
Possible to implant tissue-engineered pulp into
tooth after cleaning and shaping.
[70]
Hard tissue
Apical-pulp-derived
cells, human molar
Harvest of human apical pulp,
expansion in vitro, seed onto
hydroxyapatite scaffold, implant
subcutaneously into nude mice
Porous hydroxyapatite scaffold
The human tooth with an immature apex is an
effective source of cells for hard-tissue
regeneration.
[71]
Scaffold-
based tooth
regeneration
Tooth bud cells, rat pups
Harvest, in vitro expansion, seed
onto scaffold for in vivo
maturation
Porous hexafluoroisopropanol
(HFIP) silk scaffolds
Generation of mineralized tissues for tooth-tissue
engineering; use of silk scaffold.
[72]
Tooth bud cells, rat pups
Harvest, in vitro expansion, seed
onto scaffold for in vivo
maturation
PGA and PLGA scaffold
materials
Tooth-tissue engineering methods can be used to
generate both pig and rat tooth tissues.
[73]
Tooth bud cells, porcine
crown
Harvest, seed onto PGA mesh,
implant into omentum of rat.
PGA fibre mesh
Development of tissue-engineered tooth closely
resembles the pattern of odontogenesis.
[74]
Tooth bud cells, porcine
molar
Harvest, seed tooth cells onto
scaffold, implant into omentum of
rat, join with bone grown in
bioreactor, re-grow in rat.
PGA and PLGA scaffold
materials
Generation of hybrid toothbone for the eventual
clinical treatment of tooth loss accompanied by
alveolar bone resorption.
[75]
Organotype-
based tooth
regeneration
Dissociated single cells
from epithelial and mesen-
chymal tissues, recombined
dissociated cells
Harvest of murine tooth bud cells,
implant into tooth socket
Collagen
Proximity of ectodermal and mesenchymal cells
necessary for tooth development; generation of a
structurally correct tooth with penetration of
blood vessels and nerve tissue.
[76]
Tooth bud cells, rat pup
Isolation of tooth bud cells and
co-culture with dental pulp stem
cells, pelletize and culture in renal
capsule
N/A
Mimic the dentinogenic microenvironment from
tooth germ cells in vitro. Demonstrate that
soluble factors can produce a conditioned
medium beneficial for in vitro growth.
[77]
Rat marrow stromal
cells, mouse
embryonic stem cells
mouse embryonic
neural stem cells
Cultured embryonic oral
epithelium with other
mesenchymal cells, transfer tooth
primordium to jaw to grow tooth.
Cell pellet wrapped in epithelium
N/A
Embryonic oral tissue can guide differentiation
of other stem cells to odontoblasts; embryonic
primordium can develop in the adult
environment; generation of a functional tooth.
[78]
Cellular Homing: A Promising Approach for Tooth Regeneration
Stem cell seeding within a scaffold in vitro, was a conventional approach, which aims to mimic
cellular structure and regenerate a functional tissue equivalent in vitro or in vivo. Cellular homing
is an approach which attempts to regenerate tissue in vivo without cell-seeding. Homing is the
process by which cells migrate to and become engrafted in the tissue, on which they exert local,
functional and reparative effects. By using this process, body's own stem cells can be directed
toward the growth factor impregnated scaffolds, made of natural materials, at specific anatomical
sites. Once the stem cells have colonized the scaffold, a tissue can grow and then get merged with
the surrounding tissue. Kim and co-workers used polycaprolactone and hydroxyapatite to fabricate
tooth-shaped scaffold of human molar and rat incisor by applying three-dimensional bio printing
containing 200μm diameter interconnecting micro channels [79]. Two cell homing factors: stromal-
derived factor (SDF)-1 and bone morphogenetic protein (BMP)-7 were infused into the scaffold
micro channels, before implantation. Following mandibular incisor extraction in each of the 22
rats, a rat incisor scaffold was immediately implanted orthotopically, and simultaneously, a human
molar scaffold was implanted ectopically into the dorsum of each rat. The scaffold was colonized
by cells even without exogenously providing cells with the scaffold and for the first time de novo
formation of tooth-like structures and periodontal integration in vivo was found [79]. This strategy
avoids ex vivo cell culture and minimizes the invasiveness associated with clinical procedures. Cell
homing could provide enhancements in cellular methodology for tissue engineering and a novel,
minimally invasive option for tissue regeneration [80]. Because of harnessing the body’s innate
ability to heal and regenerate, it proves to be a meritorious approach in tooth tissue engineering [80,
81, 82]. It is worth mentioning that the tooth tissue regeneration can be achieved by endogenous
regenerative technology that avoids the expense and difficulties associated with culture, storage and
distribution of cells, and immune considerations [82].
Recent Advances in Tooth Tissue Engineering
Partial tooth structures had been regenerated well before the concept of tissue engineering was
introduced by Langer and Vacanti in 1993. In the pulp-capping procedure, dentin production was
known to be induced by calcium hydroxide, although the underlying mechanism was not known
[83]. In 1952, the pioneering work of Shirley Glasstone-Hughes demonstrated that early-stage
embryonic tooth primordia can be split into two, and each half can regenerate a complete normal
size tooth [84]. This established that the early-stage tooth primordia have an inherent level of
plasticity and regenerative capacity. The regenerative capacity of early-stage tooth primordia was
utilized in experiments by Young and co-workers [85], where scaffolds had been fabricated in the
shape of different teeth and cells, dissociated from early-stage third molar tooth germs from pigs
and rats, were seeded onto these scaffolds. The seeded scaffolds were grown in the omentum of
immune compromised rats: histological analysis revealed the formation of tiny (12 mm) tooth
crowns 2030 weeks after the in vivo implantation using porcine tooth buds [85]. The rat molar
tooth germ developed just after 12 weeks of in vivo implantation [86]. It demonstrated the
existence of stem cells in tooth primordia which are able to regenerate teeth. The demonstration of
bioengineered whole tooth crowns from pig and rat tooth bud cells provided the first evidence that
post-natal dental stem cells could be used for whole-tooth tissue engineering applications [85, 86].
Recent studies have indicated that cell-based strategies show promising potential for regenerating
the whole tooth structure in rodents [86, 87, 89]. Moreover, stem cell-based regeneration of human
tooth structures has been achieved in immunocompromised mouse models [90, 91]. One study
indicated that hybrid tooth-bone tissues were bioengineered by seeding the pig third molar tooth
bud cells onto the polyglycolide (PGA) and polyglycolide-co-lactide (PLGA) scaffolds, and
growing for 4 weeks in the omenta of adult rat hosts. There is a tremendous progress in dental stem
cell biotechnology and cell mediated murine tooth regeneration, which encouraged the researchers
to explore the potential for regeneration of living tooth with appropriate functional properties. The
recent advances in tooth tissue engineering are discussed below.
Advances in 2006:
It was proposed that bone-marrow-stem cells have the potential to turn into both dental
mesenchyme and dental epithelium offering novel possibilities for tooth-tissue engineering
[92].
Bone marrow mesenchymal stem cells (BMMSCs) were isolated from beagle dogs and
transplanted into experimental Class III periodontal defects. Four weeks after
transplantation the defects were almost regenerated with periodontal tissue. This suggests
that transplanted BMMSCs could survive and differentiate into periodontal tissue cells,
resulting in enhancement of periodontal tissue regeneration. These findings suggest that
auto-transplantation of bone marrow mesenchymal stem cells is a novel option for
periodontal tissue regeneration [93].
Advances in 2007:
Enamel-dentin complex structures were reconstituted by the cervical loop region cells (CLC)
in recombination with dental pulp horn cell population. In contrast, dentin-cementum
complex structures were reconstituted by cervical loop region cells in recombination with the
dental pulp horn cell population (PHC) within the dental cusp. This shows that cervical loop
cells play a key role in the reconstitution of tooth structure [94].
It was demonstrated that immortalized mouse dental follicle cells are able to regenerate a
PDL-like tissue in vivo [95].
It was reported a new technique for regeneration of whole tooth through production of tissue
to replace damaged or missing enamel. Epithelial cells extracted from the developing tooth
of 6-month-old pigs continue to proliferate when they are cultured on top of a special feeder
layer of cells (3T3-J2 cell line). This crucial step boosts the number of dental epithelial cells
available for enamel production and helps in tooth regeneration [96].
Mesenchymal and epithelial cells were sequentially seeded into a collagen gel drop ex vivo.
After implantation into subrenal capsules in mice, a correct tooth structure comprising
enamel, dentin, root, dental pulp and bone was observed, showing penetration of blood
vessels and nerve fibres [97].
Advances in 2008
Dentin pulp like tissue had been regenerated when polylactic-co-glycolic acid polymeric
porous scaffolds was seeded with dental pulp stem cells. In this study, it was observed that
the cells formed mineralized-like structures even without the addition of any differentiation
chemicals [98].
Autologous PDLSCs are capable of forming bone, cementum and periodontal ligament if
transplanted onto hydroxyapatite tricalcium phosphate (HA/TCP) carrier into periodontal
defects created surgically. For periodontal regeneration, non-dental derived stem cells have
been tested. ASCs (Adipose-derived Stem Cells) isolated from rat and mixed with platelet-
rich plasma (PRP), were implanted into the periodontal tissue defect generated in the test rat.
Partial alveolar bone regeneration and a periodontal ligament-like structure were observed 8
weeks after implantation [99].
Human exfoliated deciduous teeth (SHED) constitute a viable source of stem cells for dental
pulp tissue engineering. A new dental pulp-like tissue was engineered, when SHED was
seeded onto dentine slices and implanted subcutaneously in immunodeficient mice. It was
also demonstrated that SHED when co-implanted with human endothelial cells, improve the
newly formed tissue-organization, microvascular network, and oxygen and nutrient influx.
It was shown that the resulting tissue presented architecture and cellularity that closely
resemble those of a physiologic dental pulp [100].
For biological tooth to perform adequately, they must be able to grow and integrate into the
existing alveolar bone of the jaws. Amazingly, implanted embryonic tooth germs can
develop normally and form tooth when transplanted into the adult mouse. Further analyses
using 3D imaging have shown that the tooth not only have normal morphology, but also can
erupt into the oral cavity and form functional roots [16].
A viable biological substitute of a tooth was engineered by seeding the tooth shaped scaffold
with dental germs and placing it in rat jaw or omentum [101]. It has been demonstrated that
a full tooth can be produced by using the techniques of tissue engineering as illustrated in
Figure 5.
Figure 5. Schematic showing the process of bioengineering dental tissue which is grown in the
omentum or rat jaw.
Advances in 2009
The hybrid tooth-bone construct was prepared by combining tooth-bud cell seeded scaffolds
with autologous iliac-crest bone-marrow-derived stem cell seeded scaffolds, which were
transplanted into surgically created mandibular defects in the minipig. It was found small
tooth-like structures consisting of organized dentin, enamel, pulp, cementum, periodontal
ligament, surrounded by regenerated alveolar bone. This study suggests the possibility for
regeneration of teeth and associated alveolar bone, in a single procedure [102].
Evidence for engineering tooth enamel has been found that will help people to replace lost
parts of the tooth with a healthy layer of new enamel. A simple amino acid, proline, which
is repeated in the centre of proteins found in tooth enamel, makes teeth stronger and more
resilient. When the repeats are short, such as in frogs, teeth will not have the enamel prisms
that are responsible for the strength of human enamel. In contrast, when the proline repeats
are long such as in human being, they contact groups of molecules that help enamel crystals
grow [103].
Advances in 2010
Use of heterogeneous population of stem cell-mediated de novo regeneration of pulp-dentin
tissue has been demonstrated. It was found that pulp-like tissue can be regenerated de novo
in emptied root canal space by stem cells from apical papilla and dental pulp stem cells that
give rise to odontoblast-like cells producing dentin-like tissue on existing dentinal walls.
Stem/progenitor cells from apical papilla and dental pulp stem cells were isolated,
characterized, seeded onto synthetic scaffolds consisting of poly-D, L-lactide/glycolide,
inserted into the tooth fragments, and transplanted into mice. Continuous layer of dentin
like tissue has been shown to be formed on the canal dentinal walls (Figure 6) and on
mineral trioxide aggregate (MTA) cement surface [104].
Figure 6. SCID mouse as subcutaneous study model for pulp-dentin regeneration. The canal space
of human tooth root fragments (*67 mm long) was enlarged to *12.5 mm in diameter. One end
of the canal opening was sealed with MTA cement (SCID, severe combined immunodeficient;
MTA, mineral trioxide aggregate) (reproduced with permission from reference 104).
It was shown that in mice the Jagged2 gene is required for the healthy development of teeth.
Inactivation of this gene interrupts the Notch signalling pathway and due to this tooth
crowns will be malformed (Figure 7) and enamel will be lacking [105].
Figure 7, Shape of a healthy (left) and deformed molar with the Jagged2-gene deactivated (right) in
the mouse embryo (reproduced with permission from reference 105).
Advances in 2011
Recently, there is a finding which assures great promise for regenerative medicine. Many
types of cells including DPSCs, SHEDs, and SCAPs have been successfully reprogrammed
into induced Pluripotent stem (iPS) cells. Autologous DPSCs are isolated from the patient’s
own dental pulps and pluripotent iPS cells are created by driving four genes (c-Myc, Klf4,
Oct3/4, and Sox2) into DPSCs, which can be used to generate dental epithelial cells under
suitable conditions. These iPS-derived dental epithelial cells are recombined with
autologous DPSCs pellet and transplanted in vivo to make a bio-tooth. After the temporary
incubation in vivo, the bio-tooth can be transplanted into the patient’s jaws to treat tooth
loss. These dental iPS cells express the marker genes that characterize embryonic stem
cells, and maintain the developmental potential to differentiate into advanced derivatives of
all three primary germ layer [106, 107].
Takashi Tsuji and his co-workers [108] isolated stem cells from the molar teeth of mice, and
these stem cells were placed in a mold to grow. When the full tooth units were grown, it
was transplanted into the jaws of one month old mice. After 40 days, it was observed that
the transplanted teeth get fused with the jaw bones, and nerve fibres were also present in the
growing teeth. The mice that received the transplanted teeth were able to eat and chew
normally without any complications.
Challenges and Future Prospective in Tooth Tissue Regeneration
The term “tissue engineering” is now almost 35 years old, and significant progress has been made
in tooth tissue engineering during the last decades. But still, tissue engineering has not yet matured
enough into such a level so that the engineered tooth is widely accepted as autograft or allograft for
dental treatments. The regeneration of a whole tooth structure including enamel, dentin/pulp
complex, and periodontal tissues as a functional unit in humans is a challenge with the currently
available regenerative biotechnologies. It has been accepted that formation of multiple miniature
tooth crowns in the bioengineered tooth constructs is possible, but real-size whole-tooth
regeneration encounters a number of challenges due to the complexity of human tooth growth and
development. Therefore, there are still significant hurdles to overcome in developing tissue
engineered tooth and to bring it out successfully from the laboratory to clinical use. Some of the
multifold challenges, as described below, must be surmounted for rapid progress in tooth tissue
engineering.
1. Source of stem cells - Stem cells can be obtained from any source, autogenic or allogenic.
Although the autogenic stem cells have no risk of immune rejection, are least expensive and
have no ethical concerns, but to use them is time consuming procedure. Using allogenic
stem cells saves considerable time but the risk of immune rejection and pathogen
transmission limits their use. Xenogenic tooth bud cells may lead to dysmorphogenesis of
regenerated teeth, even if it is applicable to humans. Isolation, culture and storage of these
stem cells are technique-sensitive, and their immunomodulatory characteristics are still
questionable.
2. Non-vital nature of enamel - Enamel is fundamental to oral function, and yet it is
frequently the first to be lost. Innovative solutions are required to compensate for significant
loss of enamel.
3. Control of further development within the body - As the tissue engineered parts have
been developed using very potent signal molecules to induce the alteration of the growth of
stem cells, a way has to be found to insure that these alteration and growth will not continue
beyond control when implanted.
4. Implantation of the tissue engineered parts - From the stage of fabrication to the stage of
implantation, these tissue-parts have to be kept alive, and they have to find a functional
place within the human body, with a proper supply of blood, oxygen and nutrients. In
dentistry, implanting a tissue engineered tooth would be a very difficult task, when contact
with the bone, size, shape and positions of the implanted tooth are so important to a proper
and functional implantation.
5. Aging - One of the obstacles is to match the aging of the implanted tissue engineered parts
with that of the surrounding tissues and organs.
6. Cost - tissue engineered products should be economical so that the patient can afford the
treatment.
7. Storage - Cryopreservation or hypothermic storage of tissue-engineered tooth tissues should
be made so that when needed it is easily available.
8. Issues involved in the reconstruction of a bio-tooth - shape determination, tooth size
(engineered tooth is smaller than that of normal) availability of dental epithelium, directional
growth and eruption, spatial organized pulp, dental and enamel, mechanical strength and
graft rejection in the jaws remain to be resolved.
Conclusions
A new discipline in dentistry, ‘regenerative dentistry’, has been resulted due to recent advancements
in scaffold designing, better understanding of growth factor biology and interactions between
allogenic stem cells and immune system. It provides an increased level of understanding of the
mechanical and chemical stimuli that regulate tissue responses. Tissue engineering certainly has the
advantage over the other options in its low cost, small recovery time, non-intrusiveness, and as a
permanent fix to the problem. With the studies and development to come in the future, it will be
able to prove to be the most effective and long lasting procedures. The accumulating knowledge on
tooth tissue engineering can be applied to regenerate missing osseous or dental structures, repair
and replace mineralized tissues, promote oral wound healing, correct orofacial deformities and the
periodontal surgery. Biomechanical principles can also be applied to tissue engineering to enhance
the bone/tooth or bone/implant functionality and long-term stability. Dental tissue-engineering
companies, such as Dentigenix and Odontis, and large-scale enterprises such as Hitachi, have
greatly accelerated the research process and have also introduced the commercial products. Several
researches are going on mice but upscaling to humans, appears to be highly challenging.
Advancements are also being achieved in the area of biomimetics that will allow the creation of
new biologic replacements for missing oral structures. Several advancements have been made but
still improvement is necessary in the success rate of tooth formation and in the morphology of
regenerated tooth in tissue engineering methods. The opportunity for bioengineering to employ the
course of tooth regeneration is an exciting prospect and will improve the quality of life for patients
for decades to come. Tissue engineering does not end here, but will have a future in many aspects
of the medical field and each of our lives.
Acknowledgements
The authors are grateful to the Council of Scientific and Educational Research (Grant No.
27(0222)/1 O/EM R-II dated 31.05.10), India, for funding this work.
References
[1] T. Nakahara, Y. Ide. Tooth regeneration: implications for the use of bioengineered organs in
first wave organ replacement, Hum Cell, 2007, 20 (3) 63-70.
[2] P.T Sharpe, C.S Young. Test-tube teeth, Sci Am, 2005, 293(2), 34-41.
[3] P.E Petersen, Editor. Continuous improvement of oral health in the 21st century the
approach of the WHO Global Oral Health Programme. World Health Organization (2003).
[4] E. Crubzy, P. Murail, L. Girard, J.P Bernadou. False teeth of the Roman world, Nature,
1998, 391, 29.
[5] M. Esposito, J.M Hirsch, U. Lekholm, P. Thomsen. Biological factors contributing to
failures of osseointegrated oral implants (II), Etiopathogenesis, Eur J Oral Sci. 1998, 106
(3), 721-64.
[6] G.J Christensen. How to kill a tooth, J Am Dent Assoc. 2005, 136 (12) 1711-13.
[7] K.M Galler, R.N D’Souza, J.D Hartgerink. Biomaterials and their potential applications for
dental tissue engineering, J. Mater. Chem. 2010, 20, 873046.
[8] T. Uchida, C. Murakami, N. Dohi, K. Wakida, T. Satoda, O. Takahashi. Synthesis,
secretion, degradation, and fate of ameloblastin during the matrix formation stage of the rat
incisor as shown by immunocytochemistry and immunochemistry using region-specific
antibodies, J Histochem Cytochem. 1997, 45 (10) 1329-40.
[9] W.T Butler, J.C Brunn, C. Qinm, M.D McKee. Extracellular matrix proteins and the
dynamics of dentin formation, Connect. Tissue Res. 2002, 43(2-3) 301-7.
[10] R.E Walton, M. Torabinejad. Principles and Practice of Endodontics, third ed. W.B.
Saunders Company: Philadelphia, Pennsylvania, 2002, 11-3.
[11] M.I Cho, P.R Garante. Radioautographic study of [3H] mannose utilization during
cementoblast differentiation, formation of acellular cementum and development of
periodontal ligament principle fibres, Anat. Rec. 1989, 223(2) 209-22.
[12] R.T Cate, Periodontium, in: Oral Histology, Development, Structure, and Function, Edited
J.D John, J. Pendill, Mosby Elsevier: St. Louis. Missouri, China (1994), 239.
[13] I. Thesleff. Epithelial-mesenchymal signalling regulating tooth morphogenesis, J Cell Sci.
2003, 116 (9) 1647-8.
[14] P. Hölttä. Developmental aberrations of permanent teeth after high-dose anticancer therapy
in childhood [dissertation], Department of Paediatric and Preventive Dentistry, University of
Helsinki, Finland (2005).
[15] R. Langer, J. P Vacanti. Tissue engineering, Science 1993, 260 (5110) 9206.
[16] A.H.H Yen, P.T Sharpe. Stem cells and tooth tissue engineering, Cell Tissue Res. 2008, 331
(1) 359-72.
[17] C.F Ferreira, R.S Magini, P.T Sharpe. Biological tooth replacement and repair, J Oral
Rehabil. 2007, 34 (12) 933-9.
[18] W. Sonoyama, Y. Liu, D. Fang, T. Yamaza, B.M Seo, C. Zhang. Mesenchymal stem cell-
mediated functional tooth regeneration in swine, PLoS One, 2006, 1 e79.
[19] M. Taba Jr., Q. Jin, J.V Sugai, W.V Giannobile. Current concepts in periodontal
bioengineering, Orthod Craniofac Res. 2005, 8 (4) 292-302.
[20] E. Fuchs, J.A Segre. Stem Cells: A New Lease on Life, Cell 2000, 100 (1) 143-155.
[21] O. Trubiani, G. Orsini, S. Caputi, A. Piatelli. Adult mesenchymal stem cells in dental
research: a new approach for tissue engineering, Int J Immunopathol Pharmacol. 2006, 19
(3) 451-60.
[22] B. Inanç, Y.M Elçin. Stem Cells in Tooth Tissue Regeneration - Challenges and
Limitations, Stem Cell Rev and Rep. 2011, 7 (3) 683-92.
[23] C. Gandia, A. Arminan, J.M García-Verdugo, E. Lledó, A. Ruiz, M.D Miñana, J. Sanchez-
Torrijos, R. Payá, V. Mirabet, F. Carbonell-Uberos, M. Llop, J.A Montero, P. Sepúlveda.
Human dental pulp stem cells improve left ventricular function, induce angiogenesis, and
reduce infarct size in rats with acute myocardial infarction, Stem Cells 2008, 26 (3) 638-45.
[24] I.V Nosrat, J. Widenfalk, L. Olson, C.A Nosrat. Dental pulp cells produce neurotrophic
factors, interact with trigeminal neurons in vitro, and rescue motoneurons after spinal cord
injury, Dev Biol. 2001, 238 (1) 120-32.
[25] I. Kerkis, C.E Ambrosio, A. Kerkis, D.S Martins, E. Zucconi, S.A Fonseca, R.M Cabral,
C.M Maranduba, T.P Gaiad, A.C Morini, N.M Vieira, M.P Brolio, O.A Sant'Anna, M.A
Miglino, M. Zatz. Early transplantation of human immature dental pulp stem cells from
baby teeth to golden retriever muscular dystrophy (GRMD) dogs: Local or Systemic, J.
Transl Med. 2008, 6, 35-37.
[26] A. Graziano, R. d’Aquino, G. Laino, G. Papaccio. Dental pulp stem cells: a promising tool
for bone regeneration, Stem Cell Rev. 2008, 4 (1) 21-6.
[27] V. Govindasamy, A.N Abdullah, V.S Ronald, S. Musa, Z.A.C Ab. Aziz, R.B Zain. Inherent
Differential Propensity of Dental Pulp Stem Cells Derived from Human Deciduous and
Permanent Teeth, J. Endodontics 2010, 36 (9) 1504-15.
[28] W. Zhang, X.F Walboomers, S. Shi, M. Fan, J.A Jansen. Multilineage differentiation
potential of stem cells derived from human dental pulp after cryopreservation, Tissue Eng.
2006, 12 (10) 2813-23.
[29] M. Miura, S. Gronthos, M. Zhao, B. Lu, L.W Fisher, P.G Robey, S. Shi. SHED: Stem cells
from human exfoliated deciduous teeth, PNAS USA 2003, 100 (10) 5807-5812.
[30] S. Abe, S. Yamaguchi, T. Amagasa. Multilineage cells from apical pulp of human tooth
with immature apex, Oral Science Int. 2007, 4 (1) 45-58.
[31] J. Xu, W. Wang, Y. Kapila, J. Lotz, S. Kapila. Multiple differentiation capacity of STRO-
1+/CD146+ PDL mesenchymal progenitor cells, Stem Cells Dev. 2009, 18 (3) 487-96.
[32] B. Inanç, A.E Elçin, Y.M Elçin. Osteogenic induction of human periodontal ligament
fibroblasts under two-and three-dimensional culture conditions, Tissue Eng. 2006, 12 (2)
257-66.
[33] J. Yu, Y. Wang, Z. Deng, L. Tang, Y. Li, J. Shi, Y. Jin. Odontogenic capability: bone
marrow stromal stem cells versus dental pulp stem cells, Biol. Cell 2007, 99 (8) 465-74.
[34] G.T Huang, S. Gronthos, S. Shi. Mesenchymal stem cells derived from dental tissues vs
those from other sources: their biology and role in regenerative medicine, Journal of Dental
Research 2009, 88 (9) 792-806.
[35] M.E Yalvac, M. Ramazanoglu, A.A Rizvanov, F. Sahin, O. Bayrak, U. Salli, A. Palotás, G.T
Kose. Isolation and characterization of stem cells derived from human third molar tooth
germs of young adults: implications in neo-vascularization, osteo-, adipo- and neurogenesis,
The Pharmacogenomics Journal 2010, 10 (2) 105-13.
[36] Y. Oda, Y. Yoshimura, H. Ohnishi, M. Tadokoro, Y. Katsube, M. Sasao, Y. Kubo, K.
Hattori, S. Saito, K. Horimoto, S. Yuba, H. Ohgushi. Induction of Pluripotent Stem Cells
from Human Third Molar Mesenchymal Stromal Cells, The Journal of Biological Chemistry
2010, 285 (38) 29270-8.
[37] M.P Lutolf, J.A Hubbell. Synthetic biomaterials as instructive extracellular
microenvironments for morphogenesis in tissue engineering, Nat. Biotechnol. 2005, 23 (1)
47-55.
[38] D.H Kempen, L. Lu, C. Kim, X. Zhu, W.J Dhert, B.L Currier, M.J Yaszemski. Controlled
drug release from a novel injectable biodegradable microsphere/scaffold composite based on
poly(propylene fumarate), J. Biomed Mater Res A 2006, 77 (1) 103-11.
[39] T. Jiang, W.I Abdel-Fattah, C.T Laurencin. In vitro evaluation of chitosan/poly (lactic acid-
glycolic acid) sintered microsphere scaffolds for bone tissue engineering, Biomaterials 2006,
27 (28) 4894-4903.
[40] F. Decup, N. Six, B. Palmier, D. Buch, J.J Lasfargues, E. Salih, M. Goldberg. Bone
sialoprotein-induced reparative dentinogenesis in the pulp of rat’s molar, Clin Oral Investig.
2000, 4 (2) 110-9.
[41] T. Ohara, T. Itaya, K. Usami, Y. Ando, H. Sakurai, M.J Honda, M. Ueda, H. Kagami.
Evaluation of scaffold materials for tooth tissue engineering, J Biomed Mater Res A 2010,
94 (3) 800-5.
[42] H.C Arca, S. Şenel. Chitosan Based Systems for Tissue Engineering, J. Pharm. Sci. 2008,
33, 35-49.
[43] W.P Xu, W. Zhang, R. Asrican, H.J Kim, D.L Kaplan, P.C Yelick. Accurately shaped tooth
bud cell-derived mineralized tissue formation on silk scaffolds, Tissue Eng Part A 2008, 14
(4) 549-57.
[44] A. Gandhi, N. Kathuria, T. Gandhi, A. Arya. Microscale Technologies for Tooth Tissue
Engineering and Regenerative Endodontics, Internet Journal of Bioengineering 2011, 4 (2).
[45] G. Bogen, J.S Kim, L.F Bakland. Direct Pup capping with Mineral Trioxide aggregate, J
Am Dent Assoc. 2008, 139 (3) 305-315.
[46] K.A Athanasiou, G.G Niederauer, C.M Agrawal. Sterilization, toxicity, biocompatibility
and clinical applications of polylactic acid/polyglycolic acid copolymers, Biomaterials 1996,
17 (2) 93-102.
[47] C.S Young, H. Abukawa, R. Asrican, M. Ravens, M.J Troulis, L.B Kaban, J.P Vacanti, P.C
Yelic. Tissue-engineered hybrid tooth and bone Tissue Eng, 11 (2005) 1599-1610.
[48] A. Vallés-Lluch, E. Novella-Maestre, M. Sancho-Tello, M.M Pradas, G.G Ferrer, C.C
Batalla. Mimicking natural dentin using bioactive nanohybrid scaffolds for dentinal tissue
engineering, Tissue Engineering - Part A, 16 (2010) 2783-2793.
[49] S. Shi, P.M Bartold, M. Miura, B.M Seo, P.G Robey, S. Gronthos. The efficacy of
mesenchymal stem cells to regenerate and repair dental structures, Orthod Craniofacial Res.,
8 (2005) 191-199.
[50] F.M Chen, Y.M Zhao, R. Zhang, T. Jin, H.H Sun, Z.F Wu, Y. Jin. Periodontal regeneration
using novel glycidyl methacrylated dextran (Dex-GMA)/gelatin scaffolds containing
microspheres loaded with bone morphogenetic proteins, Journal of Controlled Release, 121
(2007) 81-90.
[51] T.F Kuo, H.C Lin, A.T Hung, K.C Yang, H.F Lin, T.K Tanng, S.T Chen, A.H.J Wang.
Gelatin-chondroitin-hyaluronan tri-copolymer scaffold seeded with dental bud cells for
odontogenesis: An ex vivo study on nude mice, Biomedical Engineering - Applications,
Basis and Communications, 22 (2010) 535-547.
[52] X. Yang, F. Yang, X.F Walboomers, Z. Bian, M. Fan, J.A Jansen. The performance of
dental pulp stem cells on nanofibrous PCL/gelatin/nHA scaffolds, Journal of Biomedical
Research, Part A. 93 (2010) 247-257.
[53] K. Dobie, G. Smith, A.J Sloan, A.J Smith. Effects of alginate hydrogels and TGF-β1 on
human dental pulp repair in vitro, Connective Tissue Research, 43 (2002) 387-390.
[54] A.J Sloan, R.B Rutherford, A.J Smith. Stimulation of the rat dentine-pulp complex by bone
morphogenetic protein-7 in vitro, Archives of Oral Biology, 45 (2000) 173-177.
[55] S. Yokose, H. Kadokura, N. Tajima, A. Hasegawa, H. Sakagami, K. Fujieda, T. Katayama.
Platelet-derived growth factor exerts disparate effects on odontoblast differentiation
depending on the dimers in rat dental pulp cells, Cell and Tissue Research, 315 (2004) 375-
384.
[56] H. Lovschall, O. Fejerskov, A. Flybjerg. Pulp-capping with recombinant human insulin-like
growth factor I (rhIGF-I) in rat molars, Advances in Dental Research, 15 (2001) 108-112.
[57] J. Jernvall, I. Thesleff. Reiterative signalling and patterning during mammalian tooth
morphogenesis, Mech Dev., 92 (2000) 19-29.
[58] H. Peters, R. Balling. Teeth: where and how to make them, Trends Genet., 15 (1999) 59-65.
[59] T. Mitsiadis, H. Magloire. Epidermal growth factor in tooth development, Actual
Odontostomatol., 44 (1990) 257-70.
[60] K. Takahashi, A. Yamane, P. Bringas, J. Caton, H.C Slavkin, M. Zeichner-David. Induction
of amelogenin and ameloblastin by insulin and insulin-like growth factors (IGF-I and IGF-
II) during embryonic mouse tooth development in vitro, Connect Tissue Res., 38 (1998)
269-78.
[61] Y. Chai, P. Jr Bringas, A. Mogharei, C.F Shuler, H.C Slavkin. PDGF-A and PDGFR-alpha
regulate tooth formation via autocrine mechanism during mandibular morphogenesis in
vitro, Dev Dyn., 213 (1998) 500-511.
[62] H. Tian, T. Tenzen, A.P McMahon. Dose dependency of Disp1and genetic interaction
between Disp1 and other hedgehog signalling components in the mouse, Development, 131
(2004) 4021-4033.
[63] J.K Sethi, A. Vidal-Puig. Wnt signalling and the control of cellular metabolism, Biochem.
J., 427 (2010) 117.
[64] S. Gestrelius, S.P Lyngstadaas, L. Hammarström. Emdogain - periodontal regeneration
based on biomimicry, Clin Oral Invest., 4 (2000) 120-125.
[65] W.M Philbrick, B.E Dreyer, I.A Nakchbandi, A.C Karaplis. Parathyroid hormone-related
protein is required for tooth eruption, PNAS, 95 (1998) 11846-11851.
[66] T.L. Rodriguez, K.J Nagatomo, B.L Foster, F.H Nociti, M.J Somerman. Modulation of
Phosphate/Pyrophosphate Metabolism to Regenerate the Periodontium. A Novel In Vivo
Approach, J Periodontol. (2011). Epub.
[67] S.A Hacking, A. Khademhosseini. Applications of Microscale technologies for regenerative
dentistry, J Dent Res., 88 (2009) 409-421.
[68] T. Nakahara, T. Nakamura, E. Kobayashi, K. Kuremoto, T. Matsuno, Y. Tabata. In situ
tissue engineering of periodontal tissues by seeding with periodontal ligament-derived cells,
Tissue Eng., 10 (2004) 537-544.
[69] Y. Liu, Y. Zheng, G. Ding, D. Fang, C. Zhang, P.M Bartold. Periodontal ligament stem
cell-mediated treatment for periodontitis in miniature swine, Stem Cells, 26 (2008) 1065-
1073.
[70] E.L Gotlieb, P.E Murray, K.N Namerow, S. Kuttler, F. Garcia-Godoy. An ultrastructural
investigation of tissue-engineered pulp constructs implanted within endodontically treated
teeth, J Am Dent Assoc., 139 (2008) 457-465.
[71] S. Abe, S. Yamaguchi, A. Watanabe, K. Hamada, T. Amagasa. Hard tissue regeneration
capacity of apical pulp derived cells (APDCs) from human tooth with immature apex,
Biochem Biophys Res Commun., 371 (2008) 90-93.
[72] W.P Xu, W. Zhang, R. Asrican, H.J Kim, D.L Kaplan, P.C Yelick. Accurately shaped tooth
bud cell-derived mineralized tissue formation on silk scaffolds, Tissue Eng Part A, 14
(2008) 549-557.
[73] M.T Duailibi, S.E Duailibi, C.S Young, J.D Bartlett, J.P Vacanti, P.C Yelick.
Bioengineered teeth from cultured rat tooth bud cells, J Dent Res., 83 (2004) 523-528.
[74] M.J Honda, Y. Sumita, H. Kagami, M. Ueda. Histological and immuno-histochemical
studies of tissue engineered odontogenesis, Arch Histol Cytol., 68 (2005) 89-101.
[75] C.S Young, H. Abukawa, R. Asrican, M. Ravens, M.J Troulis, L.B Kaban. Tissue-
engineered hybrid tooth and bone, Tissue Eng., 11 (2005) 1599-1610.
[76] K. Nakao, R. Morita, Y. Saji, K. Ishida, Y. Tomita, M. Ogawa. The development of a
bioengineered organ germ method, Nat Methods, 4 (2007) 227-230.
[77] J. Yu, Z. Deng, J. Shi, H. Zhai, X. Nie, H. Zhuang. Differentiation of dental pulp stem cells
into regular-shaped dentin-pulp complex induced by tooth germ cell conditioned medium,
Tissue Eng., 12 (2006) 3097-3105.
[78] A. Ohazama, S.A. Modino, I. Miletich, P.T. Sharpe. Stem-cell-based tissue engineering of
murine teeth, J Dent Res, 83 (2004) 518-522.
[79] K. Kim, C.H Lee, B.K Kim, J.J Mao. Anatomically shaped tooth and periodontal
regeneration by cell homing, J Dent Res., 89 (2010) 842-847.
[80] J.T Schantz, H. Chim, M. Whiteman. Cell Guidance in Tissue Engineering: SDF-1
Mediates Site-Directed Homing of Mesenchymal Stem Cells within Three-Dimensional
Polycaprolactone Scaffolds, Tissue Engineering., 13 (2007) 2615-2624.
[81] C.H Evans, G.D Palmer, A. Pascher, R. Porter, F.N Kwong, E. Gouze. Facilitated
endogenous repair: making tissue engineering simple, practical, and economical, Tissue
Eng., 13 (2007) 1987-1993.
[82] F.M Chen, J. Zhang, M. Zhang, Y. An, F. Chen, Z.F Wu. A review on endogenous
regenerative technology in periodontal regenerative medicine, Biomaterials, 31 (2010) 7892-
7927.
[83] H.A Zander. Reaction of the pulp to calcium hydroxide, J Dent Res., 18 (1939) 373.
[84] S. Glasstone-Hughes. The development of halved tooth germs; a study in experimental
morphology, J Anat., 86 (1952) 12-25.
[85] C.S Young, S. Terada, J.P Vacanti, M. Honda, J.D Bartlett, P.C Yelick. Tissue engineering
of complex tooth structures on biodegradable polymer scaffolds, J Dent Res., 81 (2002) 695-
700.
[86] M.T Duailibi, S.E Duailibi, C.S Young, J.D Bartlett, J.P Vacanti, P.C Yelick.
Bioengineered teeth from cultured rat tooth bud cells, J Dent Res., 83 (2004) 523-528.
[87] Y. Chai, H.C Slavkin. Prospects for tooth regeneration in the 21st century: A perspective,
Microsc. Res. Tech., 60 (2003) 469-479.
[88] A. Ohazama, S.A Modino, I. Miletich, P.T Sharpe. Stem cell based tissue engineering of
murine teeth, J Dent. Res., 83 (2004) 518-522.
[89] A.H Yen, P.T Sharpe. Regeneration of teeth using stem cell-based tissue engineering,
Expert Opin. Biol Ther., 6 (2006) 9-16.
[90] S. Gronthos, J. Brahim, W. Li, L.W Fisher, N. Cherman, A. Boyde, P. Den Besten, P.G
Robey, S. Shi. Stem cell properties of human dental pulp stem cells, J Dent Res., 81 (2002)
531-535.
[91] S. Gronthos, M. Mankani, J. Brahim, P.G Robey, S. Shi. Postnatal human dental pulp stem
cells (DPSCs) in vitro and in vivo, Proc. Natl. Acad. Sci., 97 (2000) 13625-13630.
[92] B. Hu, F. Unda, S. Bopp-Kuchler, L. Jimenez, X.J Wang, Y. Haïkel, S.L Wang, H. Lesot.
Bone Marrow Cells can give rise to Ameloblast-like Cells, J. Dent. Res., 85 (2006) 416.
[93] N. Hasegawa, H. Kawaguchi, A. Hirachi, K. Takeda, N. Mizuno, M. Nishimura, C. Koike,
K. Tsuji, H. Iba, Y. Kato, H. Kurihara. Behaviour of transplanted bone marrow-derived
mesenchymal stem cells in periodontal defects, J Periodontol., 77 (2006) 1003-1007.
[94] M. Honda, Y. Sumita, H. Kagami, I. Asahina, M. Ueda. Enamel-Dentin and Dentin-
Cementum complex structure formation in tooth-tissue regeneration, European Cells and
Materials, 14 (2007) 60.
[95] T. Yokoi, M. Saito, T. Kiyono, S. Iseki, K. Kosaka, E. Nishida, T. Tsubakimoto, H. Harada,
K. Eto, T. Noguchi, T. Teranaka. Establishment of immortalized dental follicle cells for
generating periodontal ligament in vivo, Cell Tissue Res., 327 (2007) 301-311.
[96] M. Honda. Tissue-engineering of Enamel-Dentin Complex Structures. Proceeding 1780 of
85th General Session of the International Association for Dental Research, (2007) University
of Tokyo, Japan.
[97] K. Nakao, R. Morita, Y. Saji, K. Ishida, Y. Tomita, M. Ogawa, M. Saitoh, Y. Tomooka, T.
Tsuji. The development of a bioengineered organ germ method, Nat Methods, 4 (2007)
227-230.
[98] R.M El-Backly, A.G Massoud, A.M El-Badry, R.A Sherif, M.K Marei. Regeneration of
dentine/pulp-like tissue using a dental pulp stem cell/poly(lactic-co-glycolic) acid scaffold
construct in New Zealand white rabbits, Aust Endod J, 34 (2008) 52-67.
[99] M. Tobita, A.C Uysal, R. Ogawa, H. Hyakusoku, H. Mizuno. Periodontal tissue
regeneration with adipose-derived stem cells, Tissue Eng Part A, 14 (2008) 945-953.
[100] M. Cordeiro, Z. Dong, T. Kaneko, Z. Zhang, M. Miyazawa, S. Shi, A.J Smith, J.E Nör.
Dental Pulp Tissue Engineering with Stem Cells from Exfoliated Deciduous Teeth, Journal
of Endodontics, 34 (2008) 962-969.
[101] S.E Duailibi, M.T Duailibi, W. Zhang, R. Asrican, J.P Vacanti, P.C Yelick. Bioengineered
Dental Tissues Grown in the Rat Jaw, J Dent Res., 87 (2008) 745-750.
[102] W. Zhang, H. Abukawa, M.J Troulis, L.B Kaban, J.P Vacanti, P.C Yelick. Tissue
engineered hybrid tooth-bone constructs, Methods, 47 (2009) 122-128.
[103] T. Jin, Y. Ito, X. Luan, S. Dangaria, C. Walker, M. Allen, A. Kulkarni, C. Gibson, R. Braatz,
X. Liao, T.G.H Diekwisch. Supramolecular compaction through polyproline motif
elongation as a mechanism for vertebrate enamel evolution, PLoS Biology, 7 (2009)
e1000262.
[104] G.T.J Huang, T. Yamaza, L.D Shea, F. Djouad, N.Z Kuhn, R.S Tuan, S. Shi.
Stem/progenitor cell-mediated de novo regeneration of dental pulp with newly deposited
continuous layer of dentin in an in vivo model, Tissue Eng Part A,. 16 (2010) 605-615.
[105] T.A Mitsiadis, D. Graf, H. Luder, T. Gridley, G. Bluteau. BMPs and FGFs target Notch
signalling via jagged 2 to regulate tooth morphogenesis and cytodifferentiation,
Development, 137 (2010) 3025-3035.
[106] X. Yan, H. Qin, C. Qu, R. S. Tuan, S. Shi, G.T. Huang. iPS cells reprogrammed from
human mesenchymal-like stem/progenitor cells of dental tissue origin, Stem Cells and
Development 2010, 19 (4) 469-480.
[107] M. Yan, Y. Yu, G. Zhang, C. Tang, J. Yu. A Journey from Dental Pulp Stem Cells to a Bio-
tooth, Stem Cell Rev and Rep., 2011, 7 (1) 161-171.
[108] M. Oshima, M. Mizuno, A. Imamura, M. Ogawa, M. Yasukawa, H. Yamazaki, R. Morita, E.
Ikeda, K. Nakao, T. Takano-Yamamoto, S. Kasugai, M. Saito, T. Tsuji. Functional Tooth
Regeneration Using a Bioengineered Tooth Unit as a Mature Organ Replacement
Regenerative Therapy, PLoS One, 2011, 6 (7) e21531.
... The first one involved the dissociation of tooth germ that can be seeded onto a tooth-shaped scaffold supporting tooth formation and is then transplanted to generate multiple complex tooth-like structures. The second approach relied on the interaction between epithelial and mesenchymal SCs obtained from primordial tooth germs or other sources, which induced tooth growth in culture [98]. ...
... They should also prevent the production of non-toxic molecules and chronic inflammatory responses while delivering necessary regulatory signals in a controlled way to promote healing. Finally, scaffolds should be biodegradable, allowing their replacement by regenerated tissues [98,101]. ...
... Natural biomaterials -such as natural polymers (collagen, laminin, elastin, chitosan, silk, platelet-rich plasma, bone sialoprotein, to name a few) -usually have low toxicity, are eco-friendly, and are cheaper than synthetic biomaterials. They are preferred for cell adhesion, cell-responsive degradation, proper cell signaling, and rapid degradation without immune rejection [98][99][100][101]. Tooth tissue engineering also uses synthetic materials -such as hydroxyapatite, polylactic acid, polyglycolic acid, polycaprolactone, and poly lactic-coglycolic acid -which are often more flexible and elastic than natural ones. ...
Article
Full-text available
One of the most exciting advances in life science research is the development of 3D cell culture systems to obtain complex structures called organoids and spheroids. These 3D cultures closely mimic in vivo conditions, where cells can grow and interact with their surroundings. This allows us to better study the spatio-temporal dynamics of organogenesis and organ function. Furthermore, physiologically relevant organoids cultures can be used for basic research, medical research, and drug discovery. Although most of the research thus far focuses on the development of heart, liver, kidney, and brain organoids, to name a few, most recently, these structures were obtained using dental stem cells to study in vitro tooth regeneration. This review aims to present the most up-to-date research showing how dental stem cells can be grown on specific biomaterials to induce their differentiation in 3D. The possibility of combining engineering and biology principles to replicate and/or increase tissue function has been an emerging and exciting field in medicine. The use of this methodology in dentistry has already yielded many interesting results paving the way for the improvement of dental care and successful therapies. Graphical abstract
... They work in signal transduction pathways to regulate cell proliferation, migration, and differentiation into specific phenotypes. BMPs, PDGF, FGF, TGF, EGF, and IGFs are the most common WNT proteins [87][88][89]. ...
Article
Full-text available
Maintaining dental pulp vitality and preventing tooth loss are two challenges in endodontic treatment. A tooth lacking a viable pulp loses its defense mechanism and regenerative ability, making it more vulnerable to severe damage and eventually necessitating extraction. The tissue engineering approach has drawn attention as an alternative therapy as it can regenerate dentin-pulp complex structures and functions. Stem cells or progenitor cells, extracellular matrix, and signaling molecules are triad components of this approach. Stem cells from human exfoliated deciduous teeth (SHED) are a promising, noninvasive source of stem cells for tissue regeneration. Not only can SHEDs regenerate dentin-pulp tissues (comprised of fibroblasts, odontoblasts, endothelial cells, and nerve cells), but SHEDs also possess immunomodulatory and immunosuppressive properties. The collagen matrix is a material of choice to provide structural and microenvironmental support for SHED-to-dentin pulp tissue differentiation. Growth factors regulate cell proliferation, migration, and differentiation into specific phenotypes via signal-transduction pathways. This review provides current concepts and applications of the tissue engineering approach, especially SHEDs, in endodontic treatment.
... Bone grafts, whether autogenous or allogenous, have limitations due to inadequate availability of ideal bone substitute, immune rejection, secondary trauma and osteogenic potential of the grafts [3,4]. To overcome these limitations, various studies are being done to regenerate bio-functional skeletal tissues by application of tissue engineering, which involves cells, scaffolds (a 3D polymeric network) and growth factors (signaling cues) [5]. Although all the three components are very important for tissue regeneration, but the role of scaffold is of prior importance because it interact with cells and growth factors for proper adhesion, growth, proliferation and differentiation of cells over the scaffold to regenerate a specific tissue. ...
Article
In the present study, goat-lung scaffold was fabricated by decellularization of lung tissue and verified for complete cell removal by DNA quantification, DAPI and H&E staining. The scaffold was then modified by crosslinking with quercetin and nanohydroxyapatite (nHAp), and characterized to evaluate the suitability of quercetin-crosslinked nHAp-modified scaffold for regeneration of bone tissue. The crosslinking chemistry between quercetin and decellularized scaffold was established theoretically by AutoDock Vina program (in silico docking study), which predicted multiple intermolecular hydrogen bonding interactions between quercetin and decellularized scaffold, and FTIR spectroscopy analysis also proved the same. From MTT assay and SEM studies, it was found that the quercetin-crosslinked nHAp-modified decellularized scaffold encouraged better growth and proliferation of bone-marrow derived mesenchymal stem cells (BMMSCs) in comparison to unmodified decellularized scaffold, quercetin-crosslinked decellularized scaffold and nHAp-modified decellularized scaffold. Alkaline Phosphatase (ALP) assay results showed highest expression of ALP over quercetin-crosslinked nHAp-modified scaffold among all the tested scaffolds (unmodified decellularized scaffold, quercetin-crosslinked decellularized scaffold and nHAp-modified decellularized scaffold) indicating that quercetin and nHAp is very much efficient in stimulating the differentiation of BMMSCs into osteoblast cells. Alizarin red test quantified in vitro mineralization (calcium deposits), and increased expression of alizarin red over quercetin-crosslinked nHAp-modified scaffold indicating better stimulation of osteogenesis in BMMSCs. The above findings suggest that quercetin-crosslinked nHAp-modified decellularized goat-lung scaffold provides biomimetic bone-like microenvironment for BMMSCs to differentiate into osteoblast and could be applied as a potential promising biomaterial for bone regeneration.
... Nowadays, scientists have been focusing on fabricating scaffolds using multi-polymers (more than two polymers) to mimic the properties of ECM, which also consists of multi-polymers [8,[12][13][14][15][16][17][18][19]. Here, in this study, we focus on fabricating composite scaffold with a combination of multi-polymers along with hydroxyapatite, for bone tissue engineering applications. ...
Article
A novel nano-biocomposite scaffold was fabricated in bead form by applying simple foaming method, using a combination of natural polymers — chitosan, gelatin, alginate and a bioceramic — nano-hydroxyapatite (nHAp). This approach of combining nHAp with natural polymers to fabricate the composite scaffold, can provide good mechanical strength and biological property mimicking natural bone. Environmental scanning electron microscopy (ESEM) images of the nano-biocomposite scaffold revealed the presence of interconnected pores, mostly spread over the whole surface of the scaffold. The nHAp particulates have covered the surface of the composite matrix and made the surface of the scaffold rougher. The scaffold has a porosity of 82% with a mean pore size of 112 ± 19.0 μm. Swelling and degradation studies of the scaffold showed that the scaffold possesses excellent properties of hydrophilicity and biodegradability. Short term mechanical testing of the scaffold does not reveal any rupturing after agitation under physiological conditions, which is an indicative of good mechanical stability of the scaffold. In vitro cell culture studies by seeding osteoblast cells over the composite scaffold showed good cell viability, proliferation rate, adhesion and maintenance of osteoblastic phenotype as indicated by MTT assay, ESEM of cell–scaffold construct, histological staining and gene expression studies, respectively. Thus, it could be stated that the nano-biocomposite scaffold of chitosan–gelatin–alginate–nHAp has the paramount importance for applications in bone tissue-engineering in future regenerative therapies.
... A lot of research work has been carried on chondrogenic differentiation of Mesenchymal Stem Cells (MSCs) [65][66][67][68][69]. In recent years, tissue engineering has gained attention as a potential technique for the treatment of various diseases including osteoarthritis [70][71][72][73][74][75][76]. Gupta et al. have recently used decellularized goat-lung scaffold from cadaver goat-lung tissue, by modifying with chitosan/ nanohydroxyapatite composite and shown that the chondrocytes cells which were layered on this scaffold can grow very well without changing their phenotypes. ...
Article
Full-text available
Stem cells are highly specialized biological cells that have the potential to differentiate and regenerate various cells and tissue types. Stem cell research was first initiated way back in 80’s due to the discovery of pluripotent embryonic stem cells. However, embryonic stem cells utilization spurred controversy which encouraged scientists to find an alternative for the substitution for ESCs. Human Mesenchymal Stem Cell (MSC), a type of an adult stem cell, exhibits many of the properties as that of embryonic stem cells such as pluripotency, immunomodulatory etc. These adult stromal cells are multipotent and non-hematopoietic in nature. MSCs have the potential to differentiate into diverse cell types including osteocytes, osteoblasts, adipocytes, chondrocytes, cardiomyocytes, neural cells and β-cells of islets. This differentiation property is exploited by many researchers in clinical and therapeutic applications of these cells against wide variety of diseases including osteoarticular diseases, neurodegenerative diseases, auto-immune diseases, cardiovascular diseases and in renal transplantation. Overall, this review summarises an importance of use of human Mesenchymal Stem cells (MSCs) for regenerative therapies for the cure of these major diseases in human.
... Therefore, to solve the concerns related to current surgical skeletal reconstruction, there is a necessity to develop an ideal bone substitute or biofunctional bone which should mimic the natural bone and replace the lost/damaged bone in the human body. The biofunctional bone can be regenerated by the approach of tissue engineering, which involves 3 important elements: (i) cells, (ii) growth factors, and (iii) scaffold (a 3D porous architecture made of biomaterials, whereupon the cells are seeded) [3]. The interactions among these 3 elements (cells, scaffold, and growth factors) control the process of tissue regeneration. ...
Article
Full-text available
Decellularized goat-lung scaffold was fabricated by removing cells from cadaver goat-lung tissue, and the scaffold was modified with chitosan/nanohydroxyapatite composite for the purpose of bone tissue engineering applications. MTT assay with osteoblasts, seeded over the chitosan/nanohydroxyapatite-modified decellularized scaffold, demonstrated significantly higher cell growth as compared to the decellularized scaffold without modification. SEM analysis of cell-seeded scaffold, after incubation for 7 days, represented a good cell adhesion, and the cells spread over the chitosan/nanohydroxyapatite-modified decellularized scaffold. Expression of bone-tissue-specific osteocalcin gene in the osteoblast cells grown over the chitosan/nanohydroxyapatite-modified decellularized scaffold clearly signifies that the cells maintained their osteoblastic phenotype with the chitosan/nanohydroxyapatite-modified decellularized scaffold. Therefore, it can be concluded that the decellularized goat-lung scaffold-modified with chitosan/nanohydroxyapatite composite, may provide enhanced osteogenic potential when used as a scaffold for bone tissue engineering.
Article
Full-text available
Quaternary composite scaffold consisting of chitosan, alginate, gelatin, and silk fibroin, was fabricated by applying foaming method, for tissue engineering applications. The fabricated scaffold was evaluated for its applicability in skin tissue regeneration. The environmental scanning electron microscopy (ESEM) showed the presence of interconnected pores, mostly spread over the entire surface of the scaffold with mean pore size 92611.8 lm and the porosity 88%. The scaffold showed good mechanical stability under physiological conditions as determined by short term mechanical stability testing. In vitro scaffold-degradation study showed no degradation at day 1 and from day 3 scaffold starts degrading. The degradation of the composite scaffold after 28 days was 38%. Less degradation rate of the scaffold might be beneficial, as it can provide sufficient time for the formation of neo-tissue and extracellular matrix (ECM) during tissue regeneration. In vitro cell culture studies by seeding L929 mouse fibroblast cells over composite scaffold showed good cell viability, proliferation, and adhesion as indicated by 3-(4,5-dimethylthiazol-2-yl-2,5-diphenyltetrazolium bromide) (MTT) assay and ESEM of cell-scaffold construct. Giemsa staining of L929 fibroblast cells over the scaffold showed fibroblastic morphology of L929 cells, having elongated cells with nuclei and faint cytoplasm, and these cells are positive for Oil Red stain and negative for Alizarin Red staining—indicating that they maintained their dermal fibroblastic phenotype and were not differentiated into any other cell types in presence of composite scaffold. Results of histological staining supports growth and viability of L929 fibroblasts over scaffold, thereby proving the great prospective of this scaffold for skin tissue engineering applications.
Article
Full-text available
In the present study, goat-lung was decellularized and used as a scaffold to support cellular attachment and proliferation. The biological properties of decellularized goat-lung scaffold were enhanced by modifying it with collagen and gelatin, and the effect of modification was evaluated on the growth of human skin-derived mesenchymal stem cells (S-MSCs). MTT assay demonstrated good biocompatibility, cell adhesion and proliferation of S-MSCs over collagen and gelatin modified scaffolds as compared to unmodified decellularized scaffold. Scanning electron microscopy observation confirmed the distribution of S-MSCs in the gelatin and collagen modified decellularized scaffold. Hematoxylin and Eosin staining further revealed that collagen and gelatin modified decellularized scaffold maintained good biocompatibility with S-MSCs. Expression of Keratin18 gene, along with CD105, CD73 and CD44, by S-MSCs over collagen and gelatin modified decellularized scaffold signifies that the cells are viable, proliferating and migrating, and have maintained their critical cellular functions within the scaffold. All the above results suggests that modification of goat-lung decellularized scaffold by collagen and gelatin enhances the growth and proliferation of S-MSCs, and therefore, collagen and gelatin modified decellularized scaffold could be a potential candidate for bio-artificial skin tissue engineering applications.
Article
A biologically regenerated tooth may provide a new treatment for tooth loss. In this study, a tissue engineering approach was applied to demonstrate the tooth regeneration. The dental buds of the second molar tooth from 1.5-month-old miniature pigs were harvested by surgical operation before eruption. The dental bud tissues were cultured and expanded in vitro for three weeks to obtain dental bud cells (DBCs). The phenotypes of DBCs were identified with a flowcytometry, and the DBCs were seeded into a gelatin–chondroitin–hyaluronan tri-copolymer (GCHT) scaffold. The DBCs/GCHT scaffold constructs were implanted under dermis of nude mice's thoracic dorsum. Mice were sacrificed at predetermined intervals, and the developing tooth-like tissues were harvested for histological examinations. The present results of flowcytometry showed that the DBCs expressed specific surface markers of mesenchymal stem cells. Animal study revealed that the tooth-like structures expressed cytokeratin 14 at 4, 8, and 12 weeks postoperatively. The vascular endothelial growth factor was expressed on 12 weeks. Dentin-like mineralized tissue and dentin genetic-like cells were generated that expressed dentin martrix protein-1 on 16 and 20 weeks. Osteocytes were formed on 24 weeks and expressed osteopontin. This study reveals that the DBCs combined with an appropriate scaffold regenerated tooth-like structure with specific proteins for odontogenesis in nude mice.
Article
The human tooth with immature apex is a developing organ available for investigation. In this tooth, especially in the apical pulp, the proliferation and differentiation of various cells are activated to make a complete tooth. We investigated the notion that unique cells are included in the apical pulp of human tooth with immature apex. Human impacted third molars with immature apex freshly extracted for orthodontic reasons or treatment were obtained. Histological analyses revealed that BrdU-incorporating cells and cells positive for the mesenchymal stem cell markers SH2 and SH3 were located in the same region. The cells from the apical pulp of a human tooth with immature apex, designated here as apical pulp-derived cells (APDCs), can be cultured easily in vitro under ordinary serum-supplemented culture condition. The expression of surface markers of expanded APDCs is similar to that of bone marrow mesenchymal stem cells, except for CD49d (α4-integrin). APDCs differentiated into mineralized cells, adipocytes, chondroblasts and neural cells in vitro. APDCs have a high capacity for proliferation and multilineage potential in vitro. Our results indicate that human tooth with immature apex is a precious tissue source for the research of human adult stem cells and for the advancement of dental and regenerative medicine.
Article
Engineering oral tissues as a multidisciplinary approach to build complex structures such as bone, teeth or soft dental tissues remains a challenging endeavor which will also require significant additional development of materials chemistry before it will be successful. We will highlight areas of recent success and describe major challenges which the materials chemistry community, in collaboration with clinicians, must still overcome. The isolation of stem cell populations from various sources in the oral cavity and advances in utilizing their differentiation potential has been driving the field forward. So far, bioinert materials have mainly been used as carriers and delivery vehicles, relying on the intrinsic cellular competence to form tissues. As this may not suffice to induce regeneration, there is a need for novel biomimetic scaffolds capable of providing chemical and mechanical cues to promote multiple specific interactions between cells and matrix. These signals can orchestrate processes such as cell adhesion, migration, differentiation, matrix synthesis, mineralization, and/or vasculogenesis. In this review, we give a brief description of oral anatomy and pathology, state-of-the-art treatment methods and their shortcomings. We provide an overview of current strategies to fabricate bioactive matrices, with an emphasis on nanostructured materials, and we suggest design principles for scaffolding systems specifically tailored towards dental tissue regeneration. In this review, we envision future approaches based on these emerging areas that rely on recent developments in tissue engineering and stem cell research. At the interface between material science and biology, cellular response can be controlled by materials chemistry, and potential applications for regenerative strategies are evolving.
Article
The objective of this study was to investigate the use of alginate hydrogels to present either exogenous or endogenous transforming growth factor (TGF)- g 1 to the dentin-pulp complex to signal reparative processes. Hydrogels were prepared, applied to cultured human tooth slices and the effects on tertiary dentinogenesis examined histologically. Both TGF- g 1-containing and acid-treated alginate hydrogels, but not untreated hydrogels, upregulated dentin matrix secretion and induced odontoblast-like cell differentiation with subsequent secretion of regular tubular dentin matrix on cut pulpal surfaces. It is concluded that TGF- g 1 can signal both induction of odontoblast-like cell differentiation and upregulation of their matrix secretion in the human dentin-pulp complex. Alginate hydrogels provide an appropriate matrix in which dental regeneration can take place and may also be useful for delivery of growth factors, including TGF- g s, to enhance the natural regenerative capacity of the dental pulp.
Article
Chitosan is a natural polymer with favorable properties such as biocompatibility, biodegradability, non-allergenic and non-toxic which make it a very promising material for scaffold in tissue engineering. It also exerts bioactive properties such as antibacterial and wound-healing. In the first part of this review, a general introduction to tissue engineering and chitosan will be given. Applications of chitosan based systems in hard tissue engineering such as bone, cartilage and periodontal are reviewed with recent and relevant examples, and the factors affecting the formulation properties are discussed. Furthermore, the approaches to enhance the properties of the chitosan-based scaffolds will be mentioned. Applications of chitosan in soft tissue engineering will be reviewed in the second part to be published in this journal.