You are on page 1of 13

Injury, Int. J.

Care Injured (2005) 36, 1392—1404

www.elsevier.com/locate/injury

REVIEW

Current concepts of molecular aspects


of bone healing
Rozalia Dimitriou 1, Eleftherios Tsiridis, Peter V. Giannoudis *

Academic Department of Trauma and Orthopaedic Surgery, School of Medicine,


University of Leeds, St James’s University Hospital, Backett Street, LS9 7TF, UK

Accepted 21 July 2005

KEYWORDS Summary Fracture healing is a complex physiological process. It involves the


Molecular; coordinated participation of haematopoietic and immune cells within the bone
Bone; marrow in conjunction with vascular and skeletal cell precursors, including mesench-
Ageing; ymal stem cells (MSCs) that are recruited from the surrounding tissues and the
Fracture healing; circulation. Multiple factors regulate this cascade of molecular events by affecting
Trauma different sites in the osteoblast and chondroblast lineage through various processes
such as migration, proliferation, chemotaxis, differentiation, inhibition, and extra-
cellular protein synthesis. An understanding of the fracture healing cellular and
molecular pathways is not only critical for the future advancement of fracture
treatment, but it may also be informative to our further understanding of the
mechanisms of skeletal growth and repair as well as the mechanisms of aging.
# 2005 Elsevier Ltd. All rights reserved.

Contents

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1393
Historical perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1393
Biology of fracture healing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1393
Direct or primary cortical fracture healing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1393
Indirect or secondary fracture healing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1394
Molecular aspects of fracture healing. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1394
Signalling molecules and the role of mesenchymal stem cells (MSCs) . . . . . . . . . . . . . . . . . . . . . . . . . 1394
Pro-inflammatory cytokines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1394
Growth and differentiation factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1394
Metalloproteinases and angiogenic factors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1399
The role of mesenchymal stem cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1399
Sequence of events . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1400

* Corresponding author. Present address: Academic Department of Trauma and Orthopaedics, St. James’s University Hospital, Beckett
Street, Leeds LS9 7TF, UK. Tel.: +44 113 20 66460; fax: +44 113 20 65156.
E-mail address: pgiannoudi@aol.com (P.V. Giannoudis).
1
AO Research Fellow.

0020–1383/$ — see front matter # 2005 Elsevier Ltd. All rights reserved.
doi:10.1016/j.injury.2005.07.019
Current concepts of molecular aspects of bone healing 1393

Intramembranous bone formation . . . . ........... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1400


Endochondral bone formation . . . . . . . ........... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1400
Current concepts of systemic enhancement of fracture healing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1401
Parathyroid hormone (PTH) . . . . . . . . . . ........... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1401
Growth hormone (GH) . . . . . . . . . . . . . ........... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1401
Future directions. . . . . . . . . . . . . . . . . . . ........... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1401
Tissue engineering . . . . . . . . . . . . . . . . ........... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1401
Muscle stem cells . . . . . . . . . . . . . . . . ........... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1401
Clinical applications in trauma . . . . . . . . . . ........... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1402
Conclusion . . . . . . . . . . . . . . . . . . . . . . . ........... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1402
Acknowledgements . . . . . . . . . . . . . . . . . ........... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1402
References. . . . . . . . . . . . . . . . . . . . . . . ........... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1402

Introduction ongoing to elucidate the unique role of different


molecules and their interactions in the healing
Fracture healing remains to a great extent an cascade.
unknown cascade of complex biological events. It
involves intracellular and extracellular molecular
signalling for bone induction and conduction. It is Biology of fracture healing
a multistage repair process that follows a definable
temporal and spatial sequence.20,23,63,68 Fracture healing is a complex, however, well orche-
Molecular mechanisms known to regulate skeletal strated, regenerative process initiated in response
tissue formation during embryological development to injury, resulting in optimal skeletal repair and
are recapitulated during fracture healing.25 Many restoration of skeletal function. During the repair
local and systemic regulatory factors, including process, the pathway of normal embryonic devel-
growth and differentiation factors, hormones, cyto- opment is recapitulated with the coordinated par-
kines, and extracellular matrix, interact with sev- ticipation of several cell types.25 All four
eral cell types, including bone and cartilage forming components involved in the injury site, including
primary cells or even muscle mesenchymal cells, the cortex, the periosteum, the bone marrow, and
recruited at the fracture-injury site or from the the external soft tissues, contribute in the healing
circulation. process at different extent, depending on multiple
Cellular and molecular biology provides today parameters present at the injured tissue such as
the tolls for the investigation and understanding growth factors, hormones and nutrients, pH, oxygen
of the fracture healing process. Ongoing research tension, the electrical environment and the
in this field of medicine has improved our under- mechanical stability that has been obtained.17,65
standing of fracture healing at the molecular level. In classical histological terms, fracture healing
The aim of this review article is to characterise the has been divided into direct (primary) and indirect
chain of events contributing to the healing process (secondary) fracture healing. Integrated cellular
in order to enhance the clinician’s awareness of the events, and their temporal and spatial character-
complexity of signalling pathways and molecules istics, have been elucidated by using a model of
involved. experimental fracture healing in the rat.23

Direct or primary cortical fracture healing


Historical perspectives
Direct fracture healing occurs only when there is
In 1965, Urist revolutionised the current under- anatomic reduction of the fracture fragments by
standing of fracture healing by hypothesising the rigid internal fixation and decreased intrafragmen-
existence of bone morphogenetic proteins (BMPs) tary strain.49 This process involves a direct attempt
allocated onto the extracellular collagenous by the cortex to re-establish new haversian systems
matrix.76,77 The genetic sequences of BMPs were by the formation of discrete remodelling units
first identified by Wozney et al. in 1988,82 and since known as ‘cutting cones’, in order to restore
then over 40 BMPs have been discovered. Subse- mechanical continuity.49 Vascular endothelial cells
quently, with the advances made in molecular med- and perivascular mesenchymal cells provide the
icine and molecular biology extensive research is osteoprogenitor cells to become osteoblasts. During
1394 R. Dimitriou et al.

this process, little or no periosteal response is noted (2) the transforming growth factor-beta (TGF-b)
(no callus formation).23 superfamily and other growth factors, and (3) the
angiogenic factors (Table 1).33,44
Indirect or secondary fracture healing
Pro-inflammatory cytokines
The majority of fractures heal by indirect fracture Interleukin-1 (IL-1) and Interleukin-6 (IL-6) as well
healing. It involves a combination of intramembra- as tumour necrosis factor-alpha (TNF-a) are shown
nous and endochondral ossification with the subse- to play a role in initiating the repair cascade.24,32
quent formation of a callus.23 It is generally enhanced They are secreted not only by macrophages and
by motion and inhibited by rigid fixation.49 inflammatory cells but also by cells of mesenchymal
Intramembranous ossification involves the origin present in the periosteum (Table 1).43 They
formation of bone directly, without first forming carry out central functions in the induction of down-
cartilage, from committed osteoprogenitor and stream responses to injury by having a chemotactic
undifferentiated mesenchymal cells that resides effect on other inflammatory cells, enhancing extra-
in the periosteum, farther from the fracture site.23 cellular matrix synthesis, stimulating angiogenesis,
It results in callus formation, described histologi- and recruiting endogenous fibrogenic cells to the
cally as ‘hard callus’.23 In this type of healing, the injury site.43
bone marrow’s contribution to the formation of They show peak expression within the first 24 h
bone is during the early phase of healing, when after fracture, depressed levels during the period of
endothelial cells transform into polymorphic cartilage formation, and their levels increase again
cells, which subsequently express an osteoblastic during bone remodelling (Fig. 1).32,43
phenotype.12 Cytokines also regulate endochondral bone for-
Endochondral ossification involves the recruit- mation and remodelling.7 TNF-a promotes the
ment, proliferation, and differentiation of undiffer- recruitment of mesenchymal stem cells, induces
entiated mesenchymal cells into cartilage, which apoptosis of hypertrophic chondrocytes during
becomes calcified and eventually replaced by bone. endochondral ossification and stimulates osteoclas-
Its temporal characteristics include six identifiable tic function. Absence of TNF-a result in delayed
stages including an initial stage of haematoma for- resorption of mineralized cartilage, prohibiting
mation and inflammation, subsequent angiogenesis new bone formation.32 IL-1, IL-6 and TNF-a also
and formation of cartilage, cartilage calcification, show increased levels of expression during fracture
cartilage removal, bone formation, and ultimately callus re-shaping later in the process of fracture
bone remodelling.23 This type of fracture healing, is healing and remodelling.
contributed from the adjacent to the fracture peri-
osteum and the external soft tissues, providing an Growth and differentiation factors
early bridging callus, histologically characterized as The transforming growth factor-beta superfami-
‘soft callus’, that stabilizes the fracture frag- ly. The TGF-b superfamily is a large family of
ments.23 growth and differentiation factors including bone
The classification of fracture healing in direct and morphogenetic proteins (BMPs), transforming
indirect healing reflects the histological events that growth factor-beta (TGF-b), growth differentiation
occur during the repair process. However, the factors (GDFs), activins, inhibins and the Mullerian
ongoing research in bone regeneration provided a inhibiting substance. At least 34 members have been
further understanding of the cellular and molecular identified in the human genome.75
pathways that govern these events, by demonstrat- They originate from high molecular weight pre-
ing the existence of various signalling molecules and cursors and are activated by proteolytic enzymes.73
elucidating their contribution in the initiation and They act on serine/threonine kinase membrane
control of this physiological process at the molecu- receptor on target cells.48 This ligand—receptor
lar level. interaction activates an intracellular signalling path-
way which ultimately affects gene expression in the
nucleus.
Molecular aspects of fracture healing Specific members of this superfamily including
bone morphogenetic proteins (BMPs 1—8), growth
Signalling molecules and the role of and differentiation factors (GDF-1, 5, 8, 10) and
mesenchymal stem cells (MSCs) transforming factor beta (TGF-b1, -b2, -b3), pro-
mote the various stages of intramembranous and
The signalling molecules can be categorized into endochondral bone ossification during fracture
three groups: (1) the pro-inflammatory cytokines, healing.20
Current concepts of molecular aspects of bone healing 1395

Table 1 The essential signalling molecules during fracture healing; their source and targeted cells, and their major
functions and expression patterns
Cytokines (IL-1, IL-6, TNF-a)
Source: macrophages and other inflammatory cells, cells of mesenchymal origin
Chemotactic effect on other inflammatory cells, stimulation of extracellular matrix synthesis,
angiogenesis, recruitment of endogenous fibrogenic cells to the injury site, and
at later stages bone resorption
Increased levels from days 1 to 3 and during bone remodelling

TGF-b
Source: degranulating platelets, inflammatory cells, endothelium, extracellular matrix,
chondrocytes, osteoblasts
Targeted cells: MSCs, osteoprogenitors cells, osteoblasts, chondrocytes
Potent mitogenic and chemotactic for bone forming cells, chemotactic for macrophages
Expressed from very early stages throughout fracture healing

PDGF
Source: degranulating platelets, macrophages, monocytes (during the granulation stage)
and endothelial cells, osteoblasts (at later stages)
Targeted cells: mesenchymal and inflammatory cells, osteoblasts
Mitogenic for mesenchymal cells and osteoblasts, chemotactic for inflammatory and mesenchymal cells
Released at very early stages of fracture healing

BMPs
Source: osteoprogenitors and mesenchymal cells, osteoblasts, bone extracellular matrix and chondrocytes
Targeted cells: mesenchymal and osteoprogenitor cells, osteoblasts
Differentiation of undifferentiated mesenchymal cells into chondrocytes and osteoblasts
and osteoprogenitors into osteoblasts
Various temporal expression patterns (Table 2)

FGFs
Source: monocytes, macrophages, mesenchymal cells, osteoblasts, chondrocytes
Targeted cells: mesenchymal and epithelial cells, osteoblasts and chondrocytes
Angiogenic and mitogenic for mesenchymal and epithelial cells, osteoblasts, chondrocytes
a-FGF mainly effects chondrocyte proliferation, b-FGF (more potent) involved in chondrocytes
maturation and bone resorption
Expressed from the early stages until osteoblasts formation

IGFs
Source: bone matrix, endothelial and mesenchymal cells (in granulation stage) and osteoblasts
and non-hyperthrophic chondrocytes (in bone and cartilage formation)
Targeted cells: MSCs, endothelial cells, osteoblasts, chondrocytes
IGF-I: mesenchymal and osteoprogenitor cells recruitment and proliferation, expressed
throughout fracture healing
IGF-II: cell proliferation and protein synthesis during endochondral ossification

Metalloproteinases
Source: the extracellular matrix
Degradation of the cartilage and bone allowing the invasion of blood vessels during
the final stages of endochondral ossification and bone remodelling

VEGFs
Potent stimulators of endothelial cell proliferation
Expressed during endochondral formation and bone formation

Angiopoietin (1 and 2)
Formation of larger vessel structures, development of co-lateral branches from existing vessels
Expressed from the early stages throughout fracture healing
1396 R. Dimitriou et al.

Figure 1 Schematic summary of the temporal expression patterns of the signalling molecules during fracture healing.
(The dashed line represents a difference of opinion amongst scientists in terms of the timing of expression.)

Bone morphogenetic proteins. Members of the locate into the nucleus and regulate the transcrip-
BMP family are divided into at least four separate tion of target genes (Fig. 2).75 BMPs are pleiotropic
subgroups depending on their primary amino acid morphogens and play a critical role in regulating
sequence. Group one consists of BMP-2 BMP-4, and growth, differentiation, and apoptosis of various
group two includes BMP-5, -6, and -7. The third cell types, including osteoblasts, chondroblasts,
group includes GDF-5 (or BMP-14), GDF-6 (or BMP- neural cells, and epithelial cells.67
13) and GDF-7 (or BMP-12), and finally group four Furthermore, it has been shown that BMP hetero-
includes BMP-3 (or osteogenin) and GDF-10 (or BMP- dimers, such as BMP-4/-7 and BMP-2/-7 have an
3b).67 BMP-1 is not a member of the TGF-b super- enhanced osteoinductive activity regulating more
family and it may play a role in modulating BMP efficiently differentiation and proliferation of
actions by the proteolysis of BMP antagonists/bind- mesenchymal cells to osteoblasts in vitro and in
ing proteins, such as noggin and chondrin.63 vivo.38
BMPs bind to type II serine/threonine kinase The extracellular matrix comprises the main
receptors which transphosphorylate type-I recep- source of BMPs being produced by osteoprogenitors,
tors.28,57 Subsequently, the Smad intracellular sig- mesenchymal cells, osteoblasts, and chondrocytes
nalling cascade is initiated. The Smad family (Table 1). BMPs induce a sequential cascade of
includes eight members which can be subdivided events for chondro-osteogenesis, including chemo-
into three groups: the signal-transducing receptor- taxis, mesenchymal and osteoprogenitor cells pro-
regulated Smads (R-Smads 1, 2, 3, 5, 8), the com- liferation and differentiation, angiogenesis, and
mon mediator Smad (co-Smad, such as Smad-4), and controlled synthesis of extracellular matrix.63,67
the inhibitory Smads (I-Smads, such as Smad-6 and Their regulatory effect depends upon the type of
Smad-7).37,50 R-Smad 1, 5 and 8 are substrates for the targeted cell, its differentiation stage, the local
BMP receptors and when activated they interact concentration of the ligand as well as the interac-
with Smad-4. These heteromeric complexes trans- tion with other circulating factors.35 Interestingly,
Current concepts of molecular aspects of bone healing 1397

low concentrations of BMPs in vitro favour the dif- of BMPs in fracture healing in the mouse and rat
ferentiation of mesenchymal stem cells into adipo- have shown a variety of osteogenic effects, tem-
cytes.80 poral expressions, and mitogenic capacities
BMPs are closely structurally and functionally (Tables 2 and 3).9,10,20
related, however, each has a unique role as well a In a comprehensive analysis of the osteogenic
distinct temporal expression pattern during the activity of 14 types of BMPs, Cheng et al. suggested
fracture repair process (Fig. 1). Studies of the role an osteogenic hierarchical model of BMPs. BMP-2,

Table 2 Temporal and functional characteristics of members of the TGF-b superfamily observed during fracture
healing in animal models
Member of the TGF-b Time of expression Specific responses in vivo and in vitro
superfamily
GDF-8 Restricted to day 1 20 Potential function as a negative
regulator of skeletal muscle growth 20
BMP-2 Days 1—2110,20 Recruitment of mesenchymal cells
(the earliest gene to be induced and Chondrogenesis
second elevation during osteogenesis) May initiate the fracture healing
cascade and regulate the expression of
other BMPs
BMP-2, -6, -9 may be the most potent to
induce osteoblast lineage-specific
differentiation of MSCs 19
BMP-3, -8 Days 14—2120 (restricted expression Temporal data suggest a role in the
during osteogenesis) regulation of osteogenesis
BMP-4 Transient increased expression in the Involvement in the formation of callus
surrounding soft tissues 6 h to day 5 9 at a very early stage in the healing
process
Days 14—21 20 In vitro: BMP-3 and -4 stimulate the
migration of human blood monocytes 63
Through out fracture healing 10
BMP-7 Days 14—21 20 Regulatory role in both types of
ossification
From the early stages In vitro: stimulation of relative mature
of fracture healing 9 osteoblasts 19
GDF-10, BMP-5, -6 Days 3—21 20 Regulatory role in both types of
ossification
BMP-6 may initiate chondrocyte
maturation 20
GDF-5, 1 Day 7 (maximal) to day 1420 GDF-5 an exclusive involvement in
(restricted expression chondrogenesis is suggested
during chondrogenic phase)
GDF-1 at extremely low levels Stimulation of mesenchymal aggregation
and induction of angiogenesis through
chemotaxis of endothelial cells and
degradation of matrix proteins
GDF-3, GDF-6, 9 No detectable levels within GDF-6 may be expressed only in articular
the fracture callus 20 cartilage20 and with GDF-5, 7 more
efficiently induce cartilage and
tendon-like structures in vivo 28
TGF-b1, -b2, -b3 Days 1—21 20 Potent chemotactic for bone forming
cells and macrophages
Days 3—14 20 Proliferation of undifferentiated
mesenchymal and osteoprogenitor cells,
osteoblasts, chondrocytes
Days 3—21 20
1398 R. Dimitriou et al.

BMPs may also stimulate the synthesis and secre-


tion of other bone and angiogenic growth factors
such as insulin-like growth factor (IGF) and vascular-
endothelial growth factor (VEGF), respectively.22
They may also stimulate bone formation by
directly activating endothelial cells to stimulate
angiogenesis.59,79
Recent studies have showed that the expression
of the BMP antagonists, most importantly noggin,72
which blocks BMP-2 interaction with its receptor,29
also play an important role in fracture healing reg-
ulation.36 It has been suggested that the noggin/
BMP-4 balance could be an important factor in the
regulation of callus formation.85
Transforming growth factor beta. Five isoforms
of this group have been isolated.42 Platelets release
TGF-b during the initial inflammatory phase of bone
healing and therefore this factor may be involved in
Figure 2 BMP-receptor interaction and the intracellular the initiation of callus formation.8,9 TGF-b is also
signalling pathway. produced by osteoblasts and chondrocytes, and is
stored in the bone matrix (Table 1).46 Its effect is
-6, and -9 may be the most potent to induce osteo- exerted via type-I and type-II serine/threonine
blast differentiation of mesenchymal progenitor kinase receptors, activating the Smad pathway
cells, whilst most BMPs (except BMP-3 and -13) (Smad 2 and 3).37
can promote the terminal differentiation of com- TGF-b is a potent chemotactic stimulator of
mitted osteoblastic precursors and osteoblasts.19 mesenchymal stem cells and it enhances prolifera-

20,33,43
Table 3 Timing of cellular events and expression of signalling molecules during murine fracture healing
Day 1 Haematoma formation, inflammation Cytokines: IL-1, IL-6, TNF-a released
by inflammatory cells
Recruitment of mesenchymal cells PDGF, TFG-beta released from
degranulating platelets
Osteogenic differentiation of MSCs from bone marrow BMP-2 expression and restricted to
day 1 expression of GDF-8
Day 3 MSCs proliferation begins Decline of cytokines levels
Proliferation and differentiation of Expression of TGF-b2, -b3, GDF-10,
preosteoblasts and osteoblasts in regions BMP-5, -6
of intramembranous ossification
Angiogenesis begins Angiopoietin-1 is induced
Day 7 Peak of cell proliferation in intramembranous Peak of TGF-b2 and -b3 expression
ossification between days 7 and 10
Chondrogenesis and endochondral Expression of GDF-5 and probably
ossification begins (days 9—14 maturation GDF-1
of chondrocytes)
Day 14 Cessation of cell proliferation in intramembranous Decreased levels of expression for
ossification, but osteoblastic activity continues TGF-b2, GDF-5, and probably GDF-1
Mineralization of the soft callus, cartilage resorption, Expression of BMP-3, -4, -7, and -8
and woven bone formation
Neo-angiogenesis which infiltrates along VEGFs expression
new mesenchymal cells
Phase of most active osteogenesis until day 21 Second increase of IL-1 and TNF-a
which continues during bone
remodelling
Day 21 Woven bone remodelled and subsequently Decreased expression of TGF-b1
replaced by lamellar bone and TGF-b3, GDF-10, and BMPs (2—8)
Current concepts of molecular aspects of bone healing 1399

tion of MSCs, preosteoblasts, chondrocytes and esis (Fig. 1). a-FGF mainly effects chondrocyte
osteoblasts.46 It also induces the production of proliferation and is probably important for chondro-
extracellular proteins such as collagen, proteogly- cyte maturation, whilst b-FGF is expressed by osteo-
cans, osteopontin, osteonectin, and alkaline phos- blasts and is generally more potent than a-FGF.46 In
phatase.68 Its main role is thought to be during a canine tibial osteotomy model, a single injection
chondrogenesis and endochondral bone formation of FGF-2 was associated with an early increase in
(Table 2).7 TGF-b may also initiate signalling for BMP callus size.55
synthesis by the osteoprogenitor cells,9 while it may
inhibit osteoclastic activation and promote osteo- Insulin-like growth factors (IGFs). The sources of
clasts apoptosis.51 Recently, it has been suggested IGF-I (or somatomedin-C) and IGF-II (or skeletal
that TGF-b2 and possibly TGF-b3 play more impor- growth factor) are the bone matrix, endothelial
tant roles in fracture healing than TGF-b1, as their cells, osteoblasts and chondrocytes. The serum con-
expression peak during chondrogenesis. On the centration of IGF-I is mainly regulated by the growth
other hand, TGF-b1 has a high basal level of expres- hormone (Table 1).46,73 The biological actions of the
sion in unfractured diaphyseal bone, and the level of IGFs are modulated in a cell-specific manner by IGF-
expression remains constant throughout the frac- binding proteins (IGFBPs).71
ture healing process, indicating that TGF-b2 and IGF-I promotes bone matrix formation (type I
TGF-b3 may play more important roles in fracture collagen and non-collagenous matrix proteins) by
healing than TGF-b1 (Fig. 1).20 fully differentiated osteoblasts15 and is more potent
Although various studies showed that TGF-b than IGF-II.46 IGF-II acts at a later stage of endo-
enhances cellular proliferation, its osteoinductive chondral bone formation and stimulates type I col-
potential seems limited and concern for its unfore- lagen production, cartilage matrix synthesis, and
seen side effects has been expressed.46 Therefore, cellular proliferation (Fig. 1).62 The findings from a
its therapeutic potential to enhance bone repair number of animal studies assessing the influence of
seems to be limited. IGF on skeletal repair have varied and therefore
further studies are required.7
Platelet-derived growth factor (PDGF). PDGF is a
homo- or heterodimeric polypeptide consisting of A Metalloproteinases and angiogenic factors
and B chains. PDGF effect is exerted via receptors Optimal bone regeneration requires adequate blood
that have tyrosine kinase activity. IL-1, TNF-a, and flow. During the final stages of endochondral ossifi-
TGF-b1 affect PDGF’s binding.73 It is synthesized by cation as well as during remodelling phase, specific
platelets, monocytes, macrophages, endothelial matrix metallopoteinases degrade cartilage and
cells, and osteoblasts and it is a potent mitogen bone, allowing the invasion of blood vessels
for cells of mesenchymal origin (Table 1).3 (Table 1).33
PDGF is released by platelets during the early Two separate pathways are believed to regulate
phases of fracture healing and it is a potent chemo- angiogenesis: a vascular-endothelial growth factor-
tactic stimulator for inflammatory cells and a major dependent pathway and an angiopoietin-dependent
proliferative and migratory stimulus for MSCs and pathway.33 It is speculated that both pathways are
osteoblasts (Fig. 1).46 Nash et al. showed an functional during fracture repair. VEGFs are essential
increased callus density and volume in tibial osteo- mediators of neo-angiogenesis and endothelial-cell
tomies in rabbits treated with PDGF.56 However, at specific mitogens.26 Whereas, angiopoietin 1 and 2 are
present, its therapeutic potential remains unclear. regulatory vascular morphogenetic molecules related
to the formation of larger vessel and development of
Fibroblast growth factor (FGFs). The family of FGFs co-lateral branches from existing vessels (Fig. 1).
consists of nine structurally related polypeptides. However, their contribution in bone repair is not as
The acidic and basic FGFs are the most abundant well understood (Table 1).33 Street et al. showed that
FGFs in normal adult tissue.81 Their action is exerted fracture repair was enhanced by the exogenous
by binding to tyrosine kinase receptors.83 administration of VEGF.74 Recent studies have also
During bone healing, FGFs are synthesized by shown that BMPs stimulate the expression of VEGF
monocytes, macrophages, mesenchymal cells, by osteoblasts and osteoblast like cells.22,59,84
osteoblasts and chondrocytes (Table 1). FGFs pro-
mote growth and differentiation of a variety of cells
such as fibroblasts, myocytes, osteoblasts, and The role of mesenchymal stem cells
chondrocytes. FGFs are identified during the early
stages of fracture healing and they play a critical MSCs are undifferentiated cells capable of extensive
role in angiogenesis and mesenchymal cell mitogen- replication without differentiation.13 MSCs have the
1400 R. Dimitriou et al.

potential to commit and differentiate along multi- millimetres from the fracture site and the region of
ple cell lineages. They give rise to those cells that the bone marrow with high cellular density.23 Within
form mesenchymal tissues, including bone, carti- the first 24 h of the fracture, the latter cells begin to
lage, tendon, muscle, ligament, and marrow stroma differentiate into an osteoblastic phenotype. By day
and fat.13,60 3, osteoblasts from the cortex and committed osteo-
During fracture healing, potential sources of stem progenitors derived from the periosteal cambium
cells are the bone marrow, the granulation tissue, divide and differentiate, forming woven bone (hard
the deep layer of the periosteum, the endosteum, callus). Their proliferation peaks by days 7 and 10,
and the surroundings soft tissues.23,33,53 Also, peri- and it ceases by day 14 while the osteoblastic
vascular mesenchymal stem cells that exist in blood activity continues.7 In a rat fracture healing model,
vessels walls contribute to fracture healing.11 The BMP-2, -4, and -7 showed increased levels of expres-
primary tissue source of MSCs is the periosteum53 sion during the early stages of intramembranous
and studies from Buckwalter et al. showed that the ossification.9
capacity for fracture callus development is dimin-
ished if the periosteum is removed.14 Endochondral bone formation
In regions that are mechanically less stable, endo-
Sequence of events chondral bone formation occurs. This type of ossi-
fication mainly occurs at the adjacent to the
Fracture healing, like all other repair responses, is fracture site periosteum and enhanced by the soft
initiated through the induction of an immune tissues around the fracture site. During this process,
response.24 During this initial stage, a haematoma MSCs are recruited and begin to proliferate by day 3
is formed and inflammation occurs. The major after fracture. Their subsequent differentiation into
players at this initial inflammatory phase include chondroblasts (chondrogenesis) and the prolifera-
cytokines, platelets, BMPs and MSCs. tion of these new chondrocytes occur from days 7 to
IL-1, IL-6 and TNF-a secreted by inflammatory 21, resulting to soft callus formation. These cells
cells have a chemotactic effect on other inflamma- synthesize and secrete cartilage-specific matrix,
tory cells and on the recruitment of mesenchymal including type II collagen and proteoglycans and
cells.43 Cho et al. reported a peak in expression of once firm mechanical stability is established, the
IL-1 and -6 one day after fracture followed by a rapid cartilage undergoes hypertrophy and mineralization
decline until day 3 to near undetectable levels.20 At in a spatially organized manner.7 As vasculature
the same time, platelets, activated by thrombin and begins to invade, the calcifying hypertrophic chon-
subendothelial collagen, release PDGF and TGF-b, drocytes are being removed by chondroclasts and
which play a role on the initiation of fracture woven bone formation occurs after the recruitment
repair.8 These factors induce mesenchymal cell and osteogenic differentiation of new MSCs.23 Lee
migration, activation and proliferation, angiogen- et al. hypothesised that this stage of endochondral
esis, chemotaxis of acute inflammatory cells and ossification is the final stage of a genetically pro-
further aggregation of platelets. Simultaneously, grammed process which results to apoptotic chon-
BMPs not only are released from the bone matrix drocyte death.45 Eventually, this soft callus is
but also are expressed by recruited primary replaced by woven marrow-filled bone, which
mesenchymal cells.9 During the subsequent days, undergoes significant remodelling to become
MSCs proliferate and differentiate into a chondro- weight-bearing bone following the pathway
genic or osteogenic lineage.69 During this early observed in the growth plate. Recently, Cho et al.
phase of events, angiogenesis also takes place demonstrated the different temporal patterns for
and this is a prerequisite for further progression members of the TGF-b superfamily during murine
of the regeneration cascade. The vascular ingrowth fracture healing, suggesting a potentially unique
into the developing callus is regulated by FGF, role of each BMP (Table 3).20
VEGF and angiopoietin 1 and 2.33,46 Angiopoietic The stimulation of undifferentiated MSCs and
1 has been suggested to be induced during the osteoprogenitors cells to differentiate into osteo-
initial periods of fracture healing whereas VEGF blasts (osteoinduction)73 is a morphogenetic cas-
later on mainly during endochondral and bone cade involving discrete cellular transitions. It is
formation.33 predominantly regulated by the complex interac-
tions that take place between the multiple local
Intramembranous bone formation (paracrine and autocrine) signals that are pre-
The source of the cells that contribute to intramem- sented to mesenchymal stem cells in their natural
branous bone formation appears to be the under- environment (Table 1).31 Further research is
lying cortical bone, the periosteum within a few required in order to elucidate the role of each
Current concepts of molecular aspects of bone healing 1401

factor at each discrete stage of fracture healing as healing process. Bone tissue engineering, combining
well as their combined functioning to promote its the application of the principles of orthopaedic
various stages. surgery with basic science and engineering, has
Although the cellular events during fracture heal- been heralded as an alternative when bone regen-
ing seem to be predominantly regulated by local eration is required to replace or restore the function
factors and cytokines, systemic hormones (para- of traumatised, damaged, or lost bone.66 In essence,
thyroid/thyroid hormone, the growth hormone, tissue engineering aims to combine progenitor cells,
the 1,25 dihydroxyvitamin D and the sex steroids) such as MSCs, or mature cells (for osteogenesis) with
may also modulate these events.51 biocompatible materials or scaffolds (for osteocon-
duction), with appropriate growth factors (for
osteoinduction), in order to generate and maintain
Current concepts of systemic bone.66,78 Although this new strategy is still in its
enhancement of fracture healing infancy, its clinical application offers great poten-
tials for the treatment of conditions requiring bone
Parathyroid hormone (PTH) repair.
A promising technology in the field of bone tissue
Contrary to the assumption that PTH has a catabolic engineering is the application of gene therapy as a
effect on the skeleton, intermittent exposure sti- method of growth factor delivery for the clinical
mulates osteoblasts and results in increased bone management of orthopaedic disorders, including
formation in rats. It has been shown that low-dose bone healing.18 It involves the transfer of genetic
human PTH (1—34) enhances callus formation by material into targeted cell’s genome, and thus
stimulating the early proliferation and differentia- allowing the expression of bioactive factors from
tion of osteoprogenitor cells, increasing the produc- the cells themselves and for longer periods of time.
tion of bone matrix proteins and enhancing The gene transfer can be performed using a viral
osteoclastogenesis during callus remodelling. PTH (transfection) or a non-viral (transduction) vector,
effect is likely to be mediated by osteoblastic activ- by either an in vivo or ex vivo gene-transfer strat-
ity, as PTH receptors are found on osteoblast mem- egy.18,46 With the in vivo technique, a technically
branes.1,54 easier method, the genetic material is transferred
directly into the host. However, there are safety
Growth hormone (GH) concerns with this strategy. The indirect ex vivo
technique involves the collection of cells by tissue
Growth hormone is a systemic hormone and its harvest and their genetic modification in vitro
effect on the skeleton is mediated by IGF-1 (known before transfer back into the host. It is a technically
as somatomedin-C) which promotes bone matrix more demanding but safer method, as it allows
formation (type I collagen and non-collagenous testing of the cells for any abnormal behaviour
matrix proteins) by fully differentiated osteoblasts. before re-implantation as well as selection of those
Systemically administration of GH in a rat tibial with the greatest gene expression.18
diaphyseal fracture model during the first 3 weeks Gene therapy has been used to promote fracture
of healing increased callus formation and enhanced repair through the expression of BMP-26,47 and -470
fracture strength.2 Furthermore, in a fracture heal- in animal studies. Although promising, issues of its
ing and distraction osteogenesis model in Yucatan biosafety and efficacy need to be answered before
micropigs, administration of homologous recombi- human trials take place.
nant porcine GH led to an increase in serum IGF-1,
stimulation of fracture healing and acceleration of Muscle stem cells
ossification of bone regenerate in distraction osteo-
genesis.5 Since 1965, when Urist first noted the ectopic bone
formation after demineralized bone matrix implan-
tation into skeletal muscle,76,77 subsequent studies
Future directions have reported similar observations. This phenom-
enon of osteoinduction has been attributed to the
Tissue engineering mitogenic effects of BMP-2 (in vitro)40 and BMP-3 (in
vivo)41 on cells in muscle In skeletal muscle,
Currently, as the molecular and cellular events researchers have identified adult stem cells that
during the fracture healing cascade are becoming can differentiate into cells of different line-
gradually more understood, new strategies are ages.16,58 The most well-known muscle progenitor
investigated in order to promote or facilitate the cells, termed satellite cells, are the primary source
1402 R. Dimitriou et al.

of the myoblasts, but they can also exhibit osteo- Currently, recombinant BMP-2 and -7 are com-
genic and adipogenic differentiation.4 Recently, a mercially available and, although the research is
population of stem cells that appears to be distinct ongoing, they are considered adjuncts in the sur-
from the satellite cells has also been discovered: geon’s armamentarium for the treatment of clini-
the muscle-derived stem cells (MDSCs). These cells cally challenging situations.
have the ability to differentiate into multiple
lineages including osteogenic and haematopoietic
lines.21,39 Conclusion
These muscle-based progenitor cells possess a
therapeutic potential for tissue repair and regen- Fracture healing is a complex physiological process
eration applications in various musculoskeletal as which involves a well orchestrated series of biolo-
well as cardiac muscle disorders either as a source of gical events. Whilst our knowledge has vastly
inducible progenitor cells or as gene delivery vehi- expanded, with the increasing understanding of
cles.21 For bone formation and healing in particular, the multiple factors and complex pathways
MDSCs genetically engineered to express BMP-252 involved, a lot of new developments are anticipated
and both BMP-4 and VEGF58 have been shown to be in the years to come. It is hoped that many bone
capable of stimulating osteogenesis and angiogen- disease processes secondary to trauma, aging and
esis, respectively. metabolic disorders will be successfully treated
with novel treatment protocols.

Clinical applications in trauma


Acknowledgements
Approximately 5—10%, of the 6.2 million fractures
occurring annually in the United States, are asso- Rozalia Dimitriou is a research fellow supported via
ciated with impaired healing including delayed a grant (03-943) by the AO foundation.
healing or non-union.61
Various animal studies and clinical trials in
humans have been performed and demonstrated References
the potential use of several of the biological factors
described in bone regeneration and skeletal repair, 1. Andreassen TT, Ejersted C, Oxlund H. Intermittent parathyr-
with the BMPs to be the most promising. oid hormone (1—34) treatment increases callus formation
Riedel and Valentin-Opran were the first to report and mechanical strength of healing rat fractures. J Bone
Miner Res 1999;14:960—8.
preliminary results from the use of BMP-2 to aug-
2. Andreassen TT, Oxlund H. Local anabolic effects of growth
ment the treatment of open tibial fractures.64 hormone on intact bone and healing fractures in rats. Calcif
Govender et al. in a large prospective randomized Tissue Int 2003;73:258—64.
controlled multi-center trial evaluated the effects 3. Andrew JG, Hoyland JA, Freemont AJ, Marsh DR. Platelet-
of recombinant (rh) BMP-2 on the treatment of open derived growth factor expression in normally healing human
fractures. Bone 1995;16:455—60.
tibial fractures. They reported a higher union rate,
4. Asakura A, Komaki M, Rudnicki M. Muscle satellite cells are
an accelerated time to union, improved wound multipotential stem cells that exhibit myogenic, osteogenic,
healing, reduced infection rate, and fewer second- and adipogenic differentiation. Differentiation 2001;68:
ary invasive interventions in the group of patients 245—53.
treated with rhBMP-2 and IM nail fixation, compared 5. Bail HJ, Kolbeck S, Krummrey G, et al. Systemic application
of growth hormone for enhancement of secondary and
to the group treated with IM nail fixation alone.34
intramembranous fracture healing. Horm Res 2002;58
In 1999, Geesink et al. demonstrated the ability (Suppl. 3):39—42.
of BMP-7 to cause repair of a critical-sized fibular 6. Baltzer AW, Lattermann C, Whalen JD, et al. Genetic
defect in patients undergoing opening wedge high enhancement of fracture repair: healing of an experimental
tibial osteotomy with fibulectomy.30 Friedlaender segmental defect by adenoviral transfer of the BMP-2 gene.
Gene Ther 2000;7:734—9.
et al. in a large prospective randomized controlled 7. Barnes GL, Kostenuik LC, Gerstenfeld LC, Einhorn TA. Growth
and partially blinded multicenter study, they factor regulation of fracture repair. J Bone Miner Res
assessed the efficacy of rhBMP-7 over iliac crest 1999;14:1805—15.
bone graft in the treatment of 122 patients with 8. Bolander ME. Regulation of fracture repair by growth factors.
124 tibial non-unions. The authors concluded that Proc Soc Exp Biol Med 1992;200:165—70.
9. Bostrom MP. Expression of bone morphogenetic proteins in
whilst no statistical difference was noted between
fracture healing. Clin Orthop 1998;355S:116—23.
the two groups, BMP-7 was a safe and effective 10. Bostrom MP, Lane JM, Berberian WS, et al. Immunolocaliza-
alternative to bone graft in the treatment of tibial tion and expression of bone morphogenetic proteins 2 and 4
non-unions.27 in fracture healing. J Orthop Res 1995;13:357—67.
Current concepts of molecular aspects of bone healing 1403

11. Bouletreau PJ, Warren SM, Spector JA, et al. Hypoxia and 32. Gerstenfeld LC, Cho TJ, Kon T, et al. Impaired fracture
VEGF up-regulate BMP-2 mRNA and protein expression in healing in the absence of TNF-alpha signalling: the role of
microvascular endothelial cells: implications for fracture TNF-alpha in endochondral cartilage resorption. J Bone Miner
healing. Plast Reconstr Surg 2002;109:2384—97. Res 2003;18:1584—92.
12. Brighton CT, Hunt RM. Early histological and ultrastructural 33. Gerstenfeld LC, Cullinane DM, Barnes GL, et al. Fracture
changes in medullary fracture callus. J Bone Joint Surg Am healing as a post-natal developmental process: molecular,
1991;73(6):832—47. spatial, and temporal aspects of its regulation. J Cell Bio-
13. Bruder SP, Fink DJ, Caplan AI. Mesenchymal stem cells in bone chem 2003;88:873—84.
development, bone repair, and skeletal regeneration ther- 34. Govender S, Csimma C, Genant HK, et al., BMP-2 Evaluation
apy. J Cell Biochem 1994;56:283—94. in Surgery for Tibial Trauma (BESTT) Study Group. Recombi-
14. Buckwalter JA, Einhorn TA, Marsh LJ. Bone and joint healing. nant human bone morphogenetic protein-2 for treatment of
In: Rockwood Jr CA, Green DP, Bucholz RW, Heckman JD, open tibial fractures: a prospective, controlled, randomized
editors. Fractures in adults. 5th ed., Philadelphia: Lippincott study of four hundred and fifty patients. J Bone Joint Surg
Williams & Wilkins, 2001. [Am] 2002;84-A(12):2123—34.
15. Canalis E. Effect of insulin-like growth factor I on DNA and 35. Groeneveld EHJ, Burger EH. Bone morphogenetic proteins in
protein synthesis in cultured rat calvaria. J Clin Invest human bone regeneration. Eur J Endocrinol 2000;142:9—
1980;66:709—19. 21.
16. Cao B, Huard J. Muscle-derived stem cells. Cell Cycle 36. Groppe J, Greenwald J, Wiater E, et al. Structural basis of
2004;3:104—7. BMP signalling inhibition by Noggin, a novel twelve-mem-
17. Carter DR, Van der Meulen MC, Beaupre GS. Mechanical bered cystine knot protein. J Bone Joint Surg [Am] 2003;85-
factors in bone growth and development. Bone 1996;18S: A(Suppl. 3):52—8.
5—10. 37. Heldin CH, Miyazono K, Ten Dijke P. TGF-beta signalling from
18. Chen Y. Orthopaedic application of gene therapy. J Orthop Sci cell membrane to nucleus through SMAD proteins. Nature
2001;6:199—207. 1997;390:465—71.
19. Cheng H, Jiang W, Phillips FM, et al. Osteogenic activity of the 38. Israel DI, Nove J, Kerns KM, et al. Heterodimeric bone
fourteen types of human bone morphogenetic proteins morphogenetic proteins show enhanced activity in vitro
(BMPs). J Bone Joint Surg [Am] 2003;85-A(8):1544—52. and in vivo. Growth Factors 1996;13:291—300.
20. Cho TJ, Gerstenfeld LC, Einhorn TA. Differential temporal 39. Jankowski RJ, Huard J. Myogenic cellular transplantation and
expression of members of the transforming growth factor regeneration: sorting through progenitor heterogeneity. Pan-
beta superfamily during murine fracture healing. J Bone minerva Med 2004;46:81—91.
Miner Res 2002;17:513—20. 40. Katagiri T, Yamaguchi A, Komaki M, et al. Bone morphoge-
21. Deasy BM, Li Y, Huard J. Tissue engineering with muscle- netic protein-2 converts the differentiation pathway of
derived stem cells. Curr Opin Biotechnol 2004;15:419—23. C2C12 myoblasts into the osteoblast lineage. J Cell Biol
22. Deckers MM, van Bezooijen RL, van der Horst G, et al. Bone 1994;127:1755—66.
morphogenetic proteins stimulate angiogenesis through 41. Khouri RK, Brown DM, Koudsi B, et al. Repair of calvarial
osteoblast-derived vascular endothelial growth factor A. defects with flap tissue: role of bone morphogenetic proteins
Endocrinology 2002;143:1545—53. and competent responding tissues. Plast Reconstr Surg
23. Einhorn TA. The cell and molecular biology of fracture heal- 1996;98:103—9.
ing. Clin Orthop 1998;355S:7—21. 42. Kingsley DM. The TGF superfamily: new members, new recep-
24. Einhorn TA, Majeska RJ, Rush EB, et al. The expression of tors, and new genetic tests of function in different organ-
cytokine activity by fracture callus. J Bone Miner Res isms. Genes Dev 1994;8:133—46.
1995;10:1272—81. 43. Kon T, Cho T, Aizawa T, et al. Expression of osteoprotegerin,
25. Ferguson C, Alpern E, Miclau T, Helms JA. Does adult fracture receptor activator of NF-kB ligand (osteoprotegerin ligand)
repair recapitulate embryonic skeletal formation? Mech Dev and related proinflammatory cytokines during fracture heal-
1999;87:57—66. ing. J Bone Miner Res 2001;16:1004—14.
26. Ferrara N, Davis-Smyth T. The biology of vascular endothelial 44. Le AX, Miclau T, Hu D, Helms JA. Molecular aspects of healing
growth factor. Endocr Rev 1997;18:4—25. in stabilized and non-stabilized fractures. J Orthop Res
27. Friedlaender GE, Perry CR, Cole JD, et al. Osteogenic pro- 2001;19:78—84.
tein-1 (bone morphogenetic protein-7) in the treatment of 45. Lee FYH, Choi YW, Behrens FF, et al. Programmed removal of
tibial nonunions. J Bone Joint Surg [Am] 2001;83(Suppl. chondrocytes during endochondral fracture healing. J Orthop
1):151—8. Res 1998;16:144—50.
28. Fujii M, Takeda K, Imamura T, et al. Roles of bone morpho- 46. Lieberman JR, Daluiski A, Einhorn TA. The role of
genetic protein type I receptors and Smad proteins in osteo- growth factors in the repair of bone. Biology and clinical
blast and chondroblast differentiation. Mol Biol Cell applications. J Bone Joint Surg [Am] 2002;84-A(6):1032—
1999;10:3801—13. 44.
29. Gazzero E, Gnagji V, Canalis E. Bone morphogenetic pro- 47. Lieberman JR, Daluiski A, Stevenson S, et al. The effect of
teins induce the expression of noggin, which limits their regional gene therapy with bone morphogenetic protein-2-
activity in cultured rat osteoblasts. J Clin Invest 1998;102: producing bone-marrow cells on the repair of segmental
2106—14. femoral defects in rats. J Bone Joint Surg [Am] 1999;81:
30. Geesink RGT, Hoefnagels NHM, Bulstra SK. Osteogenic activ- 905—17.
ity of OP-1 bone morphogenetic protein (BMP-7) in a human 48. Massague J, Attisano L, Wrana JL. The TGF-beta family and its
fibular defect. J Bone Joint Surg [Br] 1999;81B:710—8. composite receptors. Trends Cell Biol 1994;4:172—8.
31. Gerstenfeld LC, Barnes GL, Shea CM, Einhorn TA. Osteogenic 49. McKibbin B. The biology of fracture healing in long bones. J
differentiation is selectively promoted by morphogenetic Bone Joint Surg [Br] 1978;60-B(2):150—62.
signals from chondrocytes and synergized by a nutrient 50. Miyazono K. Signal transduction by bone morphogenetic
rich growth environment. Connect Tissue Res 2003;44 protein receptors: functional roles of Smad proteins. Bone
(Suppl. 1):85—91. 1999;25:91—3.
1404 R. Dimitriou et al.

51. Mundy GR. Regulation of bone formation by bone morpho- 69. Shea CM, Edgar CM, Einhorn TA, Gerstenfeld L. BMP treat-
genetic proteins and other growth factors. Clin Orthop ment of C3H10T1/2 mesenchymal stem cells induces both
1996;324:24—8. chondrogenesis and osteogenesis. J Cell Biochem 2003;90:
52. Musgrave DS, Pruchnic R, Bosch P, et al. Human skeletal 1112—27.
muscle cells in ex vivo gene therapy to deliver bone mor- 70. Shen HC, Peng H, Usas A, et al. Structural and functional
phogenetic protein-2. J Bone Joint Surg [Br] 2002;84:120—7. healing of critical-size segmental bone defects by transduced
53. Nakahara H, Bruder SP, Haynesworth SE, et al. Bone and muscle-derived cells expressing BMP-4. J Gene Med
cartilage formation in diffusion chambers by subcultured 2004;6:984—91.
cells derived from the periosteum. Bone 1990;11:181—8. 71. Shimasaki S, Ling N. Identification and molecular character-
54. Nakajima A, Shimoji N, Shiomi K, et al. Mechanisms for the ization of insulin-like growth factor binding proteins (IGFBP-
enhancement of fracture healing in rats treated with inter- 1, -2, -3, -4, -5 and -6). Prog Growth Factor Res 1991;3:243—
mittent low-dose human parathyroid hormone (1—34). J Bone 66.
Miner Res 2002;17:2038—47. 72. Smith WC, Harland RM. Expression cloning of noggin, a new
55. Nakamura T, Hara Y, Tagawa M, et al. Recombinant human dorsalizing factor localized to the Spemann organizer in
basic fibroblast growth factor accelerates fracture healing by Xenopus embryos. Cell 1992;70:829—40.
enhancing callus remodelling in experimental dog tibial 73. Solheim E. Growth factors in bone. Int Orthop 1998;22:410—
fractures. J Bone Miner Res 1998;13:942—9. 6.
56. Nash TJ, Howlett CR, Martin C, et al. Effect of platelet- 74. Street J, Bao M, deGuzman L, et al. Vascular endothelial
derived growth factor on tibial osteotomies in rabbits. Bone growth factor stimulates bone repair by promoting angiogen-
1994;15:203—8. esis and bone turnover. Proc Natl Acad Sci USA 2002;99:9656—
57. Onishi T, Ishidou Y, Nagamine T, et al. Distinct and overlapping 61.
patterns of localization of bone morphogenetic protein (BMP) 75. Ten Dijke P, Fu J, Schaap P, Roelen BA. Signal transduction of
family members and a BMP type II receptor during fracture bone morphogenetic proteins in osteoblast differentiation. J
healing in rats. Bone 1998;22:605—12. Bone Joint Surg [Am] 2003;85(Suppl. 3):34—8.
58. Peng H, Huard J. Muscle-derived stem cells for musculoske- 76. Urist MR. Bone: formation by autoinduction. Science
letal tissue regeneration and repair. Transpl Immunol 1965;150:893—9.
2004;12:311—9. 77. Urist MR, O’Connor BT, Burwell RG. Bone graft, derivatives
59. Peng H, Wright V, Usas A, et al. Synergistic enhancement of .and substitutes. Oxford: Butterworth—Heinemann Ltd., 1994
bone formation and healing by stem cell-expressed VEGF and 78. Vaccaro AR. The role of the osteoconductive scaffold
bone morphogenetic protein-4. J Clin Invest 2002;110: in synthetic bone graft. Orthopaedics 2002;25(Suppl. 5):
751—9. 571—8.
60. Pittenger MF, Mackay AM, Beck SC, et al. Multilineage poten- 79. Valdimarsdottir G, Goumans MJ, Rosendahl A, et al. Stimula-
tial of adult human mesenchymal stem cells. Science tion of Id1 expression by bone morphogenetic protein is
1999;284:143—7. sufficient and necessary for bone morphogenetic protein—
61. Praemer A, Furner S, Rice DP. Musculoskeletal injuries. In induced activation of endothelial cells. Circulation 2002;106:
Musculoskeletal conditions in the United States. Park Ridge, 2263—70.
IL: American Academy of Orthopaedic Surgeons; 1992. p. 85— 80. Wang EA, Israel DI, Kelly S, Luxenberg DP. Bone morphoge-
124. netic protein-2 causes commitment and differentiation in
62. Prisell PT, Edwall D, Lindblad JB, et al. Expression of insulin- C3H10T1/2 and 3T3 cells. Growth Factors 1993;9:57—71.
like growth factors during bone induction in rat. Calcif Tissue 81. Wang JS. Basic fibroblast growth factor for stimulation of
Int 1993;53:201—5. bone formation in osteoinductive or conductive implants.
63. Reddi AH. Bone morphogenetic proteins: from basic science Acta Orthop Scand 1996;269:1—33.
to clinical applications. J Bone Joint Surg [Am] 2001;83- 82. Wozney JM, Rosen V, Celeste AJ, et al. Novel regulators of
A(Suppl. 1):1—6. bone formation: molecular clones and activities. Science
64. Riedel GE, Valentin-Opran A. Clinical evaluation of rhBMP-2/ 1988;242:1528—34.
ACS in orthopaedic trauma: a progress report. Orthopaedics 83. Xu X, Weinstein M, Li C, Deng C. Fibroblast growth factor
1999;22:663—5. receptors (FGFRs) and their roles in limb development. Cell
65. Rockwood Jr CA, Green DP, Bucholz RW, Heckman JD. Frac- Tissue Res 1999;296:33—43.
tures in adults, 4th ed., Philadelphia: Lippincott, 1996. 84. Yeh LC, Lee JC. Osteogenic protein-1 increases gene expres-
66. Rose FR, Oreffo RO. Bone tissue engineering: hope vs hype. sion of vascular endothelial growth factor in primary cultures
Biochem Biophys Res Commun 2002;292:1—7. of fetal rat calvaria cells. Mol Cell Endocrinol 1999;153:113—
67. Sakou T. Bone morphogenetic proteins: from basic studies to 24.
clinical approaches. Bone 1998;22:591—603. 85. Yoshimura Y, Nomura S, Kawasaki S, et al. Colocalization of
68. Sandberg MM, Hannu TA, Vuorio EI. Gene expression during noggin and bone morphogenetic protein-4 during fracture
bone repair. Clin Orthop 1993;289:292—312. healing. J Bone Miner Res 2001;16:876—84.

You might also like