You are on page 1of 66

PROF.

PETER MARK BARTOLD (Orcid ID : 0000-0002-5695-3877)


Accepted Article
Article type : Supplement Article

Mesenchymal Stem Cells and Biologic Factors leading to Bone Formation

PM Bartold1, S Gronthos2, D Haynes3, S Ivanovski4

1
School of Dentistry, University of Adelaide, Adelaide, South Australia, Australia

2
Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and
Medical Sciences, University of Adelaide,

Adelaide, South Australia, Australia

3
Adelaide Medical School, Faculty of Health and Medical Sciences
University of Adelaide, Adelaide, South Australia, Australia

4
School of Dentistry, University of Queensland, Brisbane, Queensland, Australia

Running Title: Stem cells and biologics in osteogenesis

Key Words: Mesenchymal stem cells, growth factors, bone regeneration

Address for Correspondence

Professor PM Bartold

School of Dentistry
This article has been accepted for publication and undergone full peer review but has
not been through the copyediting, typesetting, pagination and proofreading process,
which may lead to differences between this version and the Version of Record. Please
cite this article as doi: 10.1111/jcpe.13053

This article is protected by copyright. All rights reserved.


University of Adelaide

Adelaide, South Australia


Accepted Article
Australia

Email: mark.bartold@adelaide.edu.au

Statement of Sources of Funding:

This review was not funded by any external funding agency

Disclosure of any conflicts of interests:

All authors declare they have conflicts of interest to declare.

Abstract

Background: Physiological bone formation and bone regeneration occurring during

bone repair can be considered distinct but similar processes. Mesenchymal stem

cells and associated biologic factors are crucial to both bone formation and bone

regeneration.

Aim: To perform a narrative review of the current literature regarding the role of

mesenchymal stem cells and biologic factors in bone formation with the aim of

discussing the clinical relevance of in vitro and in vivo animal studies.

Methods: The literature was searched for studies on mesenchymal stem cells and

biologic factors associated with the formation of bone in the mandible and maxilla.

The search specifically targeted studies on key aspects of how stem cells and

biologic factors are important in bone formation and how this might be relevant to

bone regeneration. The results are summarized in a narrative review format.

This article is protected by copyright. All rights reserved.


Results: Different types of mesenchymal stem cells and many biologic factors are

associated with bone formation in the maxilla and mandible.


Accepted Article
Conclusion: Bone formation and regeneration involve very complex and highly

regulated cellular and molecular processes. By studying these processes new

clinical opportunities will arise for therapeutic bone regenerative treatments.

Clinical Relevance:

Scientific Rationale: This is a review regarding the role of mesenchymal stem cells
and biologic factors in bone formation.

Principle Findings

Different types of mesenchymal stem cells and many biologic factors are associated
with bone formation in the maxilla and mandible.

Practical Implications

Although it is widely recognized that a thorough appreciation of the biological basis of


clinical therapies is essential, it is not until we understand the process of formation
that regeneration will become an achievable clinical endpoint for managing disease
and trauma to tissues. This will certainly be the case for bone regeneration.

This article is protected by copyright. All rights reserved.


INTRODUCTION

This review is presented as part of the discussion papers for the XV


Accepted Article
European Workshop in Periodontology on Bone Regeneration. The topic assigned

for this review was “Mesenchymal Stem Cells and Biologic Factors leading to Bone

Formation”. Clearly this is a difficult area to cover in one review as it involves two

philosophies of thinking and thus the scientific and clinical approaches associated

with stem cells and growth factors for bone formation can differ. With this in made

two specific aspects were reviewed namely: how stem cells and biologic factors are

important in bone formation and how this might be relevant to bone regeneration in

both preclinical and clinical settings. Since this is not a systematic review no

conclusions can be drawn concerning clinical outcomes, rather consideration of the

information to date should be helpful in defining future clinical studies.

BONE BIOLOGY, STRUCTURE AND REGENERATION

Bone is a hard connective tissue constituting the various skeletal elements

with a variety of mechanical and structural properties that are derived from its micro-

and macro-structural hierarchical arrangement (Rho et al., 1998). It has a number of

specific roles including providing mechanical support and protection, maintaining

mineral homeostasis, hematopoiesis and endocrine functions (Florencio-Silva et al.,

2015). The hierarchical arrangement of bone can be viewed at both the nano-scale

where structured cross-linked collagen matrices are embedded within a mineralized

matrix and the macro-scale where collagen fibers are aligned according to the

mechanical and functional requirements of the tissue.

This article is protected by copyright. All rights reserved.


Microstructure and Macrostructure of Bone

Bone can be divided into several different types according to its stage of
Accepted Article
development and microstructure as primary (woven), secondary (lamellar),

cancellous (trabecular) and compact (cortical) bone (Buck and Dumanian, 2012)

(Figure 1). Woven bone is a developmentally early form of bone characterized by

poorly organized collagen type I fibers with a high cellular content and large voids

resulting in low mineral content (Garcia-Rodriguez and Martinez-Reina, 2017). It is

the first form of bone to be seen in repairing and regenerating bone defects and is

eventually resorbed and replaced. Secondary, or lamellar bone, is comprised of four

general forms: osteons, trabecular, circumferential and interstitial bone (Buckwalter

et al., 1995). The osteons form the major structural unit of cortical bone and are

formed by concentric lamellae surrounding the central Haversian canal that contains

blood vessels, nerves and lymphatics. Cell processes emanate from the osteon in

the form of canaliculi and these connect the central canal to osteocytes allowing

diffusion of nutrients through the bone. Cortical, or compact, bone is the highly

mineralized, dense outer surface of bone consisting of closely packed osteons. It has

an important structural and weight-bearing role due to its high resistance to bending

and torsion. Cancellous, or trabecular, bone also contains lamellar bone but in a

different arrangement to the osteon and does not have a Haversian canal system but

rather has sinusoids that allow communication between the bone marrow and

mineralized bone tissue. The trabeculae that provide the cancellous bone with a very

high surface area are more cellular and have a higher vascular content than the

cortical bone. Overlying the external cortical bone surface is the periosteum, which is

a thick collagenous layer, comprised of Sharpey’s fibers. This provides a surface for

attachment of ligaments (including the periodontal ligament) as well as providing a

rich blood supply to the outer bone layers.

This article is protected by copyright. All rights reserved.


Bone formation and regeneration

Developmentally bone is derived from the mesoderm although there is also


Accepted Article
some contribution to the craniofacial skeletal tissues from the neural crest. Bone is

formed via two mechanisms know as endochondral and intramembranous bone

ossification. For the purposes of this review only intramembranous bone formation

will be considered since this is the mechanism through most oral osseous defects

repair and regenerate. Interesting embryonic bone formation and bone repair and

regeneration are almost identical processes (Shapiro, 2008). Intramembranous bone

formation is characterized by the formation of bone matrix without the intermediary

step of chondrogenesis (endochondral ossification). Ossification occurs through the

differentiation of mesenchymal stem cells into osteoprogenitor cells and finally into

osteoblasts. All of this is intricately orchestrated by myriad growth, differentiation and

transcription factors (Berendsen and Olsen, 2015). Once formed all bones undergo

continual remodelling dictated predominantly by mechano-transduction signals

(Robling and Turner, 2009). Thus, bone is continually resorbed and reformed

according to the forces placed upon it. These processes occur for developing bone,

newly formed bone and regenerated bone tissues. While primarily designed to

confer optimal strength and function to the osseous structures remodelling can have

significant ramifications for bone grafts or regenerated bone protected from

mechanical forces through excessive resorption resulting in lost architecture and

function.

ALVEOLAR BONE

The alveolar bone, together with the periodontal ligament and root surface

cementum provide the support structures for teeth within the maxilla and mandible

(Figure 2). During development of the maxilla and mandible the alveolar bone

This article is protected by copyright. All rights reserved.


forms via intramembranous bone formation. Structurally alveolar bone is composed

of two parts. One is the alveolar process of the maxilla and mandible that contains
Accepted Article
developing teeth as well as the roots of fully erupted teeth. The other type of alveolar

bone is termed “alveolar bone proper” and this is the portion of the alveolar bone that

houses the tooth roots of erupted teeth and associated periodontal ligament. The

periodontal ligament attaches into the alveolar bone proper via Sharpey’s fibers and

provides a very strong interface between tooth root and bone. Radiographically

alveolar bone proper can be seen as a thick radiopaque line adjacent to the alveolar

socket, termed the lamina dura. The alveolar bone proper is also commonly referred

to as “bundle bone” reflecting the embedded fiber bundles from the periodontal

ligament. Also included within the alveolar bone proper is lamellar bone (also

referred to as the cribriform plate) that is perforated by small holes, or foramina, that

allow egress of nerves and blood vessels from the bone into the periodontal

ligament. These most likely also provide a pathway for mesenchcymal stem cells to

gain access to the periodontal ligament to assist in repair and regeneration. The

alveolar bone proper encases the whole tooth root surface and at it’s most coronal

part merges with the alveolar bone to form the alveolar crest. Once teeth are

extracted there is no need for the alveolar bone proper and it resorbs very rapidly

(Araujo et al., 2015).

STEM CELLS

Stem cells are defined by their capacity to self renew, undergo extensive

proliferation and the potential to reproducibly differentiate into functional cells

indicative of several different lineages (Dominici et al., 2006). Stem/progenitor cells

have been isolated from nearly every tissue and organ of the human body.

Characterization of these cells has shown that there is considerable variability in their

This article is protected by copyright. All rights reserved.


capacity to differentiate with some having considerable pluripotent capacity while

others exhibit more limited differentiation capacity.


Accepted Article
Stem cells may be categorized on the basis of their source and associated

differentiation capacity. In general there are four main sources of stem cells; (i)

embryonic tissues, (ii) foetal tissues, (iii) postnatal tissues and (iv) reprogrammed

differentiated somatic cells (termed induced pluripotent stem cells) (Figure 3)(Lin et

al., 2008).

Embryonic stem cells have been isolated from mammalian embryos and have

a very high proliferative capacity as well as the ability to differentiate into cells with

the features of all three embryonic germ layers (ectoderm, endoderm and

mesoderm). Thus, these pluripotent cells can differentiate into almost all cell types

comprising the human body. While these cells present considerable potential for use

in regenerative medicine their clinical use has been significantly impeded due to the

ethical and legal issues relating to the use of embryos for the acquisition of these

cells. Nonetheless these cells are considered the gold standard for future

regenerative cell-based therapies (Ilic and Ogilvie, 2017).

Just over a decade ago the first reports appeared describing the

reprogramming of somatic cells into pluripotent cells with a differentiation and self-

renewal capacity comparable to embryonic stem cells (Takahashi and Yamanaka,

2006). These cells were termed “induced pluripotent stem cells” (iPSC) are

considered to provide an ethically viable alternative to embryonic stem cells in

regenerative medicine.

Adult, somatic or postnatal stem cells can be isolated from almost all organs

and tissues of the human body. These cells play a crucial role in the maintenance of

tissue integrity as well as responding to injury and restoring damage tissues through

repair and regenerative processes. However, these stem cells are far more

This article is protected by copyright. All rights reserved.


restricted in their proliferative and differentiation capacity compared to embryonic

stem cells and induced pluripotent stem cells. Notwithstanding this, adult stem cells
Accepted Article
have been studied in considerable detail with regards to their use in regenerative

medicine (Kanji and Das, 2017).

Due to their accessibility, high growth capacity and multi-potential,

mesenchymal stem cells hold great promise for tissue regeneration in clinical

applications. However, there are a number of technical issues that still need to be

overcome, including identification and isolation of appropriate precursor cell types,

establishment of optimal growth and differentiation conditions in vitro and efficient

modes of delivery for the successful transplantation of mesenchymal stem cells

(Gronthos et al., 2000). The essential components for generating effective cellular

based therapeutic strategies include a population of multi-potential progenitor cells,

presence of signalling molecules/inductive morphogenic signals and a conductive

extracellular matrix scaffold or appropriate delivery system (Nakashima, 2005,

Nakashima and Reddi, 2003, Srisuwan et al., 2006). One major obstacle that limits

the clinical use of mesenchymal stem cells is the inability to define the initial cell

preparations. The heterogeneity of the starting cell population that may result in poor

reproducibility of therapeutic outcomes, and hence it is clear that comprehensive

characterization of these different cell populations is required (Wagner et al., 2005).

In this review of stem cells we have included both in vitro and in vivo studies

that shed light on the clinical utility of mesenchymal stem cells for bone regeneration.

It is recognized that in vitro and preclinical animal studies can be of limited value but

in the absence of clinical studies they provide the best evidence to date to allow

these cells to be considered for potential clinical utility. Where possible we have

included human clinical trials to substantiate preclinical studies.

This article is protected by copyright. All rights reserved.


MESENCHYMAL STEM CELLS AND BONE REGENERATION

Postnatal stem cells were first derived and cultured from bone marrow and
Accepted Article
were defined as plastic adherent cells with clonal capacity termed colony-forming unit

fibroblasts (Friedenstein et al., 1976, Friedenstein et al., 1966). The incidence of

CFU-F in bone marrow aspirates varies between individuals ranging from 1 to 10

CFU-F per 105 cells plated. Initially, these cells were termed bone marrow stromal

cells or stromal precursor cells, but in recent times they have been generally referred

to as mesenchymal stem cells (Caplan, 1991). In 2006 the International Society for

Cellular Therapy determined that the use of the term mesenchymal stem cell was

appropriate for all multipotent fibroblast-like plastic adherent cells provided they met

specific stem cell criteria (Dominici et al., 2006, Horwitz et al., 2005). These criteria

were listed as: adherence to plastic, a specific surface antigen expression pattern

and multipotent differentiation potential (Dominici et al., 2006). Later, it was

recognized that these parameters were not sufficient to discriminate these cells from

other multipotential, clonogenic, plastic adherent cells that could be isolated from a

variety of stromal tissues. Accordingly the property of an ability to undergo self-

renewal without the loss of development potential was added to the defining features

of mesenchymal stem cells (Bianco et al., 2008).

It is now recognized that there are many defining features of mesenchymal

stem cells that should be met in order to clarify the status of cells being studied either

in vitro or in vivo. While there is considerable variability in the growth and

differentiation potential of mesenchymal stem cells (Gronthos et al., 2000, Seo et al.,

2004), a common defining feature of these cells is their capacity, under appropriate

culture conditions, to differentiate into at least three distinct phenotypic lineages,

such as osteoblasts, chondrocytes and adipocytes (Dominici et al., 2006). In addition

to these properties, cell surface markers (CD73, CD90, CD105 and CD166) have

been used to try to confirm mesenchymal stem cell identity (Dominici et al., 2006).

This article is protected by copyright. All rights reserved.


However, it is now recognized that these surface proteins alone are non-specific

markers of “stemness”, due to their expression by end-differentiated stromal cells


Accepted Article
and fibroblast populations that do not display multi- differentiation capacity. Other

studies have shown that the cell surface antigen, STRO-1 (Gronthos et al., 2003b), is

highly expressed by human bone marrow CFU-F. The STRO-1 antigen was recently

identified as a cell surface epitope on HSC70, expressed on mesenchymal stem cells

and some cancer lines (Fitter et al., 2017). Importantly, the anti-STRO-1 antibody

reacts with less than 10% of adult human bone marrow mononuclear cells,

containing all assayable CFU-F (Gronthos et al., 2003b). Moreover, STRO-1 positive

cultures lack the presence of hematopoietic stem/ progenitor cell populations and

macrophage/ endothelial cells, which are readily detected in primary stromal cultures

derived from unfractionated bone marrow aspirates (Simmons and Torok-Storb,

1991). Adult bone marrow mononuclear cells sorted on the basis of STRO-1

expression possess the capacity for multi-differentiation into various stromal cell

types including osteoblasts, adipocytes, chondrocytes, smooth muscle cells and

hematopoietic supportive stroma (Gronthos et al., 2003b, Dennis et al., 2002).

Interestingly, selection of ex vivo expanded BMSC based on their expression of

STRO-1, exhibit an enhanced capacity for growth, migration, bone formation,

hematopoietic support, immunomodulation and pro-angiogenic properties, compared

to STRO-1 negative cells (Psaltis et al., 2010, Bensidhoum et al., 2004, Goncalves et

al., 2006, Nasef et al., 2009). These developments have opened up new areas of

investigation for evaluating the properties and clinical potency of purified MSC

populations, without the presence of contaminating accessory cell populations with

the potential to influence the growth, paracrine properties and developmental

potential of cultured mesenchymal stem cells.

This article is protected by copyright. All rights reserved.


Thus, it has been recommended that in order for a cell to be labeled a

mesenchymal stem cell rigorous assessments should be carried out (Bartold and
Accepted Article
Gronthos, 2017). These should include demonstration that the individual clonal

populations have the capacity to produce the connective tissue matrix

microenvironment of their tissue of origin when implanted into immunocompromised

mice as well as demonstration of their self-renewal capacity over serial

transplantations in vivo. More recently further criteria have been proposed to define

mesenchymal stem cells and these include assessment of a number of growth

factors and cytokines that promote stem cell survival, angiogenesis, and immune cell

responses (Konala et al., 2016).

Mesenchymal stem cells are amongst the most highly studied types of adult

stem cells. They include cells derived from nearly all tissues comprising the human

body Depending on their source these cells have the capacity to differentiate into

cells with the characteristics of mesoderm (adipocytes, osteoblasts, chondrocytes,

tenocytes, skeletal myocytes and visceral stromal cells) (Gronthos et al., 2003b,

Horwitz et al., 1999, Jiang et al., 2002, Pereira et al., 1995, Pittenger et al., 1999,

Smith et al., 2004), ectoderm (neurons, astrocytes) (Woodbury et al., 2000) and

endoderm (hepatocytes) (Petersen et al., 1999). Therefore, the term mesenchymal

stem cell is very generic, and additional descriptors should be added to better

describe the tissue specificity and origin of these cells (for example: bone marrow

mesenchymal stem cells, adipose stem cells, dental pulp stem cells, periodontal

ligament stem cells).

Mesenchymal stem cells derived from specific tissues retain some “memory”

of those tissues and thus exhibit some tissue specific properties in addition to more

generic multi-potential and these can be defined by their niche environment

(Menicanin et al., 2014). Furthermore, since the first identification of mesenchymal

stem cells in the periodontal ligament (Seo et al., 2004). mesenchymal stem cell-like

This article is protected by copyright. All rights reserved.


populations have been isolated from all dental tissues except enamel (ameloblasts or

other cellular elements following tooth development) (Chen et al., 2012). Because
Accepted Article
dental tissues do not undergo continuous remodelling as do bone tissues, dental-

tissue-derived mesenchymal stem cells are more restricted in their differentiation

capacity compared to bone marrow-derived mesenchymal stem cells (Huang et al.,

2009).

Given the enormous diversity of tissues that harbour mesenchymal stem cell-

like populations this review will be limited to those cells that have been most

extensively studied for their potential to participate in bone formation and

regeneration (Table 1).

Bone Marrow Stromal Cells (BMSC)

Within the bone marrow there are three major stem cell populations:

haematopoietic stem cells that are responsible for the formation of blood cells;

angioblasts that are endothelial cell precursors cells, and bone marrow stromal stem

cell or skeletal stem cell populations responsible for the formation of the

haematopoietic microenvironment that supports and regulates the haematopoietic

stem cells (Friedenstein et al., 1970, Gronthos et al., 2003a, Pittenger et al., 1999,

Owen and Friedenstein, 1988).

These cells are comprised of a mixed population of stromal progenitor cells at

various stages of development, maintained by a minor population of multipotential,

stromal stem cells with the capacity for self-renewal(Owen and Friedenstein, 1988).

Subsequently they were demonstrated to have osteogenic capacity when implanted

into immunodeficient mice and this was controlled by at least four growth factors

including platelet derived growth factor, transforming growth factor-beta, basic

fibroblast growth factor and epidermal growth factor (Kuznetsov et al., 1997a).

This article is protected by copyright. All rights reserved.


These cells have considerable capacity to proliferate and expand in vitro as

well as the ability to differentiate into multiple cell lineages (Gronthos et al., 2003b,
Accepted Article
Kuznetsov et al., 1997b, Menicanin et al., 2010). Therefore, there has been

considerable interest in these cells with regard to their pivotal role in skeletal growth

turnover and potential use in regenerative medicine (Bianco et al., 2001). Thus in

skeletal physiology these cells are crucial in contributing to bone growth and

turnover. These unique features are also critical to their regenerative capacity. In

addition to these functions, bone marrow stromal stem cells help to maintain

hematopoietic stem cells in their niche and in general help to maintain the

hematopoietic surroundings. These properties run in parallel with their regenerative

capacity to modulate tissue function through the release of a number of soluble

mediators instructive for tissue regeneration (Bianco, 2014). This so-called

“secretome” may hold potent regenerative potential and may serve as an alternative

to whole cell transplantation (Katagiri et al., 2017).

As described above an early defining feature of bone marrow stromal stem

cells was the ability to form colonies of cells derived from a single precursor cell

denoted CFU-F (Friedenstein et al., 1974). These cells have a high capacity to

undergo self-replication and can undergo more that 50 cell doublings in vitro. It has

been estimated that billions of cells can be generated from as little as 1 ml of bone

marrow aspirate (Bianco et al., 2001).

The bone forming capacity of bone marrow stromal stem cells is well

recognized and has been extensively reviewed (Arthur et al., 2009, Bianco et al.,

2006, Bruder et al., 1998a, Caplan, 2005, Caplan, 2007, Derubeis and Cancedda,

2004, Kode et al., 2009). Early studies reported the osteoinductive (osteogenic)

capacity of human bone marrow stromal stem cells when they were combined with

an osteoconductive scaffold of hydroxyapatite / tricalcium phosphate (HA/TCP) and

subsequently implanted subcutaneously into the dorsal tissues of

This article is protected by copyright. All rights reserved.


immunocompromised mice (Kuznetsov et al., 1997b). Once excised, histological

assessment of these transplants demonstrated neovascularization, fibrous


Accepted Article
connective tissue and areas of new bone formation. The osseous regenerative

potential of bone marrow stromal stem cell within hydroxyl apatite/tricalcium

phosphate (HA/TCP) scaffolds has also been demonstrated in dogs, sheep and

goats for the repair of critical-sized bone defects (Bruder et al., 1998b, Brodke et al.,

2006, Kon et al., 2000, Viateau et al., 2007) (Bensaid et al., 2005, Petite et al., 2000,

Liu et al., 2008, Liu et al., 2010). Many other studies have assessed and confirmed

the ability of both allogeneic and autologous bone marrow stromal stem cells to

regenerate bone in different animal models (Arinzeh et al., 2003, Dai et al., 2005,

Muraglia et al., 1998, Yuan et al., 2007, Zhu et al., 2006, Field et al., 2011).

Notwithstanding these favourable outcomes, it should be noted that simple

implantation of bone marrow stromal cells into large bone defects has significant

technical difficulties. For example, early studies found that cartilage, rather than

bone, formed due to lack of vascularization (Lerner et al., 2018). These

observations led to efforts to improve the carrier vehicles and scaffolds used to

deliver bone marrow stromal cells to osseous defects using tissue engineering

concepts. A limiting factor in cell transplantation has been the properties of the tissue

engineering delivery vehicle. The fundamental concept underlying tissue engineering

is to combine a scaffold with living cells, and/or biologically active molecules to form

a “tissue engineering construct” that promotes the regeneration of tissues (Bartold et

al., 2000, Bartold et al., 2006). The scaffold should perform various functions,

including support cell colonisation, migration, proliferation and differentiation. The

scaffold design also should consider physico-chemical properties, morphology and

degradation kinetics. External size and shape of the construct are of importance,

particularly if the construct is customised for an individual patient. Scaffolds must

provide sufficient initial mechanical strength and stiffness to substitute for the loss of

This article is protected by copyright. All rights reserved.


mechanical function of the diseased, damaged or missing tissue. Continuous cell and

tissue remodelling is important for stable biomechanical conditions and


Accepted Article
vascularisation at the host site. In addition to these essentials of mechanics and

geometry, a suitable construct will (i) possess a 3D and highly porous interconnected

pore network with surface properties which are optimised for the attachment,

migration, proliferation and differentiation of cell types of interest and enable flow

transport of nutrients and metabolic waste, and (ii) be biocompatible and

biodegradable with a controllable rate to complement cell/tissue growth and

maturation (Bartold et al., 2016, Hutmacher and Cool, 2007). In the early days of

tissue engineering, it was believed that scaffolds should degrade and vanish as the

tissue is growing. Yet, tissue in-growth and maturation differs temporally from tissue

to tissue and, furthermore, tissue in-growth does not equate to tissue maturation and

remodelling, in other words a defect filled with immature tissue should not be

considered “regenerated”. Hence, many scaffold-based strategies have failed in the

past as the scaffold degradation was more rapid than tissue remodelling and/or

maturation. It is now recognized that the onset of degradation should only occur after

the regenerated tissue within the scaffold has remodelled at least once in the natural

remodelling cycle. Work over the last ten years underlines the importance of the

scaffold remaining intact as the tissue matures in the scaffold pores with bulk

degradation occurring later (Vaquette et al., 2018).

A large number of case reports have demonstrated a favourable outcome for

bone regeneration following implantation of bone marrow stromal stem cells

(Killington et al., 2018). Despite these findings, there are surprisingly few

randomised clinical trials or systematic clinical studies (i.e. not just case reports or

case series) using bone marrow stromal stem cells for bone regeneration in humans.

This finding is further highlighted in the review for this Symposium on cell therapies

for orofacial bone regeneration where it is concluded that most studies to date have

This article is protected by copyright. All rights reserved.


used whole bone marrow rather than ex vivo expanded cells (Shanbhag et al., 2018).

A recent systematic review (Killington et al., 2018) reported that 10 clinical trials were
Accepted Article
registered in the clinical trials.gov registry and of these 4 have been completed and

two were published and available for assessment (Dallari et al., 2007, Liebergall et

al., 2013). The two studies evaluated in this systematic review produced equivocal

results with one reporting improved bone formation after implantation of bone marrow

stromal stem cells (Dallari et al., 2007) and one report failing to find any such effect

(Liebergall et al., 2013). A recent study investigating the use of bone marrow stem

cells for the regeneration of alveolar cleft and trauma in adults found that while the

use of these cells is safe, their ability to facilitate complete regeneration of large

alveolar defects is limited (Bajestan et al., 2017). Most recently, significant

regeneration (in both horizontal and vertical dimensions) for severe mandibular

resorption has been reported when bone marrow stromal stem cells were used in

conjunction with biphasic calcium phosphate scaffolds and titanium-reinforced

membranes (Gjerde et al., 2018). Similar results have been reported when using

similar procedures for treating orthopedic non-unions (Gomez-Barrena et al., 2018).

All human clinical studies to date have demonstrated that bone regeneration requires

mesenchymal stem cells to have a suitable environment and this is usually facilitated

by the use of appropriate scaffold as well as biologic factors such as bone

morphogenetic proteins, transforming factor-beta and insulin-like growth factor.

These factors will be discussed later in this review.

In the context of oral reconstruction, bone marrow stromal stem cells have

been used in a number of clinical trials. In the most recent systematic review

concerning the use of mesenchymal stem cells in oral reconstructive surgery 18

studies were included of which 10 involved the use of bone marrow stromal stem

cells mixed with a variety of carriers and biologic factors (Jakobsen et al., 2013).

While these studies generally showed that bone marrow stromal stem cells may be

This article is protected by copyright. All rights reserved.


associated with new bone formation this was not a consistent finding and the

contribution of the scaffolds and biologic agents was not clear due to wide study
Accepted Article
design diversity.

Dental-derived MSC-like cells

As detailed above, mesenchymal stem cells were first isolated from bone

marrow but have since been found in nearly every tissue and organ of the human

body. Stems cells of dental origin were first isolated from the dental pulp (Gronthos

et al., 2000) and subsequently from periodontal ligament (Seo et al., 2004). Since

these early studies, mesenchymal stem cells have been isolated from every dental

tissue except enamel (Chen et al., 2012). This reflects the reparative potential of

dental tissues since following eruption of teeth into the mouth all of the periodontal

and dental structures, with the exception of enamel, have some form of regenerative

capacity presumably mediated by local mesenchymal stem cell-like populations

(Duailibi et al., 2006). Dental mesenchymal stem cells have been well characterized

and demonstrate features originally ascribed to bone marrow stromal stem cells such

as plastic adherence, colony forming unit capacity and multipotent differentiation

capacity (Gronthos et al., 2000, Miura et al., 2003, Seo et al., 2004). At least six

different mesenchymal stem cells have been isolated from dental tissues: dental pulp

stem cell, periodontal ligament stem cells, stem cells from human exfoliated

deciduous teeth, stem cells from apical papilla, dental follicle stem cells and gingival

mesenchymal stem cells.

This article is protected by copyright. All rights reserved.


Dental pulp stem cells (DPSC)

The dental pulp is not dissimilar to bone marrow in that it is a highly


Accepted Article
vascularized soft connective tissue fully encased by mineralized tissue. Thus the

initial discovery of mesenchymal stem cell populations residing within dental pulp

was not surprising, with these cells exhibiting characteristics comparable to those of

bone marrow stromal stem cells such as capacity to form mineralized tissues and

similar cell surface markers associated with endothelium, smooth muscle,

bone/dentine, and fibroblasts (Gronthos et al., 2000). Although the overall

differentiation capacity of dental pulp stem cells is lower than bone marrow stromal

stem cells, they exhibit higher proliferative rates and capacity to form colony forming

units.

The bone forming capacity of dental pulp stem cells has been demonstrated

in animal models and, as for bone marrow stromal stem cells, an appropriate scaffold

and supplementary growth factors seem to be prerequisites for these cells to

participate in bone regeneration in vivo (Wongsupa et al., 2017, Tsukamoto et al.,

2017, Chamieh et al., 2016, Asutay et al., 2015). Notwithstanding the large number

of animal preclinical studies, there are few human in vivo studies addressing the

bone regenerative capacity of dental pulp stem cells. These cells have been

investigated for their potential to assist in tooth extraction socket healing whereby

sites that had been implanted with dental pulp stem cells demonstrated regenerated

compact bone that had a matrix density greater than the controls after three years

(d'Aquino et al., 2009, Giuliani et al., 2013, Monti et al., 2017). While these results

indicate a potential role for dental pulp stem cells in bone regeneration there is need

for more studies to determine the clinical utility of dental pulp stem cells for human

bone regeneration.

This article is protected by copyright. All rights reserved.


Periodontal ligament stem cells (PDLSC)

Before their ultimate identification, the presence of precursor or stem cells


Accepted Article
had long been suspected to be present in the periodontal ligament (McCulloch et al.,

1987). Periodontal ligament stem cells were first isolated from human third molars

and were demonstrated to have mesenchymal stem cell properties similar to those

described for bone marrow stromal stem cells (Seo et al., 2004). Importantly these

cells were shown to have considerable regenerative capacity when implanted into

animals where they generated cementum/periodontal ligament structures (Seo et al.,

2004). Subsequently, these cells were demonstrated to have considerable

osteogenic and bone regenerative properties both in vitro and in vivo (Gay et al.,

2007, Grimm et al., 2011, Seo et al., 2004). As for other mesenchymal stem-like

cells, it is apparent that biocompatible scaffolds are required to maintain the viability

and osteogenic capacity of periodontal ligament stem cells and facilitate bone

formation in vivo (Kim et al., 2009, He et al., 2011, Yu et al., 2014, Tour et al., 2012).

While the periodontal regenerative capacity of periodontal ligament stem cells has

been well demonstrated in animal models and more recently in several small human

clinical case studies (Yamada et al., 2006, Feng et al., 2010, McAllister, 2011, Chen

et al., 2016), studies regarding their role in bone regeneration are limited and

restricted to animal models (Akazawa et al., 2016, Kammerer et al., 2017, Yu et al.,

2014, Kim et al., 2009, Ge et al., 2012).

Stem cells from human exfoliated deciduous teeth (SHED)

Stem cells from human exfoliated deciduous teeth are derived from remnants

of the pulp in the crown of exfoliated human deciduous teeth and represent a very

unique stem cell population (Miura et al., 2003). Interestingly, although derived from

dental pulp, stem cells from human exfoliated deciduous teeth differ from their adult-

derived counterpart dental pulp stem cells with regards to their morphology, colony

This article is protected by copyright. All rights reserved.


formation unit capacity, in vivo osteoconductive capacity, and in contrast to dental

pulp stem cells they do not form a dentin-pulp-like complex (Miura et al., 2003).
Accepted Article
Compared to dental pulp stem cells and bone marrow stromal stem cells, stem cells

from human exfoliated deciduous teeth have been demonstrated to have higher

proliferative capacity and higher expression of genes related to both cell proliferation

and extracellular matrix synthesis (Nakamura et al., 2009). Porcine and canine stem

cells from exfoliated deciduous teeth have been demonstrated to regenerate bone

when implanted into critical sized defects in the mandible and cranium (Yamada et

al., 2011, Zheng et al., 2009, Seo et al., 2008, de Mendonca Costa et al., 2008). No

human studies have been carried out to date.

Stem cells from apical papilla (SCAP)

The apical papilla is a developmental organ associated with tooth root

formation and also contributes to formation of the dental pulp. The cells found at this

site are termed stem cells from apical papilla and are a unique population of

multipotent stem cells that express high levels of survivin and telomerase that are

involved in regulating cell proliferation (Sonoyama et al., 2006). Due to their high

proliferative potential stem cells from apical papilla are considered to have better

regenerative capacity than their dental pulp counterparts, the dental pulp stem cells

(Sonoyama et al., 2006, Sonoyama et al., 2008). In vitro these cells can form

odontoblast-like cells and adipocytes (Sonoyama et al., 2006, Abe et al., 2007). In

addition these cells can express a number of neurologic cell markers (Sonoyama et

al., 2006, Sonoyama et al., 2008, Abe et al., 2007). Stem cells from apical papilla

for transplantation can be obtained from extracted third molars. It has been reported

that sufficient numbers of stem cells from apical papilla can be obtained from one

tooth for transplantation (Sonoyama et al., 2006).

This article is protected by copyright. All rights reserved.


Stem cells from apical papilla have shown osteogenic differentiation potential

in vitro (Park et al., 2009). A population of stem cells from apical papilla with high
Accepted Article
expression of CD-24 is associated with a very high osteogenic differentiation

potential (Sonoyama et al., 2006, Aguilar and Lertchirakarn, 2016). Exposure of stem

cells from apical papilla to insulin-like growth factor has been reported to enhance

osteogenic differentiation (Wang et al., 2012). However, the timing of exposure to

growth factors may be important since basic fibroblast growth factor has been noted

to both increase and decrease osteogenic induction depending on the phase of the

cell cycle at the time of exposure (Wu et al., 2012). Stem cells from apical papilla

have been shown to have regenerative potential for the dentin/pulp complex and bio-

root engineering (Sonoyama et al., 2006, Sonoyama et al., 2008). While animal

studies have demonstrated the osgteogenic potential of implanted stem cells from

apical papilla (Abe et al., 2008) to date, no studies have evaluated the potential for

stem cells from apical papilla to regenerate bone in human defects.

Dental follicle stem cells (DFC)

The dental follicle envelops the developing tooth germ prior to tooth eruption.

Because of this site’s strategic role in tooth development, it is considered to be a

good source of stem cells (Morsczeck et al., 2005). Dental follicle stem cells can be

readily obtained from unerrupted or impacted third molars. In vitro these cells

demonstrate limited osteogenic capacity after induction in osteogenic media

(Morsczeck et al., 2005) and after repeated cell passages they appear to lose their

osteogenic capacity (Yao et al., 2013).

Initial animal studies using dental follicle stem cells showed no evidence of

bone formation (Morsczeck et al., 2005). Later studies have reported bone formation

in vivo when dental follicle stem cells were implanted subcutaneously or into critical

This article is protected by copyright. All rights reserved.


size osseous defects (Tsuchiya et al., 2010, Park et al., 2012, Rezai-Rad et al.,

2015). There are no studies to date investigating the use of dental follicle stem cells
Accepted Article
in human osseous defects.

Gingival Mesenchymal stem cells (GMSC)

Cells from the gingival tissues were first noted to possess significant

heterogeneity and multipotency over 25 years ago (McCulloch and Bordin, 1991).

However, it was not until quite recently that mesenchymal stem cells from gingival

tissues have been characterized (Fournier et al., 2010, Fawzy El-Sayed and Dorfer,

2016). When first described human gingival mesenchymal stem cells were found to

have high proliferative rates, a stable morphology and retained stable karyotype and

telomerase activity over sustained passage in culture (Tomar et al., 2010). These

cells have been repeatedly demonstrated to have osteogenic potential in vitro

(Fournier et al., 2010, Tomar et al., Mitrano et al., 2010, El-Sayed et al., 2015).

While early in vivo studies failed to demonstrate osteogenic capacity of gingival

mesenchymal stem cells when implanted subcutaneously (Zhang et al., 2009),

subsequent studies have shown that these cells can promote bone formation in vivo

when implanted into periodontal defects and calvarial critical size defects (Wang et

al., 2011, Yu et al., 2013). While these cells have been proposed to be a good

source of MSC-like cells for bone regeneration due to their easy availability and

robust in vitro characteristics, no human studies have been carried out to date.

This article is protected by copyright. All rights reserved.


Adipose Tissue Stem Cells (ASC)

Mesenchymal stem cells derived from adipose tissue are emerging as a very
Accepted Article
good source of adult stem cells for regenerative therapies due to their easily

accessible nature and properties comparable to bone marrow stromal stem cells

(Bacakova et al., 2018). Mesenchymal stem cells derived by liposuction from

adipose tissue were first described in 2001 (Zuk et al., 2001). These cells were

demonstrated to have multipotent capacity to differentiate into adipocytes,

osteoblasts chondrocytes and myocytes and can form bone when implanted

subcutaneously into mice (Zuk et al., 2001, Dragoo et al., 2003). More recent studies

have demonstrated that adipose stem cells may be superior to bone marrow stromal

stem cells because they are more easily harvested, undergo later in vitro

senescence, have a higher proliferative rate and have a very high autocrine

production of growth factors and immunomodulatory factors essential for tissue

regeneration (Hsiao et al., 2012, Ding et al., 2013, Varghese et al., 2017).

The ostgeogenic capacity of adipose stem cells has been studied in

considerable detail and has led to the suggestion that these cells are amongst the

most promising for use in cell-based bone regeneration (Lindroos et al., 2011). The

osteogenic effect may arise due to their ability to differentiate into osteoblasts as well

as through paracrine mechanisms facilitating the migration and differentiation of other

precursor osteoblasts (de Girolamo et al., 2007, Tajima et al., 2015).

Adipose tissue stem cells have been found to have considerable osteogenic

bone regenerative capacity in vivo (Parrilla et al., 2011, Pourebrahim et al., 2013).

Enhanced osseous repair has been reported following implantation of these cells into

critical size osseous defects and segmental long bone defects in mice, rats and

rabbits (Levi et al., 2010, Pieri et al., 2010, Kim et al., 2012). Similar findings have

been reported in larger experimental animals such as mandibular bone defects in

This article is protected by copyright. All rights reserved.


pigs (Wilson et al., 2012). As for most mesenchymal stem cells, implantation

protocols for adipose tissue stem cells demonstrate improved efficacy when
Accepted Article
implanted within space maintaining scaffolds (Arrigoni et al., 2013) Further

improvements in the bone regenerative capacity of adipose tissue stem cells have

been reported when combined with growth factors such as bone morphogenetic

protein-2 or platelet rich plasma (Chou et al., 2011, Tajima et al., 2015).

Following successful preclinical studies adipose tissue stem cells have been

used in clinical settings for bone regeneration. A number of single case reports and

case series have demonstrated the potential of adipose tissue stem cells for osseous

regeneration in humans (Lendeckel et al., 2004, Mesimaki et al., 2009, Sandor et al.,

2013, Sandor, 2012, Thesleff et al., 2011). Adipose tissue stem cells have shown

particular promise for craniomaxillofacial osseous regeneration (Paduano et al.,

2017). Since the first case report on the use of adipose tissue stem cells for

augmentation of a post traumatic calvarial defect in a 7-year old female (Lendeckel et

al., 2004), there have been a number of case reports demonstrating the clinical utility

of these cells for craniofacial osseous reconstruction (Paduano et al., 2017). There

is now need for larger controlled clinical trials to confirm that these cells could be a

suitable alternative to bone marrow stromal stem cells.

BIOLOGIC FACTORS IN BONE FORMATION AND REGENERATION

Bone formation repair and regeneration

Osteoinduction, the biologic process of bone formation requires not only

recruitment and expansion of bone forming cells but also stimulation of non-infectious

inflammation and increased vascularization. Therefore, therapeutic adjuncts for bone

healing and regeneration require a sound understanding of the phases of bone

healing which are very complex involving a number of hierarchical and temporal

This article is protected by copyright. All rights reserved.


phases (Hankenson et al., 2014, Araujo et al., 2015). Upon bone injury (e.g. tooth

extraction) there is an immediate vascular response leading to clot formation and


Accepted Article
homeostasis. Next there is inflammatory cell migration into the site followed by a

fibrovascular response with recruitment of mesenchymal stem cells to the site

together with neovascularization. The recruited mesenchymal stem cells can then

differentiate into bone forming cells and facilitate neoossification. Finally the new

bone is remodeled following recruitment of osteoclasts and replacement of woven

bone is replaced by newly formed lamellar bone (Araujo et al., 2015). Each of these

phases is guided and controlled by the release of biologic factors in the form of

growth factors and cytokines with growth factors in particular receiving considerable

attention. A comprehensive list of growth, differentiation and transcription factors

known to be involved in bone formation and regeneration are listed in Table 2. More

specific details of the cellular aspects of bone remodeling and how this is regulated

through specific growth factors is reviewed in this volume in a paper considering the

critical interplay between bone resorbing and bone forming cells (Lerner et al., 2018).

A schematic representation of the cellular and molecular players in bone formation,

resorption and remodelling is shown in Figure 4.

It is important to recognize that much of the knowledge we have regarding

regulation of osteogenic differentiation is derived from animal models and in vitro

studies. Nonetheless, these provide pivotal information for both basic understanding

as well as studies of clinical relevance using fundamental knowledge to translate to

clinical practice through the development of novel therapeutics (Bartold et al., 2010,

Cantley et al., 2017).

This article is protected by copyright. All rights reserved.


Role of Growth Factors

Growth factors are a collective group of highly biologically active molecules


Accepted Article
that can stimulate cell proliferation, migration and differentiation by either autocrine or

paracrine mechanisms (Zarei and Soleimaninejad, 2018). While the effects of growth

factors are well recognized the mechanisms of their actions are still under intense

investigation. Growth factors are integral components regulating embryonic tissue

formation, maintaining tissue homeostasis and the co-ordinating tissue healing and

regeneration. The use of growth factors for bone regeneration has been under

intense investigation for several decades and in recent times this has resulted in

many of these being approved for human clinical use or undergoing evaluation in

human clinical trials. A search of clinicaltrials.gov (May 10, 2018) using the search

words “bone and growth factor” identified 391 registered clinical trials. The list of

known biologically active growth factors is voluminous with around 33 families of

structurally and evolutionary related proteins identified to date (Bafico and Aaronson,

2003). An appraisal of each and every known growth factor is beyond the scope of

this review. Accordingly this review will consider only the most widely studied growth

factors in the context of bone biology and regeneration. These include the bone

morphogenetic proteins (BMPs), vascular endothelial cell growth factor (VEGF),

fibroblast growth factors (FGF), platelet derived growth factors (PDGF). Although

blood derived products, such as platelet rich plasma (PRP) and platelet rich fibrin

(PRF), are a mixture of a number of growth and differentiation factors, they are

considered as a biologic agent of considerable interest in the context of bone

regeneration due to their ready availability as a chair-side source of biologic factors.

This article is protected by copyright. All rights reserved.


Inflammatory Phase of Bone Formation

Inflammation is essential for any wound healing and bone is no exception.


Accepted Article
Immediately following bone injury a blood clot forms and this serves to achieve

hemostasis. During this phase of bone healing inflammatory cells are recruited to the

site of injury through the release of a number of pro-inflammatory cytokines released

by platelets. The inflammatory infiltrate is extensive and comprises neutrophils,

lymphocytes, macrophages and eosinophils and if compromised, healing is

significantly affected (Bastian et al., 2011). The initial role of these cells is to “clean”

the wound site prior to formation of new tissue. Additional roles for these cells are to

initiate neovascularization and also to promote recruitment of fibroblasts to the site.

At this time there is a significant increase in a number of additional pro-inflammatory

mediators. These include prostaglandin-E2, bone morphogenetic protein-2,

monocyte chemoattractant proteins (MCP-1 and MCP-3), tumor necrosis factor- ,

transforming growth factor-β and numerous interleukins (IL-1, IL-3, IL-6,IL-10).

These factors have been shown to be crucial to bone repair in gene knock-out and

receptor blocking experiments that have demonstrated diminished granulation tissue

formation post clot formation at the healing site (Walters et al., 2018). The highly

iterative wound healing process leads to the formation of granulation tissue that is

slowly replaced with a more mature connective tissue rich in collagen and fibroblasts.

Cell Recruitment for Bone Formation

Mesenchymal stem cells with the capacity to differentiate into bone forming cells as

well as endothelial progenitor cells need to be attracted to the healing site. The

source of these cells for osseous healing in the maxilla, mandible and healing tooth

sockets is unclear but the periosteum, bone marrow cavity and endosteum are all

likely to be able to provide an ample supply of mesenchymal stem cells and

This article is protected by copyright. All rights reserved.


endothelial progenitor cells for bone formation. A crucial phase in this recruitment of

precursor cells is the release of initiating molecular signals by platelets such as


Accepted Article
platelet-derived growth factor (PDGF) in both its AA and BB isoforms. PDGF

promotes cell migration and proliferation. It acts through receptor tyrosine kinases

which in turn activate a number of intracellular pathways including mitogen-activated

protein kinase (MAPK), phosphoinositide-3 kinase (PI3K), Stat3 and Rho-Rac.

Through these mechanisms PDGF induces mesenchymal stem cell migration and

proliferation as well as, importantly, angiogenesis (Graham et al., 2009). While

PDGF AA has some osteogenic capacity (Li et al., 2014) it is PDGF BB that has

received most attention as a potential therapeutic agent for bone formation and

regeneration (Graham et al., 2009, Gronthos and Simmons, 1995). A number of

reports have demonstrated the clinical efficacy of PDGF for bone regeneration

(Younger et al., 2016, Li et al., 2017). Specifically, PDGF has been investigated for

its osseous regenerative properties in the management of repair and regeneration of

orthopedic and periodontal osseous defects (Hollinger et al., 2008, Friedlaender et

al., 2013, Nevins et al., 2013). The positive results from these studies have have led

to U.S. Food and Drug Administration (FDA) approval of rhPDGF for these

indications.

The fibroblast growth factors are another important group of biological

mediators involved in the recruitment and proliferation of mesenchymal stem cells

and endothelial progenitor cells (Charoenlarp et al., 2017). This family is composed

of 22 related proteins that have the capacity to play a role in bone formation. Within

this family FGF2, FGF9 and FGF 18 have been proposed to have the greatest

potential for use in bone regeneration (Charoenlarp et al., 2017) and of these FGF2

has received the most attention. Osteogenic differentiation of mesenchymal stem

cells derived from FGF2 knockout mice is reduced but this can be restored by the

addition of exogenous FGF2 (Montero et al., 2000). Furthermore, FGF knockout

This article is protected by copyright. All rights reserved.


animals demonstrate a number of developmental osseous abnormalities as well as

adverse bone healing and reduced bone formation (Du et al., 2012). Human trials
Accepted Article
have commenced using FGF to assist repair of bone fractures (Kawaguchi et al.,

2010, Tanaka et al., 2012) and also for promoting regeneration of periodontal

intraosseous defects (Kitamura et al., 2011, Li et al., 2017, Kitamura et al., 2016,

Kitamura et al., 2008) with promising results.

Angiogenesis and Bone Formation

Angiogenesis is a critical part of wound healing and tissue regeneration

(Stegen et al., 2015). For bone formation, angiogenesis is also a critical process to

prevent ischemia at the healing site and permit adequate supply of nutrients and cells

necessary for healing. Indeed ischemia is a significant cause of impaired bone

healing. The newly formed vessels also provide additional signaling molecules

involved in cell differentiation and ossification.

Until complete revascularization of the reparative tissue occurs, the wound is

hypoxic and any cells that are present must be able to adapt to these conditions

through oxygen sensing. In general, this is achieved through hypoxia inducible factor

(HIF) which leads to the release of vascular endothelial cell growth factor (VEGF)

and stimulation of neovascularization (Fan et al., 2014). VEGF is one of the most

widely studied growth factors in bone healing and regeneration because of its critical

role in neovascularization (Hu and Olsen, 2017). VEGF is highly expressed by

endothelial progenitor cells at sites of bone healing and this is thought to be an

important mechanism stimulating angiogenesis and facilitate the commencement of

bone repair (Hu and Olsen, 2017). VEFG has been shown to potentiate the bone

forming capacity of bone morphogenetic proteins (Cui et al., 2013) but these

synergistic effects have not been observed consistently (Li et al., 2016).

This article is protected by copyright. All rights reserved.


Angiopoietin is another important growth factor involved in angiogenesis.

While not directly osteogenic, it can, like VEGF, it can also act synergistically with
Accepted Article
bone morphogenetic protein 2 to upregulate the expression of osteogenic genes in

mesenchymal stem cells (Jeong et al., 2010). Furthermore, animal studies show that

implantation of angiopoietin and BMP2 into bone defects has been shown to induce

regeneration (Choi et al., 2015)

Cell differentiation and Bone Formation

Initial bone formation at sites of bone injury in the craniofacial region is

performed by osteoblasts that have differentiated from the mesenchymal stem cell

infiltrate in a manner consistent with that of intramembranous ossification. The

mesenchymal stem cell differentiation process is controlled not only by soluble

mediators, but also by the cells’ responses to hypoxia and mechanical stresses.

While mechanical forces are of interest to bone healing around teeth and dental

implants this is beyond the scope of this review and only the most significant soluble

biological mediators will be considered here.

Bone Morphogenetic Proteins, TGF- and bone cell differentiation

The discovery that demineralized bone matrix, when implanted into animals

(rabbits, rats, mice and guinea pigs) as well as humans, could induce ectopic bone

formation and new bone formation in osseous defects was a significant advance in

understanding the role of the extracellular matrix in osseoinduction (Urist, 1965).

Since then the active components have been identified as a family of proteins

collectively referred to as bone morphogenetic proteins that are part of the TGF

supergene family (Wozney et al., 1988). To date, 14 BMPs have been identified and

This article is protected by copyright. All rights reserved.


of these BMP-2,4,5,6,7 and 9 demonstrate osteogenic capacity. The osteogenic

activity of BMP-2 and BMP-7 has been the most widely studied both in experimental
Accepted Article
animals and in human trials for use in the repair of osseous defects and deformities

(Wu et al., 2016).

TGF was originally isolated from human platelets (Assoian et al., 1983) and,

like the BMPs belongs to the TGF supergene family. It is more commonly

associated with the differentiation of chondrocytes and endochondral bone formation

rather than intramembranous bone formation. While there is considerable evidence

to suggest that TGF inhibits osteoblast differentiation and intramembranous bone

formation, it may it may also have both stimulatory and inhibitory effects on

osteoblast formation mediated via complex intracellular signaling pathways. (Tang et

al., 2009).

Although the BMPs and TGF bind to related cell surface receptors and

activate similar signaling pathways (SMADS) their endpoint of action on bone

formation is different. The BMPs act through SMAD1, 5 and 8 which are associated

with osteogenic pathways whereas TGFβ acts through SMAD2 and 3 and drives

chondrocyte differentiation (Wu et al., 2016). The BMPs and TGFβ also act through

SMAD-independent pathways (e.g. p38 mitogen-activated protein kinase/p38 MAPK)

to influence mesenchymal stem cell differentiation. Both the SMAD and p38 MAPK

signaling pathways can activate specific transcription factors (e.g. Runx2) leading to

the differentiation of mesenchymal stem cells into chondrocytes and osteoblasts.

However, this is probably a highly simplistic view of the roles of TGFβ, BMPs and

SMADS, as their various interactions with many transcription factors are very

complex and yet to be fully determined (Nickel et al., 2018).

This article is protected by copyright. All rights reserved.


Cell Surface Signaling Pathways for bone cell differentiation

The Wnt signaling pathway is another important process associated with


Accepted Article
osteogenesis and bone regeneration (Leucht et al., 2018). Wnt exerts its action via

cell surface receptors (LRP 4,5 and 6) and β-catenin resulting in gene regulation

necessary for bone formation (Duan and Bonewald, 2016). The expression of Wnt

receptors is upregulated during bone regeneration (Wise et al., 2010). Disruption of

the canonical signalling pathway of Wnt has been demonstrated to influence bone

formation. For example, sclerostin is a soluble inhibitor of Wnt that, if deficient or

blocked, can lead to increased bone mass (Pietrzyk et al., 2017). Conversely,

knockout mice deficient in the Wnt cell surface receptors LRP 5 and LRP 6

demonstrate diminished bone mass (Holmen et al., 2004). Our expanding

understanding of the role of Wnt signalling in bone development, formation and

regeneration has led to translational studies investigating how this pathway can be

manipulated for regenerative purposes (Brommage, 2015, Leucht et al., 2018).

Notch signalling is another pathway that has received considerable attention

for its role in osteoinduction (Majidinia et al., 2018). The Notch signalling pathway is

mediated through four transmembrane Notch receptors (NOTCH1,2,3 & 4). Notch is

a crucial pathway involved in mesenchymal stem cell differentiation and subsequent

bone healing (Wang et al., 2016). It exerts its influence via direct osteoinductive

effects on osteoblasts. Inhibition of Notch in progenitor cells leads to reduced

osteoblastogenesis with resultant bone loss (Tu et al., 2012). Interestingly, activation

of Notch (as well as TGF and Wnt) can lead to enhanced osteoblast differentiation

at modified titanium surfaces (Chakravorty et al., 2014).

This article is protected by copyright. All rights reserved.


Remodeling of newly formed bone

The final stage in bone formation and regeneration is the remodeling phase
Accepted Article
that is mediated by both osteoclasts and osteoblasts to replace the early formed

woven bone with more mature lamellar bone. This is a complex process that can take

months or even years to complete. Bone remodeling in healing tooth sockets has

been extensively studied and provides a very useful background to understanding

how modeling and remodeling take place to result in both qualitative and quantitative

changes in bone healing and repairing bone (Araujo et al., 2015). The most critical

regulators of this process are osteoprotegerin, the signaling pathway of Receptor

Activator of Nuclear Factor B and its ligand (RANK/RANKL) and parathyroid

hormone.

RANKL is a transmembrane ligand expressed on osteoblasts, and other

stromal stromal cells, that binds to RANK, a transmembrane receptor found on

osteoclast precursor cells (Lacey et al., 1998). Interaction between RANKL and

RANK propels a cascade of signalling and gene expression that culminates in the

differentiation and maturation of osteoclast precursor cells into active osteoclasts with

bone resorbing capabilities. Osteoprotegerin is a naturally occurring inhibitor of

osteoclastogenesis (Simonet et al., 1997). It acts as a decoy receptor by binding to

RANKL and blocking its interaction with RANK, thereby preventing osteoclast

development. In addition, many of the hormones and cytokines involved in the

regulation of calcium levels in blood and bone, including prostaglandin E2,

parathyroid hormone, vitamin D3 and interleukin-11, play key roles in stimulating

osteoclastogenesis. These effects are mediated via a combination of inhibiting OPG

production and stimulating RANKL production (Martin and Sims, 2015).

This article is protected by copyright. All rights reserved.


RANK and RANKL knockout mice demonstrate a severe lack of

ostgeoclastogenesis and associated osteopetrosis, defective B & T lymphocytes and


Accepted Article
a number of skeletal defects (Ono and Nakashima, 2018). Therapeutics targeting the

RANK/RANKL axis have been developed to assist in the management of

osteoporosis, metastases to bone and giant cell tumor of bone (Deeks, 2018, Sohn

et al., 2014, Diedhiou et al., 2015, Jamshidi et al., 2018) but to date no significant

trials have been undertaken to determine if these have any effect on bone

remodeling associated with bone repair and regeneration (Vannucci and Brandi,

2016).

Parathyroid hormone is a very important mediator of bone mass which can

lead to either bone resorption or bone formation (Jilka, 2007). Parathyroid hormone

helps to maintain calcium ion levels in the blood and its effects are mediated through

a calcium-sensing receptor in the parathyroid gland. When serum calcium levels are

elevated the receptor mediates a decrease in release of parathyroid hormone.

Depending on the length of time of exposure to parathyroid hormone, its effects on

bone can be either catabolic or anabolic. For example, chronic exposure to

parathyroid hormone results in bone remodeling, whereas intermittent exposure to

the hormone results in bone apposition (Jilka, 2007). Intermittent exposure to

parathyroid hormone leads to bone apposition through three cellular mechanisms

including osteoblast proliferation, inhibition of osteoblast apoptosis and activation of

bone cells to produce new bone matrix (Lindsay et al., 2007). Induction of osteoblast

differentiation (and thereby bone apposition) by parathyroid hormone is assisted by a

number of osteogenic cytokines and growth factors, including insulin-like growth

factor-1 and fibroblast growth factor (Lindsay et al., 2007). Parathyroid hormone

stimulation of osteoclast differentiation is principally mediated through the

osteoprotegerin (OPG) and RANK/RANKL axis (Greenfield, 2012). Overall, the

anabolic and catabolic effects of parathyroid hormone on bone through its cell

This article is protected by copyright. All rights reserved.


surface receptor are mediated by complex regulatory mechanisms controlled by

growth factors, transcription factors, regulatory molecules and signaling pathways


Accepted Article
(Silva and Bilezikian, 2015).

A modified form of parathyroid hormone consisting of its first 34 amino acids

(teriparatide) has been developed and trialled for the management of osteoporosis

(Neer et al., 2001). This medication has been found to increase bone mineral

formation and density in extraction sockets and prevent periodontal bone loss in rats

with induced osteoporosis. Furthermore in a canine model teraparatide assisted new

bone formation around dental implants (Valderrama et al., 2010). Early human trials

have demonstrated the potential for teraparatide to positively influence osseous

regeneration in the oral cavity (Bashutski et al., 2010).

CELLS, BIOLOGICS AND REGENERATION

The cellular and molecular processes of bone formation during

embryogenesis, remodelling and regeneration are considered to be almost identical

(Bruder et al., 1994). It is evident that a principal underlying requirement for these

processes is the presence or supply of mesenchymal stem cells. The preferred

therapeutic strategy moving forward is the use of ex vivo expanded MSC

preparations rather than whole bone marrow aspirate transplantation or autografts

that contain limited MSC numbers diluted within a heterogeneous population of

blood/ immune cells. The move away from autograft-based therapy would also

alleviate the risk of morbidity at secondary bone sites. Originally, for the biological

process of bone formation, it was thought that mesenchymal stem cells arising from

bone were the only cells that could be involved. However, with time it has become

evident that mesenchymal stem cells from a variety of tissues have the potential to

participate in bone formation and regeneration. These include mesenchymal stem

This article is protected by copyright. All rights reserved.


cell-like populations from diverse tissues such as the tooth forming elements and

adipose tissue. While the simple injection of cells into osseous defects has been
Accepted Article
studied it is apparent that a more efficient method for the utilization of mesenchymal

stem cells for osseous regeneration is to combine them with tissue growth factors

and space maintaining and delivery systems. With these concepts in mind the field

of tissue engineering emerged and is defined as the science of reconstructing or

mimicking natural processes through the use of synthetic polymer scaffolds with the

expectation of tissue regeneration. The vision is that suitable stem cells produced in

large enough quantities through cell culture methods and aided by appropriate

growth and differentiation factors, are injected into tissues and organs in appropriate

carrier vehicles. As such, they provide a source of progenitor cells that replace the

lost or damaged resident cells allowing for regeneration of damaged tissues. Thus

stem cell based regenerative technologies rely on the input from a large number of

scientific fields including nanotechnology, three-dimension bioprinting,

bioengineering, cell biology, molecular biology, gene manipulation and good

manufacturing processes (Li et al., 2018, Oryan et al., 2017).

A number of principles have been proposed that must be taken into account

for the use of mesenchymal stem cells for bone formation, repair or regeneration

(Bruder et al., 1994):

1. An adequate supply of mesenchymal stem cells must be available

2. Mesenchymal stem cells require specific molecular cues in order to differentiate

into bone forming cells

3. Without rapid vascularization of the MSC-implanted site, the graft will fail

4. Turnover and remodelling are essential phases leading to mature and fully

regenerated bone

This article is protected by copyright. All rights reserved.


5. Delivery scaffolds and devices must be designed within the context of

biocompatibility and appropriate resorption/replacement rates, consistent with


Accepted Article
new woven bone formation and subsequent remodelling into lamellar bone.

Thus, understanding the interface between biology and engineering is central to

achieving acceptable regenerative outcomes (Li et al., 2018). Accordingly, new bone

formation and regeneration can only be achieved by combining our understanding of

cell biology, growth and differentiation factors and bioengineering principles and not

pursuing these processes in isolation.

CONCLUSION

Bone formation and regeneration involve very complex and highly regulated

cellular and molecular processes. By studying these processes new clinical

opportunities will arise for therapeutic bone regenerative treatments. These

processes can be studied both in vitro and in vivo with in vivo studies including

preclinical animal models and human clinical trials. Where possible we have

discussed human clinical trial outcomes but where this was not possible in vitro and

preclinical animal studies have been presented to highlight the potential of stem cells

and biologic agents to play significant roles in both bone formation and bone

regeneration. This review has highlighted the potential roles of stem cells and

specific growth and biologic agents in bone formation and therefore, as a corollary,

bone regeneration. Future research efforts will need to target both stem cells and

biologics through well-controlled clinical trials based on the in vitro and preclinical

published to date. Not only is there considerable opportunity for stem cell based

therapies but also by combining these with regulatory molecules through the

controlled temporal delivery using tissue engineering approaches, offer many exciting

prospects for bone regeneration. Since it is widely recognized that a thorough

This article is protected by copyright. All rights reserved.


appreciation of the biological basis of clinical therapies is essential, it is not until we

understand the process of formation that regeneration will become an achievable


Accepted Article
clinical endpoint for managing disease and trauma to tissues. This will certainly be

the case for bone regeneration.

References

Abe, S., Yamaguchi, S. & Amagasa, T. (2007) Multilineage Cells From Apical Pulp of
Human Tooth with Immature Apex. Oral Science International, 45-58.

Abe, S., Yamaguchi, S., Watanabe, A., Hamada, K. & Amagasa, T. (2008) Hard tissue
regeneration capacity of apical pulp derived cells (APDCs) from human tooth
with immature apex. Biochem Biophys Res Commun 371, 90-93.
doi:10.1016/j.bbrc.2008.04.016.

Aguilar, P. & Lertchirakarn, V. (2016) Comparison of stem cell behaviors between


indigenous high and low-CD24 percentage expressing cells of stem cells from
apical papilla (SCAPs). Tissue & Cell 48, 397-406. doi:10.1016/j.tice.2016.08.008.

Akazawa, K., Iwasaki, K., Nagata, M., Yokoyama, N., Ayame, H., Yamaki, K., Tanaka, Y.,
Honda, I., Morioka, C., Kimura, T., Komaki, M., Kishida, A., Izumi, Y. & Morita, I.
(2016) Double-layered cell transfer technology for bone regeneration. Sci Rep 6,
33286. doi:10.1038/srep33286.

Araujo, M. G., Silva, C. O., Misawa, M. & Sukekava, F. (2015) Alveolar socket healing: what
can we learn? Periodontol 2000 68, 122-134. doi:10.1111/prd.12082.

Arinzeh, T. L., Peter, S. J., Archambault, M. P., van den Bos, C., Gordon, S., Kraus, K., Smith,
A. & Kadiyala, S. (2003) Allogeneic mesenchymal stem cells regenerate bone in a
critical-sized canine segmental defect. J Bone Joint Surg Am 85-A, 1927-1935.

Arrigoni, E., de Girolamo, L., Di Giancamillo, A., Stanco, D., Dellavia, C., Carnelli, D.,
Campagnol, M., Domeneghini, C. & Brini, A. T. (2013) Adipose-derived stem cells
and rabbit bone regeneration: histomorphometric, immunohistochemical and
mechanical characterization. J Orthop Sci 18, 331-339. doi:10.1007/s00776-012-
0349-y.

Arthur, A., Zannettino, A. & Gronthos, S. (2009) The therapeutic applications of


multipotential mesenchymal/stromal stem cells in skeletal tissue repair. J Cell
Physiol 218, 237-245. doi:10.1002/jcp.21592.

This article is protected by copyright. All rights reserved.


Assoian, R. K., Komoriya, A., Meyers, C. A., Miller, D. M. & Sporn, M. B. (1983)
Transforming growth factor-beta in human platelets. Identification of a major
storage site, purification, and characterization. J Biol Chem 258, 7155-7160.
Accepted Article
Asutay, F., Polat, S., Gul, M., Subasi, C., Kahraman, S. A. & Karaoz, E. (2015) The effects of
dental pulp stem cells on bone regeneration in rat calvarial defect model: micro-
computed tomography and histomorphometric analysis. Arch Oral Biol 60, 1729-
1735. doi:10.1016/j.archoralbio.2015.09.002.

Bacakova, L., Zarubova, J., Travnickova, M., Musilkova, J., Pajorova, J., Slepicka, P.,
Kasalkova, N. S., Svorcik, V., Kolska, Z., Motarjemi, H. & Molitor, M. (2018) Stem
cells: their source, potency and use in regenerative therapies with focus on
adipose-derived stem cells - a review. Biotechnol Adv.
doi:10.1016/j.biotechadv.2018.03.011.

Bafico, A. & Aaronson, S. (2003) Classification of Growth Factors and Their Receptors. .
In: Kufe DW, Pollock RE, Weichselbaum RR, et al., editors. Holland-Frei Cancer
Medicine. 6th edition. Hamilton (ON): BC Decker; 2003.

Bajestan, M. N., Rajan, A., Edwards, S. P., Aronovich, S., Cevidanes, L. H. S., Polymeri, A.,
Travan, S. & Kaigler, D. (2017) Stem cell therapy for reconstruction of alveolar
cleft and trauma defects in adults: A randomized controlled, clinical trial. Clin
Implant Dent Relat Res 19, 793-801. doi:10.1111/cid.12506.

Bartold, P. M., Cantley, M. D. & Haynes, D. R. (2010) Mechanisms and control of


pathologic bone loss in periodontitis. Periodontol 2000 53, 55-69.
doi:10.1111/j.1600-0757.2010.00347.x.

Bartold, P. M. & Gronthos, S. (2017) Standardization of Criteria Defining Periodontal


Ligament Stem Cells. J Dent Res 96, 487-490. doi:10.1177/0022034517697653.

Bartold, P. M., Gronthos, S., Ivanovski, S., Fisher, A. & Hutmacher, D. W. (2016) Tissue
engineered periodontal products. J Periodontal Res 51, 1-15.
doi:10.1111/jre.12275.

Bartold, P. M., McCulloch, C. A., Narayanan, A. S. & Pitaru, S. (2000) Tissue engineering: a
new paradigm for periodontal regeneration based on molecular and cell biology.
Periodontol 2000 24, 253-269.

Bartold, P. M., Xiao, Y., Lyngstaadas, S. P., Paine, M. L. & Snead, M. L. (2006) Principles
and applications of cell delivery systems for periodontal regeneration.
Periodontol 2000 41, 123-135. doi:10.1111/j.1600-0757.2006.00156.x.

Bashutski, J. D., Eber, R. M., Kinney, J. S., Benavides, E., Maitra, S., Braun, T. M., Giannobile,
W. V. & McCauley, L. K. (2010) Teriparatide and osseous regeneration in the oral
cavity. N Engl J Med 363, 2396-2405. doi:10.1056/NEJMoa1005361.

Bastian, O., Pillay, J., Alblas, J., Leenen, L., Koenderman, L. & Blokhuis, T. (2011) Systemic
inflammation and fracture healing. J Leukoc Biol 89, 669-673.
doi:10.1189/jlb.0810446.

This article is protected by copyright. All rights reserved.


Bensaid, W., Oudina, K., Viateau, V., Potier, E., Bousson, V., Blanchat, C., Sedel, L.,
Guillemin, G. & Petite, H. (2005) De novo reconstruction of functional bone by
tissue engineering in the metatarsal sheep model. Tissue Eng 11, 814-824.
doi:10.1089/ten.2005.11.814.
Accepted Article
Bensidhoum, M., Chapel, A., Francois, S., Demarquay, C., Mazurier, C., Fouillard, L.,
Bouchet, S., Bertho, J. M., Gourmelon, P., Aigueperse, J., Charbord, P., Gorin, N. C.,
Thierry, D. & Lopez, M. (2004) Homing of in vitro expanded Stro-1- or Stro-1+
human mesenchymal stem cells into the NOD/SCID mouse and their role in
supporting human CD34 cell engraftment. Blood 103, 3313-3319.
doi:10.1182/blood-2003-04-1121.

Berendsen, A. D. & Olsen, B. R. (2015) Bone development. Bone 80, 14-18.


doi:10.1016/j.bone.2015.04.035.

Bianco, P. (2014) "Mesenchymal" stem cells. Annu Rev Cell Dev Biol 30, 677-704.
doi:10.1146/annurev-cellbio-100913-013132.

Bianco, P., Kuznetsov, S. A., Riminucci, M. & Gehron Robey, P. (2006) Postnatal skeletal
stem cells. Methods Enzymol 419, 117-148. doi:S0076-6879(06)19006-0 [pii]

10.1016/S0076-6879(06)19006-0.

Bianco, P., Riminucci, M., Gronthos, S. & Robey, P. G. (2001) Bone marrow stromal stem
cells: nature, biology, and potential applications. Stem Cells 19, 180-192.
doi:10.1634/stemcells.19-3-180.

Bianco, P., Robey, P. G. & Simmons, P. J. (2008) Mesenchymal stem cells: revisiting
history, concepts, and assays. Cell Stem Cell 2, 313-319.
doi:10.1016/j.stem.2008.03.002.

Brodke, D., Pedrozo, H. A., Kapur, T. A., Attawia, M., Kraus, K. H., Holy, C. E., Kadiyala, S. &
Bruder, S. P. (2006) Bone grafts prepared with selective cell retention
technology heal canine segmental defects as effectively as autograft. J Orthop Res
24, 857-866. doi:10.1002/jor.20094.

Brommage, R. (2015) Genetic Approaches To Identifying Novel Osteoporosis Drug


Targets. J Cell Biochem 116, 2139-2145. doi:10.1002/jcb.25179.

Bruder, S. P., Fink, D. J. & Caplan, A. I. (1994) Mesenchymal stem cells in bone
development, bone repair, and skeletal regeneration therapy. J Cell Biochem 56,
283-294. doi:10.1002/jcb.240560303.

Bruder, S. P., Jaiswal, N., Ricalton, N. S., Mosca, J. D., Kraus, K. H. & Kadiyala, S. (1998a)
Mesenchymal stem cells in osteobiology and applied bone regeneration. Clin
Orthop Relat Res, S247-256.

Bruder, S. P., Kraus, K. H., Goldberg, V. M. & Kadiyala, S. (1998b) The effect of implants
loaded with autologous mesenchymal stem cells on the healing of canine
segmental bone defects. J Bone Joint Surg Am 80, 985-996.

This article is protected by copyright. All rights reserved.


Buck, D. W., 2nd & Dumanian, G. A. (2012) Bone biology and physiology: Part I. The
fundamentals. Plast Reconstr Surg 129, 1314-1320.
doi:10.1097/PRS.0b013e31824eca94.
Accepted Article
Buckwalter, J. A., Glimcher, M. J., Cooper, R. R. & Recker, R. (1995) BONE BIOLOGY .1.
STRUCTURE, BLOOD-SUPPLY, CELLS, MATRIX, AND MINERALIZATION. Journal
of Bone and Joint Surgery-American Volume 77A, 1256-1275.
doi:10.2106/00004623-199508000-00019.

Cantley, M. D., Zannettino, A. C. W., Bartold, P. M., Fairlie, D. P. & Haynes, D. R. (2017)
Histone deacetylases (HDAC) in physiological and pathological bone
remodelling. Bone 95, 162-174. doi:10.1016/j.bone.2016.11.028.

Caplan, A. I. (1991) Mesenchymal stem cells. J Orthop Res 9, 641-650.


doi:10.1002/jor.1100090504.

Caplan, A. I. (2005) Review: mesenchymal stem cells: cell-based reconstructive therapy


in orthopedics. Tissue Eng 11, 1198-1211. doi:10.1089/ten.2005.11.1198.

Caplan, A. I. (2007) Adult mesenchymal stem cells for tissue engineering versus
regenerative medicine. J Cell Physiol 213, 341-347. doi:10.1002/jcp.21200.

Chakravorty, N., Hamlet, S., Jaiprakash, A., Crawford, R., Oloyede, A., Alfarsi, M., Xiao, Y. &
Ivanovski, S. (2014) Pro-osteogenic topographical cues promote early activation
of osteoprogenitor differentiation via enhanced TGFbeta, Wnt, and Notch
signaling. Clin Oral Implants Res 25, 475-486. doi:10.1111/clr.12178.

Chamieh, F., Collignon, A. M., Coyac, B. R., Lesieur, J., Ribes, S., Sadoine, J., Llorens, A.,
Nicoletti, A., Letourneur, D., Colombier, M. L., Nazhat, S. N., Bouchard, P.,
Chaussain, C. & Rochefort, G. Y. (2016) Accelerated craniofacial bone
regeneration through dense collagen gel scaffolds seeded with dental pulp stem
cells. Sci Rep 6, 38814. doi:10.1038/srep38814.

Charoenlarp, P., Rajendran, A. K. & Iseki, S. (2017) Role of fibroblast growth factors in
bone regeneration. Inflamm Regen 37, 10. doi:10.1186/s41232-017-0043-8.

Chen, F. M., Gao, L. N., Tian, B. M., Zhang, X. Y., Zhang, Y. J., Dong, G. Y., Lu, H., Chu, Q., Xu,
J., Yu, Y., Wu, R. X., Yin, Y., Shi, S. & Jin, Y. (2016) Treatment of periodontal
intrabony defects using autologous periodontal ligament stem cells: a
randomized clinical trial. Stem Cell Res Ther 7, 33. doi:10.1186/s13287-016-
0288-1.

Chen, F. M., Sun, H. H., Lu, H. & Yu, Q. (2012) Stem cell-delivery therapeutics for
periodontal tissue regeneration. Biomaterials 33, 6320-6344.
doi:10.1016/j.biomaterials.2012.05.048.

Choi, H., Jeong, B. C., Hur, S. W., Kim, J. W., Lee, K. B. & Koh, J. T. (2015) The Angiopoietin-
1 Variant COMP-Ang1 Enhances BMP2-Induced Bone Regeneration with
Recruiting Pericytes in Critical Sized Calvarial Defects. PLoS One 10, e0140502.
doi:10.1371/journal.pone.0140502.

This article is protected by copyright. All rights reserved.


Chou, Y. F., Zuk, P. A., Chang, T. L., Benhaim, P. & Wu, B. M. (2011) Adipose-derived stem
cells and BMP2: part 1. BMP2-treated adipose-derived stem cells do not improve
repair of segmental femoral defects. Connect Tissue Res 52, 109-118.
doi:10.3109/03008207.2010.484514.
Accepted Article
Cui, Q., Dighe, A. S. & Irvine, J. N., Jr. (2013) Combined angiogenic and osteogenic factor
delivery for bone regenerative engineering. Curr Pharm Des 19, 3374-3383.

d'Aquino, R., De Rosa, A., Lanza, V., Tirino, V., Laino, L., Graziano, A., Desiderio, V., Laino,
G. & Papaccio, G. (2009) Human mandible bone defect repair by the grafting of
dental pulp stem/progenitor cells and collagen sponge biocomplexes. Eur Cell
Mater 18, 75-83.

Dai, K. R., Xu, X. L., Tang, T. T., Zhu, Z. A., Yu, C. F., Lou, J. R. & Zhang, X. L. (2005)
Repairing of goat tibial bone defects with BMP-2 gene-modified tissue-
engineered bone. Calcif Tissue Int 77, 55-61. doi:10.1007/s00223-004-0095-z.

Dallari, D., Savarino, L., Stagni, C., Cenni, E., Cenacchi, A., Fornasari, P. M., Albisinni, U.,
Rimondi, E., Baldini, N. & Giunti, A. (2007) Enhanced tibial osteotomy healing
with use of bone grafts supplemented with platelet gel or platelet gel and bone
marrow stromal cells. J Bone Joint Surg Am 89, 2413-2420.
doi:10.2106/jbjs.f.01026.

de Girolamo, L., Sartori, M. F., Albisetti, W. & Brini, A. T. (2007) Osteogenic


differentiation of human adipose-derived stem cells: comparison of two different
inductive media. J Tissue Eng Regen Med 1, 154-157. doi:10.1002/term.12.

de Mendonca Costa, A., Bueno, D. F., Martins, M. T., Kerkis, I., Kerkis, A., Fanganiello, R. D.,
Cerruti, H., Alonso, N. & Passos-Bueno, M. R. (2008) Reconstruction of large
cranial defects in nonimmunosuppressed experimental design with human
dental pulp stem cells. J Craniofac Surg 19, 204-210.
doi:10.1097/scs.0b013e31815c8a54.

Deeks, E. D. (2018) Denosumab: A Review in Postmenopausal Osteoporosis. Drugs &


Aging 35, 163-173. doi:10.1007/s40266-018-0525-7.

Dennis, J. E., Carbillet, J. P., Caplan, A. I. & Charbord, P. (2002) The STRO-1+ marrow cell
population is multipotential. Cells Tissues Organs 170, 73-82.
doi:10.1159/000046182.

Derubeis, A. R. & Cancedda, R. (2004) Bone marrow stromal cells (BMSCs) in bone
engineering: limitations and recent advances. Ann Biomed Eng 32, 160-165.

Diedhiou, D., Cuny, T., Sarr, A., Norou Diop, S., Klein, M. & Weryha, G. (2015) Efficacy and
safety of denosumab for the treatment of osteoporosis: A systematic review. Ann
Endocrinol (Paris) 76, 650-657. doi:10.1016/j.ando.2015.10.009.

Ding, D. C., Chou, H. L., Hung, W. T., Liu, H. W. & Chu, T. Y. (2013) Human adipose-derived
stem cells cultured in keratinocyte serum free medium: Donor's age does not

This article is protected by copyright. All rights reserved.


affect the proliferation and differentiation capacities. J Biomed Sci 20, 59.
doi:10.1186/1423-0127-20-59.

Dominici, M., Le Blanc, K., Mueller, I., Slaper-Cortenbach, I., Marini, F., Krause, D., Deans,
Accepted Article
R., Keating, A., Prockop, D. & Horwitz, E. (2006) Minimal criteria for defining
multipotent mesenchymal stromal cells. The International Society for Cellular
Therapy position statement. Cytotherapy 8, 315-317.
doi:10.1080/14653240600855905.

Dragoo, J. L., Samimi, B., Zhu, M., Hame, S. L., Thomas, B. J., Lieberman, J. R., Hedrick, M. H.
& Benhaim, P. (2003) Tissue-engineered cartilage and bone using stem cells
from human infrapatellar fat pads. J Bone Joint Surg Br 85, 740-747.

Du, X., Xie, Y., Xian, C. J. & Chen, L. (2012) Role of FGFs/FGFRs in skeletal development
and bone regeneration. J Cell Physiol 227, 3731-3743. doi:10.1002/jcp.24083.

Duailibi, S. E., Duailibi, M. T., Vacanti, J. P. & Yelick, P. C. (2006) Prospects for tooth
regeneration. Periodontol 2000 41, 177-187. doi:PRD165 [pii]

10.1111/j.1600-0757.2006.00165.x.

Duan, P. & Bonewald, L. F. (2016) The role of the wnt/beta-catenin signaling pathway in
formation and maintenance of bone and teeth. Int J Biochem Cell Biol 77, 23-29.
doi:10.1016/j.biocel.2016.05.015.

El-Sayed, K. M., Paris, S., Graetz, C., Kassem, N., Mekhemar, M., Ungefroren, H., Fandrich,
F. & Dorfer, C. (2015) Isolation and characterisation of human gingival margin-
derived STRO-1/MACS(+) and MACS(-) cell populations. Int J Oral Sci 7, 80-88.
doi:10.1038/ijos.2014.41.

Fan, L., Li, J., Yu, Z., Dang, X. & Wang, K. (2014) The hypoxia-inducible factor pathway,
prolyl hydroxylase domain protein inhibitors, and their roles in bone repair and
regeneration. Biomed Res Int 2014, 239356. doi:10.1155/2014/239356.

Fawzy El-Sayed, K. M. & Dorfer, C. E. (2016) Gingival Mesenchymal Stem/Progenitor


Cells: A Unique Tissue Engineering Gem. Stem Cells Int 2016, 7154327.
doi:10.1155/2016/7154327.

Feng, F., Akiyama, K., Liu, Y., Yamaza, T., Wang, T. M., Chen, J. H., Wang, B. B., Huang, G. T.,
Wang, S. & Shi, S. (2010) Utility of PDL progenitors for in vivo tissue
regeneration: a report of 3 cases. Oral Dis 16, 20-28.

Field, J. R., McGee, M., Stanley, R., Ruthenbeck, G., Papadimitrakis, T., Zannettino, A.,
Gronthos, S. & Itescu, S. (2011) The efficacy of allogeneic mesenchymal
precursor cells for the repair of an ovine tibial segmental defect. Vet Comp
Orthop Traumatol 24, 113-121. doi:10-03-0046 [pii]

10.3415/VCOT-10-03-0046.

This article is protected by copyright. All rights reserved.


Fitter, S., Gronthos, S., Ooi, S. S. & Zannettino, A. C. (2017) The Mesenchymal Precursor
Cell Marker Antibody STRO-1 Binds to Cell Surface Heat Shock Cognate 70. Stem
Cells 35, 940-951. doi:10.1002/stem.2560.
Accepted Article
Florencio-Silva, R., Sasso, G. R., Sasso-Cerri, E., Simoes, M. J. & Cerri, P. S. (2015) Biology
of Bone Tissue: Structure, Function, and Factors That Influence Bone Cells.
Biomed Res Int 2015, 421746. doi:10.1155/2015/421746.

Fournier, B. P., Ferre, F. C., Couty, L., Lataillade, J. J., Gourven, M., Naveau, A., Coulomb, B.,
Lafont, A. & Gogly, B. (2010) Multipotent progenitor cells in gingival connective
tissue. Tissue Eng Part A 16, 2891-2899. doi:10.1089/ten.TEA.2009.0796.

Friedenstein, A. J., Chailakhjan, R. K. & Lalykina, K. S. (1970) The development of


fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen
cells. Cell Tissue Kinet 3, 393-403.

Friedenstein, A. J., Deriglasova, U. F., Kulagina, N. N., Panasuk, A. F., Rudakowa, S. F.,
Luria, E. A. & Ruadkow, I. A. (1974) Precursors for fibroblasts in different
populations of hematopoietic cells as detected by the in vitro colony assay
method. Exp Hematol 2, 83-92.

Friedenstein, A. J., Gorskaja, J. F. & Kulagina, N. N. (1976) Fibroblast precursors in


normal and irradiated mouse hematopoietic organs. Exp Hematol 4, 267-274.

Friedenstein, A. J., Piatetzky, S., II & Petrakova, K. V. (1966) Osteogenesis in transplants


of bone marrow cells. J Embryol Exp Morphol 16, 381-390.

Friedlaender, G. E., Lin, S., Solchaga, L. A., Snel, L. B. & Lynch, S. E. (2013) The role of
recombinant human platelet-derived growth factor-BB (rhPDGF-BB) in
orthopaedic bone repair and regeneration. Curr Pharm Des 19, 3384-3390.

Garcia-Rodriguez, J. & Martinez-Reina, J. (2017) Elastic properties of woven bone: effect


of mineral content and collagen fibrils orientation. Biomech Model Mechanobiol
16, 159-172. doi:10.1007/s10237-016-0808-z.

Gay, I. C., Chen, S. & MacDougall, M. (2007) Isolation and characterization of multipotent
human periodontal ligament stem cells. Orthod Craniofac Res 10, 149-160.
doi:10.1111/j.1601-6343.2007.00399.x.

Ge, S., Zhao, N., Wang, L., Yu, M., Liu, H., Song, A., Huang, J., Wang, G. & Yang, P. (2012)
Bone repair by periodontal ligament stem cellseeded nanohydroxyapatite-
chitosan scaffold. Int J Nanomedicine 7, 5405-5414. doi:10.2147/ijn.s36714.

Giuliani, A., Manescu, A., Langer, M., Rustichelli, F., Desiderio, V., Paino, F., De Rosa, A.,
Laino, L., d'Aquino, R., Tirino, V. & Papaccio, G. (2013) Three years after
transplants in human mandibles, histological and in-line holotomography
revealed that stem cells regenerated a compact rather than a spongy bone:
biological and clinical implications. Stem Cells Transl Med 2, 316-324.
doi:10.5966/sctm.2012-0136.

This article is protected by copyright. All rights reserved.


Gjerde, C., Mustafa, K., Hellem, S., Rojewski, M., Gjengedal, H., Yassin, M. A., Feng, X.,
Skaale, S., Berge, T., Rosen, A., Shi, X. Q., Ahmed, A. B., Gjertsen, B. T.,
Schrezenmeier, H. & Layrolle, P. (2018) Cell therapy induced regeneration of
severely atrophied mandibular bone in a clinical trial. Stem Cell Res Ther 9, 213.
Accepted Article
doi:10.1186/s13287-018-0951-9.

Gomez-Barrena, E., Rosset, P., Gebhard, F., Hernigou, P., Baldini, N., Rouard, H., Sensebe,
L., Gonzalo-Daganzo, R. M., Giordano, R., Padilla-Eguiluz, N., Garcia-Rey, E.,
Cordero-Ampuero, J., Rubio-Suarez, J. C., Stanovici, J., Ehrnthaller, C., Huber-Lang,
M., Flouzat-Lachaniette, C. H., Chevallier, N., Donati, D. M., Ciapetti, G., Fleury, S.,
Fernandez, M. N., Cabrera, J. R., Avendano-Sola, C., Montemurro, T., Panaitescu,
C., Veronesi, E., Rojewski, M. T., Lotfi, R., Dominici, M., Schrezenmeier, H. &
Layrolle, P. (2018) Feasibility and safety of treating non-unions in tibia, femur
and humerus with autologous, expanded, bone marrow-derived mesenchymal
stromal cells associated with biphasic calcium phosphate biomaterials in a
multicentric, non-comparative trial. Biomaterials.
doi:10.1016/j.biomaterials.2018.03.033.

Goncalves, R., Lobato da Silva, C., Cabral, J. M., Zanjani, E. D. & Almeida-Porada, G. (2006)
A Stro-1(+) human universal stromal feeder layer to expand/maintain human
bone marrow hematopoietic stem/progenitor cells in a serum-free culture
system. Exp Hematol 34, 1353-1359. doi:10.1016/j.exphem.2006.05.024.

Graham, S., Leonidou, A., Lester, M., Heliotis, M., Mantalaris, A. & Tsiridis, E. (2009)
Investigating the role of PDGF as a potential drug therapy in bone formation and
fracture healing. Expert Opin Investig Drugs 18, 1633-1654.
doi:10.1517/13543780903241607.

Greenfield, E. M. (2012) Anabolic effects of intermittent PTH on osteoblasts. Curr Mol


Pharmacol 5, 127-134.

Grimm, W. D., Dannan, A., Becher, S., Gassmann, G., Arnold, W., Varga, G. & Dittmar, T.
(2011) The ability of human periodontium-derived stem cells to regenerate
periodontal tissues: a preliminary in vivo investigation. Int J Periodontics
Restorative Dent 31, e94-e101.

Gronthos, S., Chen, S., Wang, C. Y., Robey, P. G. & Shi, S. (2003a) Telomerase accelerates
osteogenesis of bone marrow stromal stem cells by upregulation of CBFA1,
osterix, and osteocalcin. J Bone Miner Res 18, 716-722.
doi:10.1359/jbmr.2003.18.4.716.

Gronthos, S., Mankani, M., Brahim, J., Robey, P. G. & Shi, S. (2000) Postnatal human dental
pulp stem cells (DPSCs) in vitro and in vivo. Proc Natl Acad Sci U S A 97, 13625-
13630. doi:10.1073/pnas.240309797.

Gronthos, S. & Simmons, P. J. (1995) The growth factor requirements of STRO-1-positive


human bone marrow stromal precursors under serum-deprived conditions in
vitro. Blood 85, 929-940.

This article is protected by copyright. All rights reserved.


Gronthos, S., Zannettino, A. C., Hay, S. J., Shi, S., Graves, S. E., Kortesidis, A. & Simmons, P.
J. (2003b) Molecular and cellular characterisation of highly purified stromal
stem cells derived from human bone marrow. J Cell Sci 116, 1827-1835.
Accepted Article
Hankenson, K. D., Zimmerman, G. & Marcucio, R. (2014) Biological perspectives of
delayed fracture healing. Injury 45 Suppl 2, S8-S15.
doi:10.1016/j.injury.2014.04.003.

He, H., Yu, J., Cao, J., E, L., Wang, D., Zhang, H. & Liu, H. (2011) Biocompatibility and
Osteogenic Capacity of Periodontal Ligament Stem Cells on nHAC/PLA and
HA/TCP Scaffolds. J Biomater Sci Polym Ed 22, 179-194.
doi:10.1163/092050609x12587018007767.

Hollinger, J. O., Hart, C. E., Hirsch, S. N., Lynch, S. & Friedlaender, G. E. (2008)
Recombinant human platelet-derived growth factor: biology and clinical
applications. J Bone Joint Surg Am 90 Suppl 1, 48-54. doi:10.2106/jbjs.g.01231.

Holmen, S. L., Giambernardi, T. A., Zylstra, C. R., Buckner-Berghuis, B. D., Resau, J. H.,
Hess, J. F., Glatt, V., Bouxsein, M. L., Ai, M., Warman, M. L. & Williams, B. O. (2004)
Decreased BMD and limb deformities in mice carrying mutations in both Lrp5
and Lrp6. J Bone Miner Res 19, 2033-2040. doi:10.1359/jbmr.040907.

Horwitz, E. M., Le Blanc, K., Dominici, M., Mueller, I., Slaper-Cortenbach, I., Marini, F. C.,
Deans, R. J., Krause, D. S. & Keating, A. (2005) Clarification of the nomenclature
for MSC: The International Society for Cellular Therapy position statement.
Cytotherapy 7, 393-395. doi:10.1080/14653240500319234.

Horwitz, E. M., Prockop, D. J., Fitzpatrick, L. A., Koo, W. W., Gordon, P. L., Neel, M.,
Sussman, M., Orchard, P., Marx, J. C., Pyeritz, R. E. & Brenner, M. K. (1999)
Transplantability and therapeutic effects of bone marrow-derived mesenchymal
cells in children with osteogenesis imperfecta. Nat Med 5, 309-313.
doi:10.1038/6529.

Hsiao, S. T., Asgari, A., Lokmic, Z., Sinclair, R., Dusting, G. J., Lim, S. Y. & Dilley, R. J. (2012)
Comparative analysis of paracrine factor expression in human adult
mesenchymal stem cells derived from bone marrow, adipose, and dermal tissue.
Stem Cells Dev 21, 2189-2203. doi:10.1089/scd.2011.0674.

Hu, K. & Olsen, B. R. (2017) Vascular endothelial growth factor control mechanisms in
skeletal growth and repair. Dev Dyn 246, 227-234. doi:10.1002/dvdy.24463.

Huang, G. T., Gronthos, S. & Shi, S. (2009) Mesenchymal stem cells derived from dental
tissues vs. those from other sources: their biology and role in regenerative
medicine. J Dent Res 88, 792-806. doi:10.1177/0022034509340867.

Hutmacher, D. W. & Cool, S. (2007) Concepts of scaffold-based tissue engineering--the


rationale to use solid free-form fabrication techniques. J Cell Mol Med 11, 654-
669. doi:10.1111/j.1582-4934.2007.00078.x.

This article is protected by copyright. All rights reserved.


Ilic, D. & Ogilvie, C. (2017) Concise Review: Human Embryonic Stem Cells-What Have We
Done? What Are We Doing? Where Are We Going? Stem Cells 35, 17-25.
doi:10.1002/stem.2450.
Accepted Article
Jakobsen, C., Sorensen, J. A., Kassem, M. & Thygesen, T. H. (2013) Mesenchymal stem
cells in oral reconstructive surgery: a systematic review of the literature. J Oral
Rehabil 40, 693-706. doi:10.1111/joor.12079.

Jamshidi, K., Gharehdaghi, M., Hajialiloo, S. S., Mirkazemi, M., Ghaffarzadehgan, K. &
Izanloo, A. (2018) Denosumab in Patients with Giant Cell Tumor and Its
Recurrence: A Systematic Review. Arch Bone Jt Surg 6, 260-268.

Jeong, B. C., Kim, H. J., Bae, I. H., Lee, K. N., Lee, K. Y., Oh, W. M., Kim, S. H., Kang, I. C., Lee,
S. E., Koh, G. Y., Kim, K. K. & Koh, J. T. (2010) COMP-Ang1, a chimeric form of
Angiopoietin 1, enhances BMP2-induced osteoblast differentiation and bone
formation. Bone 46, 479-486. doi:10.1016/j.bone.2009.09.019.

Jiang, Y., Jahagirdar, B. N., Reinhardt, R. L., Schwartz, R. E., Keene, C. D., Ortiz-Gonzalez, X.
R., Reyes, M., Lenvik, T., Lund, T., Blackstad, M., Du, J., Aldrich, S., Lisberg, A., Low,
W. C., Largaespada, D. A. & Verfaillie, C. M. (2002) Pluripotency of mesenchymal
stem cells derived from adult marrow. Nature 418, 41-49.
doi:10.1038/nature00870.

Jilka, R. L. (2007) Molecular and cellular mechanisms of the anabolic effect of


intermittent PTH. Bone 40, 1434-1446. doi:10.1016/j.bone.2007.03.017.

Kammerer, P. W., Scholz, M., Baudisch, M., Liese, J., Wegner, K., Frerich, B. & Lang, H.
(2017) Guided Bone Regeneration Using Collagen Scaffolds, Growth Factors, and
Periodontal Ligament Stem Cells for Treatment of Peri-Implant Bone Defects In
Vivo. Stem Cells Int 2017, 3548435. doi:10.1155/2017/3548435.

Kanji, S. & Das, H. (2017) Advances of Stem Cell Therapeutics in Cutaneous Wound
Healing and Regeneration. Mediators Inflamm 2017, 5217967.
doi:10.1155/2017/5217967.

Katagiri, W., Watanabe, J., Toyama, N., Osugi, M., Sakaguchi, K. & Hibi, H. (2017) Clinical
Study of Bone Regeneration by Conditioned Medium From Mesenchymal Stem
Cells After Maxillary Sinus Floor Elevation. Implant Dent 26, 607-612.
doi:10.1097/id.0000000000000618.

Kawaguchi, H., Oka, H., Jingushi, S., Izumi, T., Fukunaga, M., Sato, K., Matsushita, T. &
Nakamura, K. (2010) A local application of recombinant human fibroblast
growth factor 2 for tibial shaft fractures: A randomized, placebo-controlled trial.
J Bone Miner Res 25, 2735-2743. doi:10.1002/jbmr.146.

Killington, K., Mafi, R., Mafi, P. & Khan, W. S. (2018) A Systematic Review of Clinical
Studies Investigating Mesenchymal Stem Cells for Fracture Non-Union and Bone
Defects. Curr Stem Cell Res Ther 13, 284-291.
doi:10.2174/1574888x12666170915121137.

This article is protected by copyright. All rights reserved.


Kim, H. P., Ji, Y. H., Rhee, S. C., Dhong, E. S., Park, S. H. & Yoon, E. S. (2012) Enhancement
of bone regeneration using osteogenic-induced adipose-derived stem cells
combined with demineralized bone matrix in a rat critically-sized calvarial
defect model. Curr Stem Cell Res Ther 7, 165-172.
Accepted Article
Kim, S. H., Kim, K. H., Seo, B. M., Koo, K. T., Kim, T. I., Seol, Y. J., Ku, Y., Rhyu, I. C., Chung, C.
P. & Lee, Y. M. (2009) Alveolar bone regeneration by transplantation of
periodontal ligament stem cells and bone marrow stem cells in a canine peri-
implant defect model: a pilot study. J Periodontol 80, 1815-1823.
doi:10.1902/jop.2009.090249.

Kitamura, M., Akamatsu, M., Kawanami, M., Furuichi, Y., Fujii, T., Mori, M., Kunimatsu, K.,
Shimauchi, H., Ogata, Y., Yamamoto, M., Nakagawa, T., Sato, S., Ito, K., Ogasawara,
T., Izumi, Y., Gomi, K., Yamazaki, K., Yoshie, H., Fukuda, M., Noguchi, T., Takashiba,
S., Kurihara, H., Nagata, T., Hamachi, T., Maeda, K., Yokota, M., Sakagami, R., Hara,
Y., Noguchi, K., Furuuchi, T., Sasano, T., Imai, E., Ohmae, M., Koizumi, H.,
Watanuki, M. & Murakami, S. (2016) Randomized Placebo-Controlled and
Controlled Non-Inferiority Phase III Trials Comparing Trafermin, a Recombinant
Human Fibroblast Growth Factor 2, and Enamel Matrix Derivative in Periodontal
Regeneration in Intrabony Defects. J Bone Miner Res 31, 806-814.
doi:10.1002/jbmr.2738.

Kitamura, M., Akamatsu, M., Machigashira, M., Hara, Y., Sakagami, R., Hirofuji, T.,
Hamachi, T., Maeda, K., Yokota, M., Kido, J., Nagata, T., Kurihara, H., Takashiba, S.,
Sibutani, T., Fukuda, M., Noguchi, T., Yamazaki, K., Yoshie, H., Ioroi, K., Arai, T.,
Nakagawa, T., Ito, K., Oda, S., Izumi, Y., Ogata, Y., Yamada, S., Shimauchi, H.,
Kunimatsu, K., Kawanami, M., Fujii, T., Furuichi, Y., Furuuchi, T., Sasano, T., Imai,
E., Omae, M., Yamada, S., Watanuki, M. & Murakami, S. (2011) FGF-2 stimulates
periodontal regeneration: results of a multi-center randomized clinical trial. J
Dent Res 90, 35-40. doi:10.1177/0022034510384616.

Kitamura, M., Nakashima, K., Kowashi, Y., Fujii, T., Shimauchi, H., Sasano, T., Furuuchi, T.,
Fukuda, M., Noguchi, T., Shibutani, T., Iwayama, Y., Takashiba, S., Kurihara, H.,
Ninomiya, M., Kido, J., Nagata, T., Hamachi, T., Maeda, K., Hara, Y., Izumi, Y.,
Hirofuji, T., Imai, E., Omae, M., Watanuki, M. & Murakami, S. (2008) Periodontal
tissue regeneration using fibroblast growth factor-2: randomized controlled
phase II clinical trial. PLoS One 3, e2611. doi:10.1371/journal.pone.0002611.

Kode, J. A., Mukherjee, S., Joglekar, M. V. & Hardikar, A. A. (2009) Mesenchymal stem
cells: immunobiology and role in immunomodulation and tissue regeneration.
Cytotherapy 11, 377-391. doi:912826134 [pii]

10.1080/14653240903080367.

Kon, E., Muraglia, A., Corsi, A., Bianco, P., Marcacci, M., Martin, I., Boyde, A., Ruspantini, I.,
Chistolini, P., Rocca, M., Giardino, R., Cancedda, R. & Quarto, R. (2000)
Autologous bone marrow stromal cells loaded onto porous hydroxyapatite
ceramic accelerate bone repair in critical-size defects of sheep long bones. J

This article is protected by copyright. All rights reserved.


Biomed Mater Res 49, 328-337. doi:10.1002/(SICI)1097-
4636(20000305)49:3<328::AID-JBM5>3.0.CO;2-Q [pii].

Konala, V. B., Mamidi, M. K., Bhonde, R., Das, A. K., Pochampally, R. & Pal, R. (2016) The
Accepted Article
current landscape of the mesenchymal stromal cell secretome: A new paradigm
for cell-free regeneration. Cytotherapy 18, 13-24.
doi:10.1016/j.jcyt.2015.10.008.

Kuznetsov, S. A., Friedenstein, A. J. & Robey, P. G. (1997a) Factors required for bone
marrow stromal fibroblast colony formation in vitro. Br J Haematol 97, 561-570.

Kuznetsov, S. A., Krebsbach, P. H., Satomura, K., Kerr, J., Riminucci, M., Benayahu, D. &
Robey, P. G. (1997b) Single-colony derived strains of human marrow stromal
fibroblasts form bone after transplantation in vivo. J Bone Miner Res 12, 1335-
1347. doi:10.1359/jbmr.1997.12.9.1335.

Lacey, D. L., Timms, E., Tan, H. L., Kelley, M. J., Dunstan, C. R., Burgess, T., Elliott, R.,
Colombero, A., Elliott, G., Scully, S., Hsu, H., Sullivan, J., Hawkins, N., Davy, E.,
Capparelli, C., Eli, A., Qian, Y. X., Kaufman, S., Sarosi, I., Shalhoub, V., Senaldi, G.,
Guo, J., Delaney, J. & Boyle, W. J. (1998) Osteoprotegerin ligand is a cytokine that
regulates osteoclast differentiation and activation. Cell 93, 165-176.

Lendeckel, S., Jodicke, A., Christophis, P., Heidinger, K., Wolff, J., Fraser, J. K., Hedrick, M.
H., Berthold, L. & Howaldt, H. P. (2004) Autologous stem cells (adipose) and
fibrin glue used to treat widespread traumatic calvarial defects: case report. J
Craniomaxillofac Surg 32, 370-373. doi:10.1016/j.jcms.2004.06.002.

Lerner, U. H., Kindstedt, E. & Lundberk, P. (2018) The critical interplay between bone
resorbing and bone forming cells. Journal of Clinical Periodontology.

Leucht, P., Lee, S. & Yim, N. (2018) Wnt signaling and bone regeneration: Can't have one
without the other. Biomaterials. doi:10.1016/j.biomaterials.2018.03.029.

Levi, B., James, A. W., Nelson, E. R., Vistnes, D., Wu, B., Lee, M., Gupta, A. & Longaker, M. T.
(2010) Human adipose derived stromal cells heal critical size mouse calvarial
defects. PLoS One 5, e11177. doi:10.1371/journal.pone.0011177.

Li, A., Xia, X., Yeh, J., Kua, H., Liu, H., Mishina, Y., Hao, A. & Li, B. (2014) PDGF-AA
promotes osteogenic differentiation and migration of mesenchymal stem cell by
down-regulating PDGFRalpha and derepressing BMP-Smad1/5/8 signaling. PLoS
One 9, e113785. doi:10.1371/journal.pone.0113785.

Li, B., Wang, H., Qiu, G., Su, X. & Wu, Z. (2016) Synergistic Effects of Vascular Endothelial
Growth Factor on Bone Morphogenetic Proteins Induced Bone Formation In
Vivo: Influencing Factors and Future Research Directions. Biomed Res Int 2016,
2869572. doi:10.1155/2016/2869572.

Li, F., Yu, F., Xu, X., Li, C., Huang, D., Zhou, X., Ye, L. & Zheng, L. (2017) Evaluation of
Recombinant Human FGF-2 and PDGF-BB in Periodontal Regeneration: A

This article is protected by copyright. All rights reserved.


Systematic Review and Meta-Analysis. Sci Rep 7, 65. doi:10.1038/s41598-017-
00113-y.

Li, J. J., Ebied, M., Xu, J. & Zreiqat, H. (2018) Current Approaches to Bone Tissue
Accepted Article
Engineering: The Interface between Biology and Engineering. Adv Healthc Mater
7, e1701061. doi:10.1002/adhm.201701061.

Liebergall, M., Schroeder, J., Mosheiff, R., Gazit, Z., Yoram, Z., Rasooly, L., Daskal, A.,
Khoury, A., Weil, Y. & Beyth, S. (2013) Stem cell-based therapy for prevention of
delayed fracture union: a randomized and prospective preliminary study. Mol
Ther 21, 1631-1638. doi:10.1038/mt.2013.109.

Lin, N. H., Gronthos, S. & Bartold, P. M. (2008) Stem cells and periodontal regeneration.
Aust Dent J 53, 108-121. doi:10.1111/j.1834-7819.2008.00019.x.

Lindroos, B., Suuronen, R. & Miettinen, S. (2011) The potential of adipose stem cells in
regenerative medicine. Stem Cell Rev 7, 269-291. doi:10.1007/s12015-010-
9193-7.

Lindsay, R., Zhou, H., Cosman, F., Nieves, J., Dempster, D. W. & Hodsman, A. B. (2007)
Effects of a one-month treatment with PTH(1-34) on bone formation on
cancellous, endocortical, and periosteal surfaces of the human ilium. J Bone
Miner Res 22, 495-502. doi:10.1359/jbmr.070104.

Liu, G., Zhao, L., Zhang, W., Cui, L., Liu, W. & Cao, Y. (2008) Repair of goat tibial defects
with bone marrow stromal cells and beta-tricalcium phosphate. J Mater Sci
Mater Med 19, 2367-2376. doi:10.1007/s10856-007-3348-3.

Liu, X., Li, X., Fan, Y., Zhang, G., Li, D., Dong, W., Sha, Z., Yu, X., Feng, Q., Cui, F. & Watari, F.
(2010) Repairing goat tibia segmental bone defect using scaffold cultured with
mesenchymal stem cells. J Biomed Mater Res B Appl Biomater 94, 44-52.
doi:10.1002/jbm.b.31622.

Majidinia, M., Sadeghpour, A. & Yousefi, B. (2018) The roles of signaling pathways in
bone repair and regeneration. J Cell Physiol 233, 2937-2948.
doi:10.1002/jcp.26042.

Martin, T. J. & Sims, N. A. (2015) RANKL/OPG; Critical role in bone physiology. Rev
Endocr Metab Disord 16, 131-139. doi:10.1007/s11154-014-9308-6.

McAllister, B. S. (2011) Stem cell-containing allograft matrix enhances periodontal


regeneration: case presentations. Int J Periodontics Restorative Dent 31, 149-155.

McCulloch, C. A. & Bordin, S. (1991) Role of fibroblast subpopulations in periodontal


physiology and pathology. J Periodontal Res 26, 144-154.

McCulloch, C. A., Nemeth, E., Lowenberg, B. & Melcher, A. H. (1987) Paravascular cells in
endosteal spaces of alveolar bone contribute to periodontal ligament cell
populations. Anat Rec 219, 233-242. doi:10.1002/ar.1092190304.

This article is protected by copyright. All rights reserved.


Menicanin, D., Bartold, P. M., Zannettino, A. C. & Gronthos, S. (2010) Identification of a
common gene expression signature associated with immature clonal
mesenchymal cell populations derived from bone marrow and dental tissues.
Stem Cells Dev 19, 1501-1510. doi:10.1089/scd.2009.0492.
Accepted Article
Menicanin, D., Mrozik, K. M., Wada, N., Marino, V., Shi, S., Bartold, P. M. & Gronthos, S.
(2014) Periodontal-ligament-derived stem cells exhibit the capacity for long-
term survival, self-renewal, and regeneration of multiple tissue types in vivo.
Stem Cells Dev 23, 1001-1011. doi:10.1089/scd.2013.0490.

Mesimaki, K., Lindroos, B., Tornwall, J., Mauno, J., Lindqvist, C., Kontio, R., Miettinen, S. &
Suuronen, R. (2009) Novel maxillary reconstruction with ectopic bone formation
by GMP adipose stem cells. Int J Oral Maxillofac Surg 38, 201-209.
doi:10.1016/j.ijom.2009.01.001.

Mitrano, T. I., Grob, M. S., Carrion, F., Nova-Lamperti, E., Luz, P. A., Fierro, F. S., Quintero,
A., Chaparro, A. & Sanz, A. (2010) Culture and characterization of mesenchymal
stem cells from human gingival tissue. J Periodontol 81, 917-925.
doi:10.1902/jop.2010.090566.

Miura, M., Gronthos, S., Zhao, M., Lu, B., Fisher, L. W., Robey, P. G. & Shi, S. (2003) SHED:
stem cells from human exfoliated deciduous teeth. Proc Natl Acad Sci U S A 100,
5807-5812. doi:10.1073/pnas.0937635100

0937635100 [pii].

Montero, A., Okada, Y., Tomita, M., Ito, M., Tsurukami, H., Nakamura, T., Doetschman, T.,
Coffin, J. D. & Hurley, M. M. (2000) Disruption of the fibroblast growth factor-2
gene results in decreased bone mass and bone formation. J Clin Invest 105, 1085-
1093. doi:10.1172/jci8641.

Monti, M., Graziano, A., Rizzo, S., Perotti, C., Del Fante, C., d'Aquino, R., Redi, C. A. &
Rodriguez, Y. B. R. (2017) In Vitro and In Vivo Differentiation of Progenitor Stem
Cells Obtained After Mechanical Digestion of Human Dental Pulp. J Cell Physiol
232, 548-555. doi:10.1002/jcp.25452.

Morsczeck, C., Gotz, W., Schierholz, J., Zeilhofer, F., Kuhn, U., Mohl, C., Sippel, C. &
Hoffmann, K. H. (2005) Isolation of precursor cells (PCs) from human dental
follicle of wisdom teeth. Matrix Biol 24, 155-165.
doi:10.1016/j.matbio.2004.12.004.

Muraglia, A., Martin, I., Cancedda, R. & Quarto, R. (1998) A nude mouse model for human
bone formation in unloaded conditions. Bone 22, 131S-134S.

Nakamura, S., Yamada, Y., Katagiri, W., Sugito, T., Ito, K. & Ueda, M. (2009) Stem cell
proliferation pathways comparison between human exfoliated deciduous teeth
and dental pulp stem cells by gene expression profile from promising dental
pulp. J Endod 35, 1536-1542. doi:10.1016/j.joen.2009.07.024.

This article is protected by copyright. All rights reserved.


Nakashima, M. (2005) Bone morphogenetic proteins in dentin regeneration for potential
use in endodontic therapy. Cytokine Growth Factor Rev 16, 369-376. doi:S1359-
6101(05)00051-1 [pii]
Accepted Article
10.1016/j.cytogfr.2005.02.011.

Nakashima, M. & Reddi, A. H. (2003) The application of bone morphogenetic proteins to


dental tissue engineering. Nat Biotechnol 21, 1025-1032. doi:10.1038/nbt864

nbt864 [pii].

Nasef, A., Zhang, Y. Z., Mazurier, C., Bouchet, S., Bensidhoum, M., Francois, S., Gorin, N. C.,
Lopez, M., Thierry, D., Fouillard, L. & Chapel, A. (2009) Selected Stro-1-enriched
bone marrow stromal cells display a major suppressive effect on lymphocyte
proliferation. Int J Lab Hematol 31, 9-19. doi:10.1111/j.1751-
553X.2007.00997.x.

Neer, R. M., Arnaud, C. D., Zanchetta, J. R., Prince, R., Gaich, G. A., Reginster, J. Y.,
Hodsman, A. B., Eriksen, E. F., Ish-Shalom, S., Genant, H. K., Wang, O. & Mitlak, B.
H. (2001) Effect of parathyroid hormone (1-34) on fractures and bone mineral
density in postmenopausal women with osteoporosis. N Engl J Med 344, 1434-
1441. doi:10.1056/nejm200105103441904.

Nevins, M., Kao, R. T., McGuire, M. K., McClain, P. K., Hinrichs, J. E., McAllister, B. S., Reddy,
M. S., Nevins, M. L., Genco, R. J., Lynch, S. E. & Giannobile, W. V. (2013) Platelet-
derived growth factor promotes periodontal regeneration in localized osseous
defects: 36-month extension results from a randomized, controlled, double-
masked clinical trial. J Periodontol 84, 456-464. doi:10.1902/jop.2012.120141.

Nickel, J., Ten Dijke, P. & Mueller, T. D. (2018) TGF-beta family co-receptor function and
signaling. Acta Biochim Biophys Sin (Shanghai) 50, 12-36.
doi:10.1093/abbs/gmx126.

Ono, T. & Nakashima, T. (2018) Recent advances in osteoclast biology. Histochem Cell
Biol 149, 325-341. doi:10.1007/s00418-018-1636-2.

Oryan, A., Kamali, A., Moshiri, A. & Baghaban Eslaminejad, M. (2017) Role of
Mesenchymal Stem Cells in Bone Regenerative Medicine: What Is the Evidence?
Cells Tissues Organs 204, 59-83. doi:10.1159/000469704.

Owen, M. & Friedenstein, A. J. (1988) Stromal stem cells: marrow-derived osteogenic


precursors. Ciba Found Symp 136, 42-60.

Paduano, F., Marrelli, M., Amantea, M., Rengo, C., Rengo, S., Goldberg, M., Spagnuolo, G. &
Tatullo, M. (2017) Adipose Tissue as a Strategic Source of Mesenchymal Stem
Cells in Bone Regeneration: A Topical Review on the Most Promising
Craniomaxillofacial Applications. Int J Mol Sci 18. doi:10.3390/ijms18102140.

Park, B. W., Hah, Y. S., Choi, M. J., Ryu, Y. M., Lee, S. G., Kim, D. R., Kim, J. R. & Byun, J. H.
(2009) In vitro osteogenic differentiation of cultured human dental papilla-

This article is protected by copyright. All rights reserved.


derived cells. J Oral Maxillofac Surg 67, 507-514.
doi:10.1016/j.joms.2008.08.037.

Park, B. W., Kang, E. J., Byun, J. H., Son, M. G., Kim, H. J., Hah, Y. S., Kim, T. H., Mohana
Accepted Article
Kumar, B., Ock, S. A. & Rho, G. J. (2012) In vitro and in vivo osteogenesis of
human mesenchymal stem cells derived from skin, bone marrow and dental
follicle tissues. Differentiation 83, 249-259. doi:10.1016/j.diff.2012.02.008.

Parrilla, C., Saulnier, N., Bernardini, C., Patti, R., Tartaglione, T., Fetoni, A. R., Pola, E.,
Paludetti, G., Michetti, F. & Lattanzi, W. (2011) Undifferentiated human adipose
tissue-derived stromal cells induce mandibular bone healing in rats. Arch
Otolaryngol Head Neck Surg 137, 463-470. doi:10.1001/archoto.2011.61.

Pereira, R. F., Halford, K. W., O'Hara, M. D., Leeper, D. B., Sokolov, B. P., Pollard, M. D.,
Bagasra, O. & Prockop, D. J. (1995) Cultured adherent cells from marrow can
serve as long-lasting precursor cells for bone, cartilage, and lung in irradiated
mice. Proc Natl Acad Sci U S A 92, 4857-4861.

Petersen, B. E., Bowen, W. C., Patrene, K. D., Mars, W. M., Sullivan, A. K., Murase, N., Boggs,
S. S., Greenberger, J. S. & Goff, J. P. (1999) Bone marrow as a potential source of
hepatic oval cells. Science 284, 1168-1170.

Petite, H., Viateau, V., Bensaid, W., Meunier, A., de Pollak, C., Bourguignon, M., Oudina, K.,
Sedel, L. & Guillemin, G. (2000) Tissue-engineered bone regeneration. Nat
Biotechnol 18, 959-963. doi:10.1038/79449.

Pieri, F., Lucarelli, E., Corinaldesi, G., Aldini, N. N., Fini, M., Parrilli, A., Dozza, B., Donati, D.
& Marchetti, C. (2010) Dose-dependent effect of adipose-derived adult stem cells
on vertical bone regeneration in rabbit calvarium. Biomaterials 31, 3527-3535.
doi:10.1016/j.biomaterials.2010.01.066.

Pietrzyk, B., Smertka, M. & Chudek, J. (2017) Sclerostin: Intracellular mechanisms of


action and its role in the pathogenesis of skeletal and vascular disorders. Adv
Clin Exp Med 26, 1283-1291. doi:10.17219/acem/68739.

Pittenger, M. F., Mackay, A. M., Beck, S. C., Jaiswal, R. K., Douglas, R., Mosca, J. D.,
Moorman, M. A., Simonetti, D. W., Craig, S. & Marshak, D. R. (1999) Multilineage
potential of adult human mesenchymal stem cells. Science 284, 143-147.

Pourebrahim, N., Hashemibeni, B., Shahnaseri, S., Torabinia, N., Mousavi, B., Adibi, S.,
Heidari, F. & Alavi, M. J. (2013) A comparison of tissue-engineered bone from
adipose-derived stem cell with autogenous bone repair in maxillary alveolar
cleft model in dogs. Int J Oral Maxillofac Surg 42, 562-568.
doi:10.1016/j.ijom.2012.10.012.

Psaltis, P. J., Paton, S., See, F., Arthur, A., Martin, S., Itescu, S., Worthley, S. G., Gronthos, S.
& Zannettino, A. C. (2010) Enrichment for STRO-1 expression enhances the
cardiovascular paracrine activity of human bone marrow-derived mesenchymal
cell populations. J Cell Physiol 223, 530-540. doi:10.1002/jcp.22081.

This article is protected by copyright. All rights reserved.


Rezai-Rad, M., Bova, J. F., Orooji, M., Pepping, J., Qureshi, A., Del Piero, F., Hayes, D. & Yao,
S. (2015) Evaluation of bone regeneration potential of dental follicle stem cells
for treatment of craniofacial defects. Cytotherapy 17, 1572-1581.
doi:10.1016/j.jcyt.2015.07.013.
Accepted Article
Rho, J. Y., Kuhn-Spearing, L. & Zioupos, P. (1998) Mechanical properties and the
hierarchical structure of bone. Med Eng Phys 20, 92-102.

Robling, A. G. & Turner, C. H. (2009) Mechanical signaling for bone modeling and
remodeling. Crit Rev Eukaryot Gene Expr 19, 319-338.

Sandor, G. K. (2012) Tissue engineering of bone: Clinical observations with adipose-


derived stem cells, resorbable scaffolds, and growth factors. Ann Maxillofac Surg
2, 8-11. doi:10.4103/2231-0746.95308.

Sandor, G. K., Tuovinen, V. J., Wolff, J., Patrikoski, M., Jokinen, J., Nieminen, E.,
Mannerstrom, B., Lappalainen, O. P., Seppanen, R. & Miettinen, S. (2013) Adipose
stem cell tissue-engineered construct used to treat large anterior mandibular
defect: a case report and review of the clinical application of good manufacturing
practice-level adipose stem cells for bone regeneration. J Oral Maxillofac Surg 71,
938-950. doi:10.1016/j.joms.2012.11.014.

Seo, B. M., Miura, M., Gronthos, S., Bartold, P. M., Batouli, S., Brahim, J., Young, M., Robey,
P. G., Wang, C. Y. & Shi, S. (2004) Investigation of multipotent postnatal stem
cells from human periodontal ligament. Lancet 364, 149-155.
doi:10.1016/s0140-6736(04)16627-0.

Seo, B. M., Sonoyama, W., Yamaza, T., Coppe, C., Kikuiri, T., Akiyama, K., Lee, J. S. & Shi, S.
(2008) SHED repair critical-size calvarial defects in mice. Oral Dis 14, 428-434.

Shanbhag, S., Suliman, S., Pandis, N., Stavropoulos, A., Sanz, M. & Mustafa, K. (2018) Cell
therapy for orofacial bone regeneration: a systematic review and meta-analysis.
Journal of Clinical Periodontology, 315-.

Shapiro, F. (2008) Bone development and its relation to fracture repair. The role of
mesenchymal osteoblasts and surface osteoblasts. Eur Cell Mater 15, 53-76.

Silva, B. C. & Bilezikian, J. P. (2015) Parathyroid hormone: anabolic and catabolic actions
on the skeleton. Curr Opin Pharmacol 22, 41-50.
doi:10.1016/j.coph.2015.03.005.

Simmons, P. J. & Torok-Storb, B. (1991) Identification of stromal cell precursors in


human bone marrow by a novel monoclonal antibody, STRO-1. Blood 78, 55-62.

Simonet, W. S., Lacey, D. L., Dunstan, C. R., Kelley, M., Chang, M. S., Luthy, R., Nguyen, H.
Q., Wooden, S., Bennett, L., Boone, T., Shimamoto, G., DeRose, M., Elliott, R.,
Colombero, A., Tan, H. L., Trail, G., Sullivan, J., Davy, E., Bucay, N., Renshaw-Gegg,
L., Hughes, T. M., Hill, D., Pattison, W., Campbell, P., Sander, S., Van, G., Tarpley, J.,
Derby, P., Lee, R. & Boyle, W. J. (1997) Osteoprotegerin: a novel secreted protein
involved in the regulation of bone density. Cell 89, 309-319.

This article is protected by copyright. All rights reserved.


Smith, J. R., Pochampally, R., Perry, A., Hsu, S. C. & Prockop, D. J. (2004) Isolation of a
highly clonogenic and multipotential subfraction of adult stem cells from bone
marrow stroma. Stem Cells 22, 823-831. doi:10.1634/stemcells.22-5-823.
Accepted Article
Sohn, W., Simiens, M. A., Jaeger, K., Hutton, S. & Jang, G. (2014) The pharmacokinetics
and pharmacodynamics of denosumab in patients with advanced solid tumours
and bone metastases: a systematic review. Br J Clin Pharmacol 78, 477-487.
doi:10.1111/bcp.12355.

Sonoyama, W., Liu, Y., Fang, D., Yamaza, T., Seo, B. M., Zhang, C., Liu, H., Gronthos, S.,
Wang, C. Y., Wang, S. & Shi, S. (2006) Mesenchymal stem cell-mediated functional
tooth regeneration in swine. PLoS One 1, e79.
doi:10.1371/journal.pone.0000079.

Sonoyama, W., Liu, Y., Yamaza, T., Tuan, R. S., Wang, S., Shi, S. & Huang, G. T. (2008)
Characterization of the apical papilla and its residing stem cells from human
immature permanent teeth: a pilot study. J Endod 34, 166-171.
doi:10.1016/j.joen.2007.11.021.

Srisuwan, T., Tilkorn, D. J., Wilson, J. L., Morrison, W. A., Messer, H. M., Thompson, E. W. &
Abberton, K. M. (2006) Molecular aspects of tissue engineering in the dental
field. Periodontol 2000 41, 88-108. doi:PRD176 [pii]

10.1111/j.1600-0757.2006.00176.x.

Stegen, S., van Gastel, N. & Carmeliet, G. (2015) Bringing new life to damaged bone: the
importance of angiogenesis in bone repair and regeneration. Bone 70, 19-27.
doi:10.1016/j.bone.2014.09.017.

Tajima, S., Tobita, M., Orbay, H., Hyakusoku, H. & Mizuno, H. (2015) Direct and indirect
effects of a combination of adipose-derived stem cells and platelet-rich plasma
on bone regeneration. Tissue Eng Part A 21, 895-905.
doi:10.1089/ten.TEA.2014.0336.

Takahashi, K. & Yamanaka, S. (2006) Induction of pluripotent stem cells from mouse
embryonic and adult fibroblast cultures by defined factors. Cell 126, 663-676.
doi:10.1016/j.cell.2006.07.024.

Tanaka, T., Kitasato, S., Chazono, M., Kumagae, Y., Iida, T., Mitsuhashi, M., Kakuta, A. &
Marumo, K. (2012) Use of an Injectable Complex of beta-Tricalcium Phosphate
Granules, Hyaluronate, and Fibroblast Growth Factor-2 on Repair of Unstable
Intertrochanteric Fractures. Open Biomed Eng J 6, 98-103.
doi:10.2174/1874120701206010098.

Tang, Y., Wu, X., Lei, W., Pang, L., Wan, C., Shi, Z., Zhao, L., Nagy, T. R., Peng, X., Hu, J., Feng,
X., Van Hul, W., Wan, M. & Cao, X. (2009) TGF-beta1-induced migration of bone
mesenchymal stem cells couples bone resorption with formation. Nat Med 15,
757-765. doi:10.1038/nm.1979.

This article is protected by copyright. All rights reserved.


Thesleff, T., Lehtimaki, K., Niskakangas, T., Mannerstrom, B., Miettinen, S., Suuronen, R. &
Ohman, J. (2011) Cranioplasty with adipose-derived stem cells and biomaterial:
a novel method for cranial reconstruction. Neurosurgery 68, 1535-1540.
doi:10.1227/NEU.0b013e31820ee24e.
Accepted Article
Tomar, G. B., Srivastava, R. K., Gupta, N., Barhanpurkar, A. P., Pote, S. T., Jhaveri, H. M.,
Mishra, G. C. & Wani, M. R. Human gingiva-derived mesenchymal stem cells are
superior to bone marrow-derived mesenchymal stem cells for cell therapy in
regenerative medicine. Biochem 393, 377-383. Epub 2010 Feb 2016.

Tomar, G. B., Srivastava, R. K., Gupta, N., Barhanpurkar, A. P., Pote, S. T., Jhaveri, H. M.,
Mishra, G. C. & Wani, M. R. (2010) Human gingiva-derived mesenchymal stem
cells are superior to bone marrow-derived mesenchymal stem cells for cell
therapy in regenerative medicine. Biochem Biophys Res Commun 393, 377-383.
doi:10.1016/j.bbrc.2010.01.126.

Tour, G., Wendel, M., Moll, G. & Tcacencu, I. (2012) Bone repair using periodontal
ligament progenitor cell-seeded constructs. J Dent Res 91, 789-794.
doi:10.1177/0022034512452430.

Tsuchiya, S., Ohshima, S., Yamakoshi, Y., Simmer, J. P. & Honda, M. J. (2010) Osteogenic
differentiation capacity of porcine dental follicle progenitor cells. Connect Tissue
Res 51, 197-207. doi:10.3109/03008200903267542.

Tsukamoto, J., Naruse, K., Nagai, Y., Kan, S., Nakamura, N., Hata, M., Omi, M., Hayashi, T.,
Kawai, T. & Matsubara, T. (2017) (*) Efficacy of a Self-Assembling Peptide
Hydrogel, SPG-178-Gel, for Bone Regeneration and Three-Dimensional
Osteogenic Induction of Dental Pulp Stem Cells. Tissue Eng Part A 23, 1394-1402.
doi:10.1089/ten.TEA.2017.0025.

Tu, X., Chen, J., Lim, J., Karner, C. M., Lee, S. Y., Heisig, J., Wiese, C., Surendran, K., Kopan,
R., Gessler, M. & Long, F. (2012) Physiological notch signaling maintains bone
homeostasis via RBPjk and Hey upstream of NFATc1. PLoS Genet 8, e1002577.
doi:10.1371/journal.pgen.1002577.

Urist, M. R. (1965) Bone: formation by autoinduction. Science 150, 893-899.

Valderrama, P., Jung, R. E., Thoma, D. S., Jones, A. A. & Cochran, D. L. (2010) Evaluation of
parathyroid hormone bound to a synthetic matrix for guided bone regeneration
around dental implants: a histomorphometric study in dogs. J Periodontol 81,
737-747. doi:10.1902/jop.2010.090562.

Vannucci, L. & Brandi, M. L. (2016) Healing of the bone with anti-fracture drugs. Expert
Opin Pharmacother 17, 2267-2272. doi:10.1080/14656566.2016.1241765.

Vaquette, C., Pilipchuk, S. P., Bartold, P. M., Hutmacher, D. W., Giannobile, W. V. &
Ivanovski, S. (2018) Tissue Engineered Constructs for Periodontal Regeneration:
Current Status and Future Perspectives. Adv Healthc Mater, e1800457.
doi:10.1002/adhm.201800457.

This article is protected by copyright. All rights reserved.


Varghese, J., Griffin, M., Mosahebi, A. & Butler, P. (2017) Systematic review of patient
factors affecting adipose stem cell viability and function: implications for
regenerative therapy. Stem Cell Res Ther 8, 45. doi:10.1186/s13287-017-0483-8.
Accepted Article
Viateau, V., Guillemin, G., Bousson, V., Oudina, K., Hannouche, D., Sedel, L., Logeart-
Avramoglou, D. & Petite, H. (2007) Long-bone critical-size defects treated with
tissue-engineered grafts: a study on sheep. J Orthop Res 25, 741-749.
doi:10.1002/jor.20352.

Wagner, W., Wein, F., Seckinger, A., Frankhauser, M., Wirkner, U., Krause, U., Blake, J.,
Schwager, C., Eckstein, V., Ansorge, W. & Ho, A. D. (2005) Comparative
characteristics of mesenchymal stem cells from human bone marrow, adipose
tissue, and umbilical cord blood. Exp Hematol 33, 1402-1416. doi:S0301-
472X(05)00364-4 [pii]

10.1016/j.exphem.2005.07.003.

Walters, G., Pountos, I. & Giannoudis, P. V. (2018) The cytokines and micro-environment
of fracture haematoma: Current evidence. J Tissue Eng Regen Med 12, e1662-
e1677. doi:10.1002/term.2593.

Wang, C., Inzana, J. A., Mirando, A. J., Ren, Y., Liu, Z., Shen, J., O'Keefe, R. J., Awad, H. A. &
Hilton, M. J. (2016) NOTCH signaling in skeletal progenitors is critical for
fracture repair. J Clin Invest 126, 1471-1481. doi:10.1172/jci80672.

Wang, F., Yu, M., Yan, X., Wen, Y., Zeng, Q., Yue, W., Yang, P. & Pei, X. (2011) Gingiva-
derived mesenchymal stem cell-mediated therapeutic approach for bone tissue
regeneration. Stem Cells Dev 20, 2093-2102. doi:10.1089/scd.2010.0523.

Wang, S., Mu, J., Fan, Z., Yu, Y., Yan, M., Lei, G., Tang, C., Wang, Z., Zheng, Y., Yu, J. & Zhang,
G. (2012) Insulin-like growth factor 1 can promote the osteogenic differentiation
and osteogenesis of stem cells from apical papilla. Stem Cell Res 8, 346-356.
doi:10.1016/j.scr.2011.12.005.

Wilson, S. M., Goldwasser, M. S., Clark, S. G., Monaco, E., Bionaz, M., Hurley, W. L.,
Rodriguez-Zas, S., Feng, L., Dymon, Z. & Wheeler, M. B. (2012) Adipose-derived
mesenchymal stem cells enhance healing of mandibular defects in the ramus of
swine. J Oral Maxillofac Surg 70, e193-203. doi:10.1016/j.joms.2011.10.029.

Wise, J. K., Sena, K., Vranizan, K., Pollock, J. F., Healy, K. E., Hughes, W. F., Sumner, D. R. &
Virdi, A. S. (2010) Temporal gene expression profiling during rat femoral
marrow ablation-induced intramembranous bone regeneration. PLoS One 5.
doi:10.1371/journal.pone.0012987.

Wongsupa, N., Nuntanaranont, T., Kamolmattayakul, S. & Thuaksuban, N. (2017)


Assessment of bone regeneration of a tissue-engineered bone complex using
human dental pulp stem cells/poly(epsilon-caprolactone)-biphasic calcium
phosphate scaffold constructs in rabbit calvarial defects. J Mater Sci Mater Med
28, 77. doi:10.1007/s10856-017-5883-x.

This article is protected by copyright. All rights reserved.


Woodbury, D., Schwarz, E. J., Prockop, D. J. & Black, I. B. (2000) Adult rat and human
bone marrow stromal cells differentiate into neurons. J Neurosci Res 61, 364-
370. doi:10.1002/1097-4547(20000815)61:4<364::aid-jnr2>3.0.co;2-c.
Accepted Article
Wozney, J. M., Rosen, V., Celeste, A. J., Mitsock, L. M., Whitters, M. J., Kriz, R. W., Hewick, R.
M. & Wang, E. A. (1988) Novel regulators of bone formation: molecular clones
and activities. Science 242, 1528-1534.

Wu, J., Huang, G. T., He, W., Wang, P., Tong, Z., Jia, Q., Dong, L., Niu, Z. & Ni, L. (2012) Basic
fibroblast growth factor enhances stemness of human stem cells from the apical
papilla. J Endod 38, 614-622. doi:10.1016/j.joen.2012.01.014.

Wu, M., Chen, G. & Li, Y. P. (2016) TGF-beta and BMP signaling in osteoblast, skeletal
development, and bone formation, homeostasis and disease. Bone Res 4, 16009.
doi:10.1038/boneres.2016.9.

Yamada, Y., Ito, K., Nakamura, S., Ueda, M. & Nagasaka, T. (2011) Promising cell-based
therapy for bone regeneration using stem cells from deciduous teeth, dental
pulp, and bone marrow. Cell Transplant 20, 1003-1013.
doi:10.3727/096368910x539128.

Yamada, Y., Ueda, M., Hibi, H. & Baba, S. (2006) A novel approach to periodontal tissue
regeneration with mesenchymal stem cells and platelet-rich plasma using tissue
engineering technology: A clinical case report. Int J Periodontics Restorative Dent
26, 363-369.

Yao, S., He, H., Gutierrez, D. L., Rad, M. R., Liu, D., Li, C., Flanagan, M. & Wise, G. E. (2013)
Expression of bone morphogenetic protein-6 in dental follicle stem cells and its
effect on osteogenic differentiation. Cells Tissues Organs 198, 438-447.
doi:10.1159/000360275.

Younger, A., Penner, M. & Montijo, H. E. (2016) Vancouver Experience of Recombinant


Human Platelet-Derived Growth Factor. Foot Ankle Clin 21, 771-776.
doi:10.1016/j.fcl.2016.07.009.

Yu, B. H., Zhou, Q. & Wang, Z. L. (2014) Periodontal ligament versus bone marrow
mesenchymal stem cells in combination with Bio-Oss scaffolds for ectopic and in
situ bone formation: A comparative study in the rat. J Biomater Appl 29, 243-
253. doi:10.1177/0885328214521846.

Yu, M., Ge, S., Wang, F., Wen, Y., Yan, X., Zeng, Q., Yue, W., Yang, P. & Pei, X. (2013) The
role of systemically delivered bone marrow-derived mesenchymal stem cells in
the regeneration of periodontal tissues. Int J Oral maxillofac Implants 28, e503-
511. doi:10.11607/jomi.te31.

Yuan, J., Cui, L., Zhang, W. J., Liu, W. & Cao, Y. (2007) Repair of canine mandibular bone
defects with bone marrow stromal cells and porous beta-tricalcium phosphate.
Biomaterials 28, 1005-1013. doi:S0142-9612(06)00885-4 [pii]

10.1016/j.biomaterials.2006.10.015.

This article is protected by copyright. All rights reserved.


Zarei, F. & Soleimaninejad, M. (2018) Role of growth factors and biomaterials in wound
healing. Artif Cells Nanomed Biotechnol, 1-6.
doi:10.1080/21691401.2018.1439836.
Accepted Article
Zhang, Q., Shi, S., Liu, Y., Uyanne, J., Shi, Y., Shi, S. & Le, A. D. (2009) Mesenchymal stem
cells derived from human gingiva are capable of immunomodulatory functions
and ameliorate inflammation-related tissue destruction in experimental colitis. J
Immunol 183, 7787-7798. doi:10.4049/jimmunol.0902318.

Zheng, Y., Liu, Y., Zhang, C. M., Zhang, H. Y., Li, W. H., Shi, S., Le, A. D. & Wang, S. L. (2009)
Stem cells from deciduous tooth repair mandibular defect in swine. J Dent Res
88, 249-254. doi:10.1177/0022034509333804.

Zhu, L., Liu, W., Cui, L. & Cao, Y. (2006) Tissue-engineered bone repair of goat-femur
defects with osteogenically induced bone marrow stromal cells. Tissue Eng 12,
423-433. doi:10.1089/ten.2006.12.423.

Zuk, P. A., Zhu, M., Mizuno, H., Huang, J., Futrell, J. W., Katz, A. J., Benhaim, P., Lorenz, H. P.
& Hedrick, M. H. (2001) Multilineage cells from human adipose tissue:
implications for cell-based therapies. Tissue Eng 7, 211-228.
doi:10.1089/107632701300062859.

Figure Legends

Figure 1. Bone is composed of periosteal, cortical and cancellous compartments

Figure 2. Structural components of the alveolar bone include the alveolar process of
the maxilla and mandible that contains developing teeth as well as the roots of fully
erupted teeth. The other type of alveolar bone is termed “alveolar bone proper” and
this is the portion of the alveolar bone that houses the tooth roots of erupted teeth
and associated periodontal ligament.

Figure 3. Stem cell lineages. Classification of stem cells is based upon their
differentiation potency and is determined by the site, stage of development or cell
culture induction environment in which the populations are derived. Cells may be
categorised as totipotent, pluripotent, multipotent or induced pluripotent. (Adapted
with permission (Lin et al., 2008).

This article is protected by copyright. All rights reserved.


Figure 4. Cellular and molecular players in bone formation, resorption and
remodelling. The complex interplay between stem cells, growth factors, cell surface
ligands and bone cells is illustrated.
Accepted Article
Abbreviations:

BMPs: bone morphogenetic proteins; Ca2+; Calcium ions CTGF: connective tissue
growth factor; EGF: epidermal growth factor ERBs: a family of receptor tyrosine
kinase receptors related to the epidermal growth factor receptor ET-1: endothelin 1;
IGFs; insulin-like growth factors; IL-1: interleukin-1; IL-11: interleukin-11 IL-6:
interleukin-6; OPG: osteoprotogerin; PDGF: platelet derived growth factor; PTHrP;
parathyroid related protein; RANK: Receptor activator of nuclear factor kappa-
Β; RANKL: Receptor activator of nuclear factor kappa-Β ligand; TGF-
growth factor beta; uPA: urokinase-type plasminogen activator.

Table 1

Different Stem Cell Populations used for Bone and Periodontal Regeneration

Bone Marrow Stromal Cells (BMSC)

Dental Derived Mesenchymal Stem cell-like Cells

Dental Pulp Stem Cells (DPSC)

Periodontal Ligament Stem Cells (PDLSC)

Stem Cells from Human Exfoliated Deciduous Teeth (SHED)

Stem Cells from Apical Papilla

Dental Follicle Cells (DFC)

Gingival Mesenchymal Stem Cells (GMSC)

Adipose Tissue Stem Cells (ASC)

This article is protected by copyright. All rights reserved.


Table 2

Biologic Factors Associated with Bone Formation and Regeneration


Accepted Article
Bone–derived Growth Factors & Differentiation Factors

Bone Morphogenetic Proteins (BMPs)

BMP-2

BMP-7

Platelet Derived Growth Factor (PDGF)

Fibroblast Growth Factors (FGF)

aFGF

bFGF

Transforming Growth Factor (TGF-beta)

Growth Differentiation Factors (GDF)

Insulin-like Growth Factors (IGF)

IGF-1

IGF-2

Vascular Endothelial Cell Growth Factor (VEGF)

Skeletal Growth Factor (SGF)

Parathyroid Hormone-related Peptide (PTHrP)

Bone Growth and Regeneration Signalling Pathways

TGF-β Family Signaling

FGF Signaling
Wnt Signaling
Hh Signaling

This article is protected by copyright. All rights reserved.


Bone Growth and Regeneration Families of Transcription Factors
Homeobox Gene Family of Transcription Factors
Accepted Article
Dlx Homeobox Gene Family

Homeobox Gene Family

Hox Homeobox Gene Family

Paired Box (Pax) Homeobox Gene Family

LIM Homeobox Gene Family (Lhx)

Paired-Like (Pitx) Homeobox Gene Family

Runx Transcription Factors


SRY-Related HMG-Box Family of Transcription Factors
bHLH Family of Transcription Factors
Twist

D proteins

The myogenic regulatory factors (MRFs)


Snail Family of Transcription Factors
Smad Transcription Factors
β-Catenin/LEF/TCF Transcription Factors
Gli Transcription Factors
Forkhead Family of Transcription Factors

This article is protected by copyright. All rights reserved.


Accepted Article

This article is protected by copyright. All rights reserved.


Accepted Article

This article is protected by copyright. All rights reserved.


Accepted Article

This article is protected by copyright. All rights reserved.

You might also like