+ All Categories
Home > Documents > Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair...

Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair...

Date post: 14-Mar-2018
Category:
Upload: vodien
View: 217 times
Download: 4 times
Share this document with a friend
100
Possible Role of Osteoblasts in Regulating the Initiation of Endochondral Repair Process during Fracture Healing by Yasha Amani Andabili A thesis submitted in conformity with the requirements for the degree of Master of Science Department of Laboratory Medicine and Pathobiology University of Toronto © Copyright by Yasha Amani Andabili, 2012
Transcript
Page 1: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

Possible Role of Osteoblasts in Regulating the Initiation of Endochondral Repair Process during Fracture Healing

by

Yasha Amani Andabili

A thesis submitted in conformity with the requirements for the degree of Master of Science

Department of Laboratory Medicine and Pathobiology University of Toronto

© Copyright by Yasha Amani Andabili, 2012

Page 2: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

ii

Possible Role of Osteoblasts in Regulating the Initiation of

Endochondral Repair Process during Fracture Healing

Yasha Amani Andabili

Master of Science

Department of Laboratory Medicine and Pathobiology University of Toronto

2012

Abstract

Fracture repair is a regenerative event that involves the precise coordination of a variety

of cells for successful healing process. Within the microstructure hierarchy of bone repair, the

predominant cells involved include the chondrocytes, osteocytes, osteoblasts, and osteoclasts.

Although the role of osteoblasts during fracture healing has been previously shown, their role

during the initiation phase of endochondral fracture repair remains unclear. In order to study

the role of osteoblasts during fracture repair, we used a transgenic mouse model expressing the

herpes simplex virus thymidine kinase gene in early differentiating osteoblasts, which allows

conditional ablation of cells in osteoblastic lineage upon treatment with the Gancicolvir drug.

Results from this study suggest that not only are osteoblasts required in later stages of fracture

repair as the medium for bone synthesis, and osteoclast activation during bone remodelling, but

could also be required for the initiation and advancement of the endochondral ossification

process.

Page 3: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

iii

Acknowledgments

First and foremost, I would like to demonstrate my gratitude to a cast of people without

whom the completion of this project would not have been possible. I want to thank my

supervisor Dr. Benjamin Alman for his advice, guidance and infinite patience over the course of

my masters program. It has been a very interesting journey and a wonderful experience working

in his lab, especially for being given the opportunity to make a contribution in the field of bone

research. Thank you for your belief and support.

I want to thank all the past and present members of my advisory committee, Dr. Freda

Miller, Dr. Marc Grynpas and Dr. Jane Aubin, for their helpful advice, useful guidance and

constructive discussions. I have had the benefit of learning from your questions, which have

helped me mature as a scientist and allowed me test my abilities and limits. I specially want to

express my gratitude to Dr. Freda Miller for her efforts and support over the years, both as a co-

supervisor and a member of my committee. Thank you.

I also want to thank all the past and present members of Dr. Alman and Dr. Miller’s

laboratories who have helped me grow as a scientist, forge friendships, and most importantly for

their moral support and encourangement, thought-provoking conversations, technical help and

sharing of their expertise and knowlege. A special thanks to Dr. Saeid Amini Nik and Dr. Harry

Elsholtz for their guidance, encouragement and mentorship over the years as well as their belief

in me as a student.

This work was carried out in the laboratories of Dr. Benjamin Alman, Dr. Freda

Miller/Dr. David Kaplan. Special thanks to the laboratory of Dr. Marc Grynpas for making

available the equipments for histomorphometric analysis as well as their technical support and

knowledge. This work was done at the Sickkids division of the MaRS research facility and was

partially funded by a fellowship provided by the University of Toronto.

Finally I want to thank my family for their love and encourangment as well as their moral

support and unrelenting belief in my ability to grow and mature as a scientist.

Page 4: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

iv

Table of Contents

Contents 

Acknowledgments ........................................................................................................................... ii 

Table of Contents ........................................................................................................................... iv 

List of Figures ............................................................................................................................... vi 

List of Abbreviations .................................................................................................................. vii 

Chapter 1 General Introduction .................................................................................................. 1 

1.1  Bone Formation ................................................................................................................. 1 

1.1.1  Endochondral Ossification ................................................................................... 2 

1.1.2  Intramembranous Ossification ............................................................................ 3 

1.1.3  Fracture Repair ..................................................................................................... 4 

1.2  Skeletal Cell Types .......................................................................................................... 10 

1.2.1  Chondrocytes ....................................................................................................... 10 

1.2.2  Osteoclasts ........................................................................................................... 11 

1.2.3  Osteoblasts ........................................................................................................... 14 

1.2.4  Osteocytes ............................................................................................................ 17 

1.3  Osteogenic Proteins ......................................................................................................... 18 

1.3.1  Vascularization in Successful Bone Healing ..................................................... 18 

1.3.2  Matrix Metalloproteinases ................................................................................. 20 

1.3.3  Bone Morphogenetic Proteins ............................................................................ 23 

1.3.4  Transforming Growth Factor Beta ................................................................... 24 

1.3.5  Wnt Signalling and Bone Formation ................................................................. 25 

1.4  Thymidine Kinase/Ganciclovir System ......................................................................... 27 

References .................................................................................................................................... 34 

Page 5: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

v

Chapter 2 Possible Role of Osteoblasts in Regulating the Initiation of Endochondral Repair Process during Fracture Healing ............................................................................. 60 

2  Summary ................................................................................................................................. 60 

2.1  Introduction ..................................................................................................................... 60 

2.2  Methods and Materials ................................................................................................... 63 

2.2.1  Generation of fractures: ........................................................................................ 63 

2.2.2  Mechanism of action of GCV: .............................................................................. 63 

2.2.3  Real-time PCR: ..................................................................................................... 64 

2.2.4  Staining method – Safranin-O/Haematoxylin-Eosin: ........................................... 64 

2.2.5  Histomorphometric, Cell quantification and X-ray analysis: ............................... 64 

2.2.6  Collagen type X and TRAP staining: .................................................................... 65 

2.2.7  Statistical Analysis: ............................................................................................... 65 

2.3  Results .............................................................................................................................. 65 

2.3.1  Pretreatment of DTK transgenic mice with GCV leads to ablation of osteoblasts ............................................................................................................ 65 

2.3.2  Osteoblast depletion delays initiation of endochondral bone repair .............. 66 

2.3.3  Continuous ablation of osteoblasts hinders the progression of endochondral ossification past the soft callus stage ......................................... 67 

2.4  Discussion ......................................................................................................................... 70 

2.5  Figures .............................................................................................................................. 73 

References .................................................................................................................................... 84 

Chapter 3 Summary and Conclusions ...................................................................................... 87 

Chapter 4 Future Directions ...................................................................................................... 88 

Appendix ...................................................................................................................................... 90 

Acknowledgements ..................................................................................................................... 92 

Page 6: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

vi

List of Figures

Figure 1: Genetic Control of Skeletal Cell Type Differentiation.

Figure 2: Bone Formation Occurs Through Two Different Modes of Ossification Processes.

Figure 3: Model of Fracture Healing Stages and Cellular Participants Involved During the Process of Repair.

Figure 4: Continuous GCV Treatment Leads to Non-union of the Fractured Bone.

Figure 5: In Vivo Experimental Design for Studying the Role of Osteoblasts During Fracture Repair Process.

Figure 6: GCV Pretreatment is Effective in Ablating Pre-existing Osteoblasts.

Figure 7: GCV Treatment of the DTK Transgenic Mice Leads to a Decrease in Bone-Lining Osteoblast Population.

Figure 8: Continuous GCV Treatment Leads to a Delay in Initiation and Progression of Endochondral Ossification Process.

Figure 9: Matrix Metalloproteinase 13 Gene Expression is Reduced in Absence of Osteoblasts.

Figure 10: Osteocytes are Unaffected by GCV Treatment.

Figure 11: Upon GCV Withdrawal Bones are Able to Replenish the Osteoblast Population as Early as 7 Days Post Fracture.

Figure 12: Osteoblast Ablation Leads to a Decrease in TRAP Positive Osteoclasts During the Remodeling Stage of Fracture Healing, and the Decrease in Osteoclast Population Hinders the Healing Process.

Figure 13: In Absence of Osteoblasts There is a Lack of Collagen Type X Matrix Resorption.

Figure 14: Continuous Depletion of Osteoblasts Results in Unresorbed Cortical Bone and Cartilage Matrix at the Fracture Site.

Figure 15: Fluorescent and Immunohistochemistry staining for GFP-tagged SKP-injected into fracture site of continuous GCV treated DTK mice.

Page 7: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

vii

List of Abbreviations

ALP – Alkaline phosphatase

bHLH – A basic helix-loop-helix domain present in two Twist genes (Twist-1 and -2)

BMP – Bone morphogenetic protein

BSP – Bone sialoprotein

BV – Bone volume

CAT – Chloramphenicol acetyltransferase

Cbfa1 – Core binding factor 1

Col-I – Collagen type I

Col-II – Collagen type II

Col-X – Collagen type X

CV – Cartilage volume

CXCR4 – A CXC chemokine receptor (type 4) in the G-protein coupled receptor family specific

for stromal cell-derived factor-1.

DAPI – A polpular flourecent stain (4’, 6-diamidino-2-phenylindole) used for nuclear

counterstain in immunohistochemistry.

DTK – Refers to the transgenic mouse model expressing the HSV-tk gene under the control of a

2.3 kilobase fragment of the rat α1 type I collagen promoter (Col2.3∆tk)

ECM – Extra cellular matrix

EDTA – Ethylenediaminetetraacetic acid is a chelating agent used for bone decalcification.

FGF – Fibroblast growth factor

GCV – Ganciclovir

GFP – Green fluorescent protein

HE – Haematoxylin eosin

HIF – Hypoxia inducible factor

HSV-TK – Herpes simplex virus thymidine kinase

HTC – Hypertrophic chondrocyte

IGF – Insulin-like growth factor

Ihh – Indian hedgehog

IL – Interleukin

M-CSF – Macrophage colony stimulating factor

Page 8: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

viii

MMP – Matrix metalloproteinase

MSC – Mesenchymal stromal cells

NF-κB – Nuclear factor-κB

NOD-SCID – Refers to the non-obese diabetic severe combined immunodeficiency mouse

model

OC – Osteocalcin

OG2 – Osteocalcin gene 2

OCIL – Osteoclast inhibitory lectin

OPG – Osteoprotegerin

OPN – Osteopontin

Osx – Osterix

PDGF – Platelet-derived growth factor

PGE2 – Prostaglandin E2

PTH – Parathyroid hormone

PTHrP – Parathyroid hormone related peptide

RANK – Receptor activator of nuclear factor-κB

RANKL – Receptor activator of nuclear factor-κB ligand

RFP – Red fluorescent protein

Runx2 – Runt-related transcription factor 2

SDF-1 – Stromal cell-derived factor-1

SKP – Skin derived precursors

SO – Safranin O

TGF – Transforming growth factor

TNF – Tumor necrosis factor

TRAF – TNF receptor-associated factor

TRAP – Tartrate-resistant acid phosphatase

TV – Tissue volume

VEGF – Vascular endothelial growth factor

Page 9: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

1

Chapter 1 General Introduction

1.1 Bone Formation

Bone is composed of two distinct tissues, cartilage and bone, each of which includes

specific cell types that fulfill unique functions that are critical for the growth, maintenance and

integrity of the skeleton (Ducy, 1998; Ducy, 2000). Chondrocytes, found in cartilage, are

essential for longitudinal growth of bone from the two metaphyseal ends, and synthesize the

cartilaginous templates during development and postnatal skeletogenesis, onto which osteoblasts

can deposit the bone matrix (Ducy, 2000). Osteoblasts and osteoclasts, found in bone, closely

collaborate in basic multicellular units (BMU) to coordinate the anabolic build-up (synthesis)

and catabolic break down (resorption) of the bone tissue, which occurs regularly throughout the

skeleton at micro scales during adult life (Hadjidakis, 2006; Hill, 1998; Neumann, 2007; Proff,

2009).

Certain genes are critical for regulating the progression of skeletal formation through the

stage of chondrogenesis, angiogenesis, and osteogenesis to the process of remodelling. The

initial phase of mesenchymal condensation involves the expression of transcription factors core

binding factor 1/runt-related transcription factor 2 (Cbfa1/Runx2) and Sox9 genes, which play a

role in determining the population of cells that will occupy and contribute to the condensation

(Hall, 1992; Hall, 1995; Zhou, 2006). Expression of certain growth factors, such as transforming

growth factor beta (TGFβ) superfamily, regulate the proliferation of mesenchymal progenitors

and their differentiation into chondrocytes (Mundlos, 1997a; Mundlos, 1997b). The cartilage

anlage is developed by the secretion of various structural proteins such as syndecan-3, versican

and tenascin by cells within the condensate (Koyama, 1995; Koyama, 1996).

The master regulator, Runx2/Cbfa1, also plays important role in chondrocyte maturation,

angiogenesis and the remodelling of extracellular matrix, as well as being required for

intramembranous ossification (Ferguson, 1999; Yoshida, 2005). Furthermore, it is essential for

fetal osteogenesis (Komori, 1997; Otto, 1997), as deletion of the cbfa1 gene in homozygous null

mutants (cbfa1-/-) results in an almost complete lack of endochondral and intramembranous

Page 10: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

2

ossification, while development of the cartilage scaffold proceeds normally (Komori, 1997;

Mundlos, 1997a; Mundlos, 1997b; Otto, 1997).

This transcription factor regulates the maturation of chondrocytes by controlling the

expression of osteocalcin and osteopontin (OPN), which despite being known as osteoblast-

specific markers, do get expressed in hypertrophic chondrocytes (Gerstenfeld, 1996; Lian, 1993;

Nomura, 1989). By controlling the expression of OPN, which is responsible for the attachment

of cells to the extracellular matrix (Miyauchi, 1991; Reinholt, 1990; Somerman, 1987),

Runx2/Cbfa1 indirectly affects terminal differentiation and apoptosis of chondrocytes. It is also

responsible for directly regulating the expression of matrix metalloproteinase 13 (MMP13), a

collagenase that is involved in remodelling of extracellular matrix that is laid down by

hypertrophic chondrocytes (Vu, 1998).

Another population of cells, the osteoblasts, synthesize a matrix rich in type I collagen

which eventually becomes mineralized, and osteoclasts function to resorb this mineralized

matrix, allowing the remodelling of the rigid bone tissue (Ducy, 2000). Aside from the

transcription factor Runx2/Cbfa1 (Maruyama, 2007), another important signalling molecule

during skeletogenesis is the Wnt-family of molecules, which functions during both embryology

and adult bone repair by regulating the differentiation of mesenchymal precursors into

osteoblasts and the subsequent osteoblastic bone formation (Chen, 2009; Topol, 2009).

Given the biologic importance of these three cell types, and the nonredundancy of their

functions, a defect in differentiation and function of any of these cells leads to severe

repercussions during development that manifests as various diseases of bone and cartilage

(Ducy, 2000). Therefore, knowledge of the genetic network controlling their differentiation and

function (Figure 1) becomes important for developing new and effective therapeutic strategies.

1.1.1 Endochondral Ossification

Endochondral ossification, an anabolic response driven by osteoblasts, includes the

formation of a cartilaginous template that is eventually replaced with bone matrix (Schindeler,

2009). It is initiated by the condensation of mesenchymal cells and their differentiation into

chondrocytes, which secrete a type-II collagen and aggrecan-rich cartilage matrix. Cells

surrounding the condensate form the perichondrium, while cells within the cartilage template

Page 11: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

3

further differentiate into hypertrophic chondrocytes, which requires a cessation of proliferation

and exit from the cell cycle (Hartmann, 2009). These cells produce type-X collagen, as well as

the angiogenic vascular endothelial growth factor (VEGF) and various osteoblast-specific

proteins including alkaline phosphatase (ALP) and OPN, which results in recruitment of new

blood vessels to the avascular cartilage and the mineralization of the cartilage matrix (Iyama,

1991; Kronenberg, 2003). This matrix is eventually resorbed, and the degradation of this

cartilage matrix during endochondral ossification is dependent on the activity of MMPs,

particularly MMP9 (gelatinase B) and MMP13, that regulate remodeling and neovascularization

of the cartilage anlage (Johansson, 1997; Vu, 1998). The invasion of blood vessels into the

cartilage matrix leads to recruitment of osteoblast and osteoclast precursors.

Development of adequate vasculature is critical during the endochondral ossification, as

it determines the rate of bone formation by coupling the processes of chondrogenesis and

osteogenesis (Gerber, 2000). It has been determined that the coordination of metaphyseal and

epiphyseal vascularization, ossification and cartilage formation during endochondral process are

dependent on functions of VEGF isoforms (Maes, 2002). These factors are secreted by the

developing hypertrophic chondrocytes, and lead to recruitment of osteoblasts, osteoclasts and

haematopoietic cells.

Osteoblasts residing in the perichondrium will create a collar of compact bone around the

middle region of the cartilage template, referred to as diaphysis, the site of primary ossification

center. At the two ends of the developing bone, known as epiphyses, secondary ossification

centers are formed, and the cartilaginous plate separating the diaphysis and epiphysis will

become the growth plate, which regulates the longitudinal growth of the long bones (Collin-

Osdoby, 1994; Gerber, 2000; Hall, 2006) (Figure 2A).

1.1.2 Intramembranous Ossification

In contrast to endochondral ossification, no signal from cartilage is required for bone

formation via intramembranous ossification, which is responsible for the formation of flat bones

of the face and skull, as well as parts of the mandibles and clavicles. In this process, cells of the

mesenchymal condensate differentiate directly into osteoblasts (Hartmann, 2009), with the cells

at the edge of the condensates forming the periosteum, a connective tissue that wraps the bone

and holds most of the osteoprogenitor cells (Augustin, 2007). Newly differentiated osteoblasts

Page 12: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

4

immediately start producing the bone matrix osteoid (Hartmann, 2009). Deposition of bone

matrix by the maturing osteoblasts leads to formation of bone spicules, which grow and

eventually fuse with neighboring spicules to create newly formed trabeculae. The increase in size

and number and the ultimate interconnection of these trabeculaes results in formation of a weak

and disorganized structure with high incidences of entrapped osteoblasts (Hermey, 1996a;

Hermey 1996b). This woven bone is gradually replaced with a more organized, stronger lamellar

bone. In this process, the emergence and differentiation of mesenchymal precursors directly into

mature osteoblasts leads to formation of compact bones, and osteoblasts that become entrapped

within lacunae of the matrix differentiate into osteocytes (Hartmann, 2009) (Figure 2B).

1.1.3 Fracture Repair

Fracture healing is a complex process that recapitulates the specific patterns of embryonic

development and bone regeneration and involves the interplay of numerous cell types (Mersell,

2011; Schindeler, 2008) (Figure 3). During the last few decades, various studies investigating

the biochemical and anatomical events of bone healing have rapidly advanced our understanding

of the signalling cascade and pathways that result in successful fracture repair. There are two

types of fracture healing processes, including direct (primary) and indirect (secondary)

(Gerstenfeld, 2003; Rahn, 2002). Also, many of the mechanisms involved in the skeletal repair

process recap the events of embryonic development (Einhorn, 2001; Ferguson, 1999;

Gerstenfeld, 2003).

Primary direct mode of skeletal repair is an uncommon event, which happens at regions

of biomechanical instability and requires rigid stabilization of the fracture site (Allgower, 1979;

Perren, 1979; Schatzker, 1989). This rigid stabilization suppresses the generation of a callus in

either the cancellous or cortical bone. In contrast to indirect bone healing, this process involves

generation of a weak intermediate and does not occur in anaerobic environment (Madison,

1993). Two forms of primary skeletogenesis exist, gap healing and contact healing, where bone

union is achieved without an external callus formation, fibrous tissue or cartilaginous scaffold

formation within the fracture gap (Liu, 1999; Perren, 1979; Schenk, 1967; Schenk, 1987). Gap

healing is initiated by formation of woven bone, followed by generation of parallel oriented

lamellar bone that provides support (Schenk, 1967; Schenk, 1987). This is ensued by

remodelling of the longitudinal haversian canals, reconstruction of the necrotic fracture ends and

Page 13: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

5

formation of new bone at the fracture site that replaces and reorients the fractured bone back to

the original configuration (DeLacure, 1994). In contrast, contact healing occurs in cases where

bone fragments are in direct appositions and is not preceded by a transverse bone formation

between the fracture ends. In this form of direct healing osteons are able to grow parallel to the

long axis of the bone across the fracture site, supplemented by tunnelling osteoclastic activity

forming cutting cones across the fracture line that allow the penetration of blood vessels,

accompanied by osteoprogenitor and endothelial cells (Perren, 1969; Perren, 1979; Schenk,

1967; Schenk, 1987; Thomas, 1998). This form of bone repair will also lead to regeneration of

normal bone architecture.

Secondary indirect bone healing is characterized by spontaneous bone formation in

absence of a rigid stable fixation of the fracture site, and is the most common method of fracture

healing consisting of both endochondral and intramembranous process of bone generation

(Gerstenfeld, 2006). This form of healing is improved by micro-motion and weight-bearing,

although delayed healing or non-union has been observed in cases where too much motion

and/or load is applied (Green, 2005; Pape, 2002; Perren, 2002). It occurs in areas where

application of motion cannot be avoided, such as intramedullary nailing, external or internal

fixation of complicated fractures (Pape, 2002; Perren, 2002). It consists of several overlapping

phases that span an inflammatory phase, a reparative phase that consists of both

intramembranous and endochondral ossification resulting in formation of soft callus followed by

remodelling and establishment of hard callus, and a final remodelling phase that re-establishes

the orientation of the original bone orientation (Bolander, 1992; Frost, 1989; Madison, 1993;

Marsh, 1999; McKibbin, 1978; Nemeth, 1988; Thomas, 1998; White, 1977).

Bone formation during adult growth and regeneration has been shown to be reflected in

the processes occurring during fetal development (Ferguson, 1999). To understand the

similarities and differences between these two processes, a series of expression assays was

performed to examine the molecular signals critical for bone synthesis and repair (Ferguson,

1999). The study showed that some aspects of bone development are regulated by similar

molecular signals during fetal development and adult growth/repair. These include the

maturation of chondrocyte, invasion of vasculature and the process of ossification. Major

differences between these two developmental processes are the initial inflammation response

Page 14: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

6

during adult healing that influences the mechanisms controlling the mesenchymal cell

condensation during adult repair process (Ferguson, 1999).

1.1.3.1 Inflammatory Response

The immediate response to injury from fracture trauma is usually associated with damage

of the local soft tissue integrity along with disruption to normal vascular function and nutrient

flow at the fracture site. The bleeding within the fracture site triggers the infiltration of

macrophages, degranulating platelets, and other inflammatory cells into the developing

haematoma that aid in clearing the degenerated cells and secrete various cytokines and growth

factors (Einhorn, 1998; Gerstenfeld, 2003; Schineler, 2008). The eventual coagulation of the

haematoma around the site of injury provides a template for the formation of future callus

(Gerstenfeld, 2003).

Current knowledge suggests that the cascades of growth factors secreted by the

inflammatory cells regulate different step in fracture bone healing. The signalling molecules

involved in this process include TGF-β, platelet-derived growth factor (PDGF), fibroblast growth

factor-2 (FGF-2), VEGF, macrophage colony stimulating factor (M-CSF), interleukins-1 and -6

(IL-1 and -6), bone morphogenetic proteins (BMPs), and tumor necrosis factor-α (TNF-α)

(Bolander, 1992; Einhorn, 1998; Gerstenfeld, 2003). This cellular response creates a positive

feedback loop that assists the recruitment of additional inflammatory cells, triggering the

migration and invasion of mesenchymal progenitor cells as well as influencing the initiation of

repair process and development of fracture callus (Barnes, 1999; Bolander, 1992; Iwaki, 1997;

Shapiro, 2008). Although prolonged or chronic expression of inflammatory cytokines is known

to have a negative effect on bone healing or any implanted materials, a transient highly regulated

response is critical for tissue regeneration (Gerstenfeld, 2003).

1.1.3.2 Formation of Soft Callus

The first 7 to 10 days of fracture healing involves the process of chondrogenesis, where

mesenchymal progenitor derived chondrocytes proliferate and produce a semi-rigid cartilaginous

template adjacent to the fracture site. It has been suggested that the fracture line in the bone may

set up the overall spatial relationships of the generated mesenchymal condensations, which is

exhibited by the development of various discrete crescent shaped centers of condensates that are

Page 15: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

7

symmetric with respect to the fracture line (Gerstenfeld, 2003). The formation of the

cartilaginous tissue occurs in regions that are mechanically less stable, such as between the

fracture ends and external to the periosteal site, and serves as a soft callus that provides the initial

mechanical support to the fracture (Rahn, 2002). This tissue will later undergo mineralization,

resorption and is superseded by the production of the bony callus.

On the cellular level, this stage of bone healing represents the involvement of

chondrocytes and fibroblasts, with the relative proportion of these cells varying between

different fractures (Gerstenfeld, 2003). In cases where the cartilage production is absent or

delayed, the fracture site is instead infiltrated by fibroblasts that generate a fibrous tissue.

Mesenchymal condensations and the subsequent chondrogenesis occurs at multiple discrete sites

within the fracture site that will eventually proliferate and merge to form a centralized

fibrocartilaginous structure between the fracture ends (Barnes, 1999). Coordinated expression of

various growth factors, such as TGF-β2, TGF-β3, PDGF, FGF-1, and insulin-like growth factor

(IGF), stimulate the proliferation/differentiation of the emerging fibroblastic and chondrocytic

cells (Cho, 2002, Einhorn, 1998; Gerstenfeld, 2003), while chondrogenesis is promoted by

various members of the BMP family of cytokines, such as BMP-2, -4, -5, and -6 (Al-Aql, 2008).

In response to these factors, chondrocytes are able to produce extensive amounts of

extracellular matrix proteins, such as collagen type II. As chondrocytes within the condensate

mature, they undergo hypertrophy and begin to secrete type X collagen (Al-Aql, 2008). Prior to

the synthesis of hard callus, the deposited cartilage scaffold undergoes calcification, gradual

removal of soft callus, neovascularisation, and the systemic deposition of woven bone through

the interplay of an array of molecular cytokines and pro-angiogenic factors that include VEGF,

BMPs, FGF-1 and TGF-β (Al-Aql, 2008; Cho, 2002; Gerstenfeld, 2003; Marsell, 2009). Other

factors involved in the cascade that initiates resorption include the macrophage colony-

stimulating factor (M-CSF), receptor activator of nuclear factor NF-κB ligand (RANKL),

osteoprotegrein (OPG) and TNF-α (Barnes, 1999; Gerstenfeld, 2003).

1.1.3.3 Formation of Hard Callus

The maturation of prehypertrophic chondrocytes of the soft callus is accompanied by

enlargement of cellular volume and increase in extracellular matrix production. The deposited

matrix undergoes spontaneous calcification that result in formation of hydroxyapatite crystals.

Page 16: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

8

The calcification and nucleation mechanism begins with the accumulation of calcium-containing

granules and their transport into the extracellular to the extracellular matrix where they

precipitate with the surrounding phosphate ions and form the initial mineral deposits (Ketenjian,

1975). This calcified cartilage needs to be resorbed prior to its replacement by the woven bone

that is secreted by osteoblasts (Einhorn, 1998; Giannoudis, 2011).

This hard callus formation stage of bone repair is characterized by high levels of

osteoblast activity and represents the most active period of osteogenesis, leading to formation of

mineralized bone matrix. The ability of the early differentiated osteoblasts to progress down the

osteogenic lineage in an ordered manner is a critical prerequisite for efficient fracture healing.

The progression of the hard callus stage involves the replacement of the calcified cartilage with

woven bone that provides mechanical rigidity to the healing bone (Gerstenfeld, 2006). This is

carried out primarily through the function of MMP13, which is produced by cells in the

osteoblastic lineage (Nakamura, 2004) that invade the cartilage matrix. Mice deficient in

MMP13 function exhibit a defect in removal of the deposited cartilage which cannot be rescued

with normal bone marrow transplantation (Behonick, 2007; Kosaki 2007). Osteoclast cells that

follow the early osteoblast in-growth phase synthesize MMP9 (Andersen, 2004), another

important degradative enzyme shown to be important for cartilage resorption (Colnot, 2003).

As the process of cartilage template removal continues the outer layer of the callus forms

the structure that will make up a new cortical shell (Cao, 2002; Komatsubara, 2005; Li, 1999).

The new woven bone matrix deposited by newly differentiated osteoblasts consists of both

proteinaceous and mineralized bone extracellular matrix, which is typically irregular and under-

remodelled (Gerstenfeld, 2006). The bone forming osteoblasts populate the newly formed

endosteal surface of the callus, and a thin layer of osteoclastic cells followed by a layer of

myofibroblastic cells advance toward the outer periosteal surface of the shell (Ushiku, 2010).

The outer cortical shell is formed with a distinctive dynamic appearance, which is influenced by

the mechanical load that is applied during motion (Ushiku, 2010). As the base of the shell

thickens the active region of bone formation contracts toward the apex of the shell and the

process of remodelling occurs in an inward manner from outside toward inside of the convex

cortical shell (Ushiku, 2010). The initial chondrocytes at the center of the fracture zone are

products of the progenitor population residing from the periosteum. The bone formation over the

surface of the callus to form the new periosteum and the outer cortical shell also derive from the

Page 17: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

9

periosteal progenitors (Gerstenfeld, 2009; McDonald, 2008). However, the subsequent

remodelling of the fracture callus requires osteoclasts. Osteoclast inhibitors do not affect the

early stages of fracture healing, but in the absence of sufficient osteoclast function there is a

delay of inward remodelling and failure to resorb the mineralized cartilage core (Gerstenfeld,

2009; McDonald, 2008).

1.1.3.4 Bone Remodeling

In order to fully restore the biomechanical properties to the normal bone, the generated

rigid hard callus that is composed mostly of woven bone, requires to be remodelled back to the

original bone configuration. This is done through a second phase of resorption that replaces the

unorganized woven bone of the hard callus with a more organized lamellar bone structure and

creates the central medullary cavity (Gerstenfeld, 2003; Marsell, 2011). The orchestration of this

phase is regulated by a series of biochemical factors, such as IL-1, TNF-α, BMP-2, that show

reasonably high expression level during this stage of bone healing (Al-Aql, 2008; Marsell, 2009;

Mountziaris, 2008). This process involves the balanced coordination of hard callus resorption by

osteoclasts, followed by lamellar bone synthesis by osteoblasts (Marsell, 2009; Schindeler,

2008).

Based on histological evidence that shows the presence of large multinucleated cells at

the edge of the soft callus, along with the invading vascular endothelial cells, osteoclasts have

been suggested to play a key role in both soft callus and hard callus remodeling (Schell, 2006).

Although these cells display similar morphology to osteoclasts and stain for classic osteoclast

markers (such as TRAP staining), they have been termed chondroclasts, based on their physical

location (Cole, 1987).

Active bone resorption results in the release of various growth factors and products, such

as TGFβ, BMPs, and collagen peptides that signal the breakdown of deposited osteoid.

Concurrently, high levels of bone resorption are responsible for stimulation of a local anabolic

response, which is influenced and modulated by mechanical forces (Martin, 2005; Mohan, 1991).

This is seen in cases where lack of loading on the developing bone, such as disuse and/or rigid

fixation, leads to impaired bone healing (Rubin, 2002). Furthermore, it has been observed in

rodent animal models that the anabolic activity of osteoblasts can still be driven by the secreted

osteoclast factors, such as osteoclast inhibitory lectin (OCIL) (Nakamura, 2007), even in

Page 18: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

10

presence of non-functional osteoclast cells (Marzia, 2000; Schaller, 2004). This suggests that the

release of factors from resorbed bone matrix may not be critical in stimulating osteoblast

function, and the exact mechanism for the anabolic/catabolic response coupling during bone

remodelling needs to be further investigated (Martin, 2005).

1.2 Skeletal Cell Types

Unlike other organs that generate scar tissue upon healing, the skeleton possesses a

unique regenerative potential where the newly formed bone is indistinguishable from the

uninjured adjacent tissue. As well, there are various conserved similarities and notable

differences between fetal skeletal tissue development and adult reparative process. For instance,

in both processes progenitor cells aggregate at site of future bone formation to form

condensations (Hall, 1988; Thompson, 2002) that proceed down a chonrogenic/osteogenic

lineage via the expression of a conserved set of molecular markers that regulate cell

differentiation and maturation (Ferguson, 1999; Karsenty, 2002; Vortkamp, 1998). Furthermore,

both fetal or adult skeletal formation require extracellular matrix remodelling and vascularisation

as prerequisite for ossification, and disruption of these events can lead to delay of subsequent

bone formation (Thompson, 2002; Vu, 1998). In contrast, mechanical forces (Carter, 1998; Le,

2001; Probst, 1997; Thompson, 2002) and inflammatory response (Simon, 2002) have

substantial influence on cartilage and bone formation during adult fracture repair, but are not

required for the initiation of chondrogenesis or osteogenesis during fetal growth. Chondrocytes,

osteoblasts, osteocytes and osteoclasts are key players in the process of bone formation and

repair, where the absence of any one of these lineages will disrupt the complex process of

skeletogenesis.

1.2.1 Chondrocytes

Chondrocytes, the cartilage forming cells, promote bone growth throughout development.

During endochondral growth plate development, four chondrocyte populations become arranged

in distinct narrow zones, which range from the resting zone closest to the joint surface to the

proliferating, prehypertrophic and hypertrophic zones at the calcified end of the growth plate

(Adams, 2002; Wagner, 2001). These cells are organized in columns and divide along the long

axis of the plate, changing in shape and phenotype as they progress through the various zones.

As the cells become hypertrophic, they become swollen and increase in volume while expressing

Page 19: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

11

type X collagen. Secretion of this matrix acts to initiate mineralization, whereby trabeculae of

woven bones are deposited by osteoblasts onto the calcified cartilage septa (Adams, 2002).

A complex network of signalling molecules regulates the proliferation and differentiation

of these different chondrocyte subpopulations (Figure 1A). The earliest molecule to be

expressed during chondrogenesis is the transcription factor Sox9, which is required for

chondrogenesis of the mesenchymal condensate that prefigures bone formation (Wright, 1995).

Various studies, looking at either conditional knock-out or null Sox9 transgenic mouse lines,

have shown that in absence of Sox9 there was no cartilage formed in the limbs of these mice and

cells in the mesenchymal condensate do not adopt a chondrogenic morphology (Akiyama, 2002;

Bi, 1999). Sox9 is the master gene that regulates the expression of various genes involved in

chondrogenesis, such as Sox5 and Sox6, and together these genes are implicated in induction of

cartilage matrix proteins, aggrecan and type II collagen (Han, 2008; Lefebvre, 1997; Lefebvre,

1998a; Lefebvre, 1998b).

Furthermore, recent studies have shown Runx2/Cbfa1 to be one of the transcription

factors genetically downstream of Sox9 controlling the differentiation of chondrocyte

hypertrophy (Takeda, 2001; Ueta, 2001). This was shown in mice lacking Runx2/Cbfa1 that

lacked hypertophic chondrocytes in several but not all skeletal elements (Inada, 1999; Kim,

1999). Two other growth factors, parathyroid hormone related peptide (PTHrP) and Indian

hedgehog (Ihh), provide a tightly regulated feedback loop that play critical roles in controlling

chondrocyte maturation through the different zones. Cells from the perichondrium secrete PTHrP

that signal via their PTH/PTHrP receptors on prehypertrophic chondrocytes that suppress their

differentiation into hypertrophic chondrocytes (Karaplis, 1994; Lanske, 1996). Furthermore, Ihh

is secreted by prehypertrophic chondrocytes that signal to the cells in the perichondrium to

upregulate PTHrP synthesis that in turn inhibits chondrocyte hypertrophy and endochondral bone

formation (St-Jacques, 1999; Vortkamp, 1996).

1.2.2 Osteoclasts

Bone acts as a reservoir for calcium and phosphate in the body and is resorbed/ deposited

via feedback mechanisms in accordance with the stresses that are placed upon it (Robert, 2009).

Bone remodelling, which is essential for repairing microdamages and optimizing bone structures

for mechanical function, is a continuous episode that is mediated by specialized bone resorbing

Page 20: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

12

cells, the osteoclasts (Robert, 2009). The haematopoietic stem cell-derived osteoclasts require

close interaction with osteoblasts for their activation (Figure 1B).

Receptor activator of nuclear factor-κB ligand (RANKL), a member of the TNF

superfamily, is expressed by osteoblasts and is the primary mediator of osteoclastogenesis

(Lacey, 1998; Robert, 2009). Through its interaction with its receptor RANK, which is expressed

on osteoclasts and their precursors, RANKL promotes osteoclast differentiation, fusion and

activation (Lacey, 1998). After ligand-induced trimerization, RANK promotes cytoplasmic

signaling by recruiting adaptor molecules that activate downstream signaling pathways mediated

by protein kinases that include inhibitor of NF- κB kinase (IKK), c-Jun N-terminal kinase (JNK),

p38, Src and extracellular signal-regulated kinase (ERK) pathways (Boyle, 2003; Teitelbaum,

2003). A key preliminary step in RANK signaling involves the binding of TNF receptor-

associated factors (TRAFs) to specific cytoplasmic domains of RANK, which directs activation

of transcription factor NF- κB and activator protein-1 (AP-1) (Boyle, 2003). NF- κB has been

shown to be pivotal to osteoclastogenesis and deletion of NF- κB leads to osteoclast-autonomous

osteopetrosis, characterized by absence of multinucleated bone-resorbing cells (Franzoso, 1997).

Another transcriptional influence of RANKL include the activation of osteoclastogenic

transcription factor NFATc1 (also called NFAT2), which prompts osteoclastogenesis when

expressed in precursor cells even in the absence of RANKL (Ishida, 2002; Takayanagi 2002).

Active osteoclasts attach to bone, creating a tight seal, and are identified by expression of

the phosphatase TRAP. In addition, they secrete a range of factors involved in progenitor

mobilization (Heissig, 2002a; Kollet, 2003; Pruijt, 1999) and migration of preosteoclasts to the

resorption site (Blavier, 1995), that include the cytokine IL-8 (Rothe, 1998) and the proteolytic

enzyme MMP-9 (Vu, 1998) and cathepsin K (Kollet, 2003). The role of MMP-9 in bone

remodelling has been shown in MMP-9 knockout mice, where the growth–plate cartilage suffers

delayed ossification suggesting a role for this enzyme in cartilage resorption (Vu, 1998).

Osteoclasts are distinguished by their polarization toward the bone and the formation of a

ruffled membrane structure, which acts as the resorptive organelle of the cell and contains a

villous-like complex with numerous ‘spike-like’ vacuolar H+ATPase proton pumps (Blair, 1989).

The formation of this structure is dependent on the contact of osteoclast with bone and allows the

creation of an acidic environment between the cell and the juxtaposed bone structure. Cathepsin

Page 21: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

13

K, one of the major bone-resorbing proteases, is essential for bone remodelling and degradation

of type I collagen (Goto, 2003). Secretion of these enzymes by osteoclasts into the resorptive

pits, which are isolated by tight seals, allows the degradation of the skeletal matrix and

progression of bone remodelling.

Osteoprotegerin (OPG), a soluble decoy receptor, is secreted by osteoblast cells and

inhibits osteoclastogenesis by blocking RANKL binding to its cellular receptor RANK on

osteoclasts (Boyle, 2003). Mice overexpressing or deficient in OPG have been shown to exhibit

phenotypes of marked increased or decreased in bone density, respectively (Bucay, 1998;

Simonet, 1997). Overexpression of OPG leads to inhibition of RANKL activation of osteoclasts

and the resulting decreased osteoclastogenesis is manifested in increased bone density, as seen in

osteopetrotic pathologies (Simonet, 1997). In contrast, a deficiency in OPG is characterized by

increased bone resorption, severe trabecular and cortical proposity, leading to an osteoporotic

phenotype with decreased bone density and high occurrences of fractures (Bucay, 1998). The

cytokine M-CSF is expressed by osteoblasts and their precursors as a soluble and membrane-

bound protein and promotes survival and proliferation of osteoclast precursors (Teitelbaum,

2003). The role of M-CSF in osteoclastogenesis was established by studies of op/op mouse that

display osteoclast-deficient osteopetrosis due to nonfunctional M-CSF (Yoshida, 1990).

A chemokine, SDF-1, together with its receptor CXCR4 play pivotal role in survival,

proliferation, anchorage of stem cells to the endosteal region of bone or the endothelial

microenvironment within the bone tissue (Adams, 2006; Cottler-Fox, 2003; Lapidot, 2005). This

ligand is highly expressed by bone marrow endothelial cells as well as endosteal osteoblasts

(Adams, 2006; Kollet, 2006; Ponomaryov, 2000), which have been shown to support the

maintenance of the haematopoietic stem cell niche (Yin, 2006) and retaining stem cells in

quiescent state (Arai, 2004). An increase in SDF-1 is seen under stress situations such as injury,

chemotherapy, inflammation or treatment with granulocyte colony-stimulating factor (G-CSF),

which triggers an imbalance of steady-state homeostasis and induce massive stem cell

mobilization, proliferation and differentiation (Cottler-Fox, 2003; Heissig, 2002). Ultimately this

leads to enhanced secretion of proteolytic enzymes such as MMP-9, elastases and cathepsin K,

resulting in increased activity of osteoclasts and higher bone resorption (Takamatsu, 1998;

Watanbe, 2003). Through a feedback mechanism, following the increase in proteolytic enzymes

Page 22: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

14

by osteoclasts there is degradation of SDF-1, increased expression of CXCR4 and mobilization

of maturing progenitors and stem cells (Levesque, 2003; Petit, 2002).

1.2.3 Osteoblasts

Osteoblasts are highly differentiated extracellular matrix-producing cells that are derived

from an undifferentiated progenitor population through a series of differentiation and maturation

events (Aubin, 1998). During their differentiation process, osteoblasts can either (1) undergo

programmed cell death, (2) become quiescent and transform into inactive bone-lining cells, or

(3) merge progressively within the bone matrix to become mature osteoblasts (Jilka, 1998;

Manolagas, 2000; Noble, 1997). Some researchers also report cases where osteoblasts

transdifferentiate into cells able to lay down chondroid bone (Li, 2004). Based on efforts by

numerous groups embarking in the research to identify osteoblast-specific transcription factors,

several aspects of osteoblast biology have been recognized. Primarily it has been noted that

throughout the differentiation process, osteoblasts remain similar to fibroblasts and the only

difference is slow to appear and seen late in the process, with the secretion of extracellular

matrix that is only produced by mature osteoblasts (Aubin, 1996). Due to absence of phenotypic

changes throughout osteoblast differentiation, it has been difficult to study osteoblast biology at

cellular levels. As a result, gene expression assays have been utilized to assess the various

aspects of osteoblast differentiation.

Cells of osteoblast origin have been shown to play essential role in orchestrating bone

formation and remodelling. They are involved in bone formation during embryonic development

and fracture repair by depositing new bone matrix known as osteoid, while playing a role in bone

resorption and remodelling by producing a wide range of compounds that regulate osteoclast

activity (Hofbauer, 2000; Lerner, 2000; Puzas, 1992; Suda, 1997). These regulating molecules

include prostaglandin E2 (PGE2), IL-1, IL-6, and RANKL (Hofbauer, 2000; Lerner, 2000;

Puzas, 1992; Suda, 1997).

Several publications have shown the involvement of mononuclear cells in phagocytosis

of collagen at sites adjacent to osteoclast resorption pits (Beertsen, 1978; Garant, 1976; Rifkin,

1979; Tran, 1982; Yajima, 1999). Prior to bone resorption, osteoblasts aid in removing any

nonmineralized osteoid from bone surface by secreting a number of MMPs (Chambers, 1985a;

Chambers, 1985b), and osteoblasts mediate the initiation of bone formation after resorption,

Page 23: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

15

which results in a finely tuned remodelling process (Harris, 1969; Kahn, 1987; Martin, 1994;

Rodan, 1997). Quite a few studies have shown that after resorption by osteoclasts, the resorption

pit still contains non-digested demineralised bone matrix (Delaisse, 1987; de Saint-Georges,

1989; Holliday, 1997; Jones, 1986; Tran, 1982). A study by Everts and colleagues looked at the

involvement of bone lining cells in coordination with bone resorption and regulation of bone

formation after resorption by osteoclasts (Everts, 2002). By analysing mouse long bone and

calvariae they determined that after osteoclasts have exerted their activity, bone lining cells enter

the resorption pit and subsequently digest any protruding collagen fibrils that are left by

osteoclasts. Several publications have reported that osteoblasts are driven toward the resorption

pits via signalling molecules such as TGF-β (Erlebacher, 1998; Oursler, 1994; Pfeilschifter,

1990), IGF (Hayden, 1995), or OPN (Dodds, 1995).

Runx2/Cbfa1 is necessary for both endochondral and intramembranous bone formation

and is expressed in MSC during early embryonic development, where it acts as a master

regulator in commitment of these cells to osteoblastic lineage. Mice lacking in Runx2 gene fail to

form mineralized bone in any part of the skeleton, lack mature osteoblasts, and have significantly

decreased expression of osteoblast differentiation markers such as ALP, OPN, and osteocalcin

(Komori, 1997; Otto, 1997). Furthermore, other studies have shown that expression of

Runx2/Cbfa1 is cooperatively regulated by BMP-2 and TGF-β signalling, two genes that play

essential role in the process of skeltogenesis (Lee, 2003). Current knowledge indicates that most

of the proteins factors expressed by osteoblasts are also expressed by other cell types such as

fibroblasts or chondrocytes. As a membrane bound enzyme, bone ALP is a maker for

hypertrophic chondrocytes (Althoff, 1982; Habuchi, 1993), is associated with early

differentiation of osteoblasts (Stein, 1993), and plays important role in extracellular

hydroxyapatite formation (Hui, 1997). The matrix protein osteocalcin is expressed exclusively in

bone and attenuates the process of bone formation since osteocalcin-deficeint mice display

increased bone mass (Ducy, 1996; Mark, 1988; McKee 1992). Osteocalcin is present in the

beginning of the mineralization phase while ALP expression is linked to matrix maturation

(Tuckermann, 2000). The only true osteoblast-specific gene that is expressed by fully

differentiated osteoblasts and regulates their function is osteocalcin (Desbois, 1994; Ducy, 1996;

Hauschka, 1989).

Page 24: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

16

The novel zinc finger-containing transcription factor, osterix (Osx), was originally

identified via a simple screen to isolate osteoblast-specific cDNA (Nakashima, 2002), and found

to be required for osteoblast differentiation and bone formation. Osx is expressed in osteoblasts

of all endochondral and intramembranous bones and in absence of Osx there is no cortical or

trabeculae bone formation. In Osx null mice cartilage anlagen is fully formed and mesenchymal

cells, along with blood vessels and osteoclasts, are able to invade the mineralized cartilage

matrix during endochondral ossification, but do not deposit any bone matrix (Nakashima, 2002).

Similarly, the osteogenic cells in intramembranous skeletal elements of these mice are

completely blocked in their differentiation into osteoblasts. These cells do however exhibit

Runx2/Cbfa1 expression at levels comparable to those in wildtype osteoblasts. In contrast, Osx is

not expressed in Runx2/Cbfa1 null mice, indicating that Osx is not required for Runx2/Cbfa1

expression and acts genetically downstream of Runx2/Cbfa1 pathway (Nakashima, 2002).

During mouse embryonic development Runx2/Cbfa1 expression is detected as early as

E10, however expression of molecular markers of differentiated osteoblasts is seen around E13

and replacement of the cartilaginous template by bone occurs at later E15 time point (Bianco,

1991; Chen, 1992; Ducy, 2000; Kauffman, 1992). This delay implies that other regulatory

proteins such as Osx, an activator of transcription (Nakashima, 2002), are involved in this

process. Other factors such as twist proteins, expressed transiently in osteoblast progenitors, act

as inhibitors of Runx2/Cbfa1 function. In vertebrates, two twist genes (Twist-1 and -2) exist that

code for the basic helix-loop-helix (bHLH)-containing transcription factors (Wolf, 1991; Li,

1995). Bialek and colleagues demonstrated that during skeletogenesis Twist proteins transiently

inhibit osteoblast differentiation via the interaction of a novel antiosteogenic domain within these

proteins with Runx2/Cbfa1 DNA binding domain (Bialek, 2004), thereby interfering with its

function. This Twist box, a domain distinct from the bHLH domains, is a 20 amino-acid long

domain that allows the antiosteogenic function of Twists (Castanon, 2002).

Regulation of the Runx2/Cbfa1 gene expression and protein functionality occurs at

multiple other levels, where various cytokines, growth factors, and hormones, including TGF-β,

BMP, FGF, sonic hedgehog, vitamin D3, and estrogen, control the spatial and temporal

expression pattern of Runx2/Cbfa1 (D’Souza, 1999; Ducy, 1997; Kim, 2003; Lee, 1999;

Takamoto, 2003; Tou, 2001; Tsuji, 1998; Zhou, 2000). Taken together, these data suggest that

aside from its critical role in osteoblast growth and chondrocyte hypertrophy, Runx2/Cbfa1 may

Page 25: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

17

indirectly control osteoclast function by mediating RANKL and OPG production by osteoblasts

(Enomoto, 2003; Jonason, 2009; Mori, 2006; Usui, 2008).

1.2.4 Osteocytes

Osteocytes are the most abundant cellular components found in the cortical bone

structure, making up 95% of all cells in bone and covering close to 94% of all bone surfaces

(Frost, 1960; Marotti, 1996). An individual bone consists of ten times more osteocytes than

osteoblasts, embedded in the cortical bone tissue (Parfitt, 1990). They are formed during various

modes of ossification when advancing active osteoblasts lay down bone matrix that engulfs the

osteoblasts already at the ossification front (Knothe, 2004).

Similar to the cross-talk between osteoblasts and osteoclasts that has previously been

established (Lacey, 1998; Yasuda, 1998), there is cross-talk between osteocytes and osteoblasts

(Burger, 2003). Osteocytes form a meshwork of cell processes with one another through a

system of canaliculi that is formed in the bone matrix and allows communication between

adjacent osteocytes (Palumbo, 1990a). Once osteoblasts, which are estimated to only live up to

three month in human bones (Manolagas, 2000) or even shorter (10-20 days) in mouse alveolar

bone (McCulloch, 1988) become embedded in the developing bone, they mature into osteocytes

that have a greater lifespan than active osteoblasts.

Numerous terminologies have been used to describe the osteoblast to osteocyte transition

(Holtrop, 1990; Manolagas, 2000; Meunier, 1989), however not much research has been

conducted in the field of bone biology that focuses on the formation and biology of osteocytes.

The process of osteoblast transformation into osteocytes is dependent on a variety of factors,

ranging from the mode of ossification, the type of bone generated (woven vs lamellar bone), the

location of bone formation, the species, and on the gender/age of the model being studied (Hall,

2005). Once the osteoblasts become embedded in the bone matrix and transform to osteocytes

they cease their bone deposition activity and instead function to maintain the integrity of bone

structure by acting as strain and stress sensors (Burger, 2003; Knothe, 2004). The embedded

osteocytes are also able to deposit and resorb bone around the lacunae in which they reside, as a

result changing the shape of the lacuna in a process called osteocyte osteolysis. Although this

process is not seen in human osteocytes, there is evidence for its occurrence in other vertebrates

such as squirrels (Haller, 1978), rats (Belanger, 1977; Tazawa, 2004), snakes (Alcobendas,

Page 26: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

18

1991), eels (Lopez, 1980), and bats (Doty, 1985; Kwiecinski, 1985; Kwiecinski, 1987), among

other species (Steinberg, 1981; Witten, 2000; Zhang, 2000).

Several hypotheses exist in the literature as to how the transition from osteoblasts to

osteocytes occurs within the bone matrix. Unpolarized osteoblasts can become entrapped by their

own secretion by laying down bone in all directions, while polarized osteoblasts can become

embedded within the matrix via their neighbouring cells (Nefussi, 1991; Palumbo, 1990a;

Palumbo, 1990b). Another scheme is that of acellular, osteocyte-deprived bone that occurs when

a group of highly polarized osteoblasts function as a unit to lay down bone matrix in a

synchronized matter, moving away from the osteogenic front and resulting in creation of

acellular bone (Ekanayake, 1987; Ekanayake, 1988; Witten, 1997). The fates of the remaining

osteoblasts include apoptosis or becoming bone-lining cells (Franz-Odendaal, 2006). The

number of osteoblasts that become osteocytes has been estimated to be 10-20% (Aubin, 1998),

whereas apoptosis is thought to eliminate approximately 60-80% of these osteoblasts (Jilka,

2007). In this fully formed bone, the osteocytes constitute 95% of the cells in the bone (Franz-

Odendaal, 2006).

1.3 Osteogenic Proteins

From a physiological perspective, both molecular and genetic regulatory mechanisms are

essential for the recruitment, differentiation and maturation of each of the cell types involved in

skeletogenesis. As the process of bone formation is a complex multistep phenomenon, requiring

the participation of a variety of cells, there is a broad spectrum of physiological regulatory

signaling proteins that manage the initiation and progression of individual stages of ossification,

ranging from neovascularization to chondro/osteogenesis and matrix remodeling (Einhorn,

1995). This introduction will explore the mechanism and function of a few of these proteins that

have been determined to be important during bone formation.

1.3.1 Vascularization in Successful Bone Healing

Adequate blood supply during development and bone repair has been viewed in the

literature as one of the key elements that warrants accurate and timely restoration of the bone

tissue (Schindeler, 2009). The primary underlying cause of poor bone repair has been proposed

to be due to vascular deficiency (Pelissier, 2003) and hypoxic stress has been shown to stimulate

Page 27: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

19

secretion of pro-angiogenic factors from osteochondral cells that in turn promote the re-

establishment of a vascular supply to support bone cell differentiation (Wang, 2007a).

Furthermore, compiling evidence that argues for the temporal occurrence of bone

formation and vascularisation has generated an emerging concept that vascular cells, pericytes,

may contribute to bone repair. Pericytes have been shown to have myogenic differentiation

capacity (Dellavalle, 2007), and to exhibit osteogenic phenoytpes in vitro, expressing

osteoblastic markers and ability to form mineralized nodules (Brighton, 1992; Doherty, 1998;

Schor, 1990). This is further supported by data exhibiting a synergistic interplay between

vaslcular and osteoprogenitor cells. It has been shown that overexpression of the key factor

hypoxia inducible factor α (HIF1α) in osteoblasts results in development of overly dense bones,

while disruption of this signalling pathway produces the opposite phenotype of osteoporotic

bones (Wang, 2007b). Additionally, co-expression of the potent pro-angiogenic factor VEGF

with bone inducing molecule BMP4, transplanted in vivo via implanted stem cells, resulted in

increased ectopic bone formation (Peng, 2002).

Arrays of various molecular effectors that are involved in normal embryonic vascular

development also play a role in neovascularisation in adults (Carmeliet, 2003). These factors

range from VEGF, angiopoietins (Ang-1), basic fibroblast growth factor (bFGF), hypoxia

inducible factor α (HIF1α) and various members of TGFβ, to PDGF, IGF family (IGF-1, IGF-2),

and hepatocyte growth factor (HGF) (Madeddu, 2005). Interestingly, OPN-deficient mice, aside

from an increase in osteoclastic activity that leads to increased bone resorption and lower bone

density, display medial calcification of renal and aorta, which suggests a role for OPG in

correlation involving vascular calcification and osteoporosis (Bucay, 1998).

The family of VEGFs and their receptors are critical regulators of angiogenesis that play

an important role in skeletal growth and repair by initiating an array of signal transduction

cascades that eventually lead to development of vasculogenesis, or angiogenesis in the case of

tissue repair (Carmeliet, 2000; Ferrara, 2001; Rossant, 2002; Tammela, 2005; Yancopoulos,

2000). This creates a network between neighbouring cells, which by providing an adequate

system for flow of different cytokines, chemokines, and growth factors helps maintain the

homeostatis of the surrounding tissues. Any inhibitions in development of this system in

Page 28: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

20

repairing bone tissue leads to various pathologies such as osteonecrosis (Childs, 2005),

osteomyelitis (Lazzarini, 2002), and osteoporosis (Alagiakrishnan, 2003; Burkhardt, 1987).

There are multiple stages during growth and development where vascularisation of

cartilage regions is required. During embryonic development, there is invasion of blood vessels

originating from the perichondrium, into the emerging cartilage structure. Second, during

development, rapid vascularisation of the growth plates allows for elevated postnatal growth.

Third, during adulthood, the process of angiogenesis is controlled periodically when anabolic or

catabolic responses are required due to bone trauma or pathophysiologic conditions, such as

rheumatoid arthritis (RA) and osteoarthritis (OA) (Gerber, 2000). Several prior studies involving

early block in osteoblastogenesis via genetic inactivation of Runx2/Cbfa1, Osx, Indian hedgehog

or β-catenin have suggested that osteoblast differentiation of progenitor cells, up to the stage of

Runx2/Osx expression, is required for vascular invasion of cartilage and primary ossification

(Colnot, 2005; Hill, 2005; Komori, 1997; Nakashima, 2002; Otto, 1997; St-Jacques, 1999).

1.3.2 Matrix Metalloproteinases

The matrix metalloproteinases (MMPs), a family of enzymes sharing common functional

domains and activation mechanisms (Sternlight, 1999),(243) play a role in regulating cell

migration and ECM remodelling, which is essential for morphogenesis, embryonic development

and tissue remodeling (Birkedal-Hansen, 1993; Brinckerhoff, 2002; Nagase, 1999).(244-246) Bone

formation involves extensive remodelling of the extracellular matrices (ECM), and MMPs 2, 8, 9

and 13 have been implicated in this process (Chin, 1997; Gack, 1995; Johansson, 1997; Maruya,

2003; Mattot, 1995; Sasano, 2002). In addition, MMPs have also been recognized to be

important in events such as osteoclast recruitment (Blavier, 1995; Engsig, 2000; Karsdal, 2001;

Sato, 1998), coupling of bone resorption and formation (Everts, 2002), angiogenesis (Vu, 1998)

and osteoblast survival (Karsdal, 2002).

Itagaki and colleagues examined the expression of MMPs, type I collagen and

osteocalcin during bone healing in an experimental rat calvarial defect model. Their study

determined that the expression of MMP8 was highest on day 1, while type I collagen,

osteocalcin, MMP2 and MMP13 expression increased toward week 2 (Itagaki, 2008). Type I

collagen and osteocalcin mRNA transcripts were shown to be localized in osteoblasts and

Page 29: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

21

osteocytes, and the expression of MMPs 2, 8, and 13 in some of these cell types suggested that

they may play a role in remodelling of the ECM during bone healing (Itagaki, 2008).

During endochondral ossification, expression of various MMPs in the developing bone

allows the remodelling of cartilage anlage and the subsequent neovascularisation. Among the

members of MMPs thought to be important for skeletal development, MMP-9 and MMP-13 have

been shown to be two of the main proteinases involved in endochondral ossification (Inada,

2004; Stickens, 2004; Uusitalo, 2000). The critical role of these MMPs can be seen in knockout

transgenic animals that display dramatic expansion of the hypertrophic zone in the growth plate

of the primary ossification site.

Of the collagenases, MMP-13 is the most effective at cleaving type II collagen

(Billinghurst, 1997; Knauper, 1996) and its expression has been shown to be regulated by

transcription factor Runx2/Cbfa1 (Porte, 1999; Selvamurugan, 1998). A study by Wu and

colleagues, using bovine fetal chondrocytes isolated from physeal cartilages, showed an increase

in expression of transcription factor Runx2/Cbfa1, type X collagen and MMP-13 upon

differentiation of these cells into hypertrophic chondrocytes (Wu, 2002). This was followed by

increased incorporation of 45Ca2+ in the extracellular matrix associated with matrix calcification.

Inhibition of type II collagen degradation by a nontoxic carboxylate inhibitor of MMP-13

resulted in suppression of chondrocyte differentiation as seen by a repression in Runx2/Cbfa1

and type X collagen gene expression, as well as decreased incorporation of 45Ca2+ in the

extracellular matrix. Their results suggested that expression and extracellular proteolysis activity

of MMP-13 is required for chondrocyte differentiation, which is associated with matrix

mineralization (Wu, 2002). This is also seen in mice lacking Runx2/Cbfa1, where the absence of

MMP-13 expression in the growth plates results in impaired chondrocyte maturation and matrix

mineralization during endochondral development (Inada, 1999; Jimenez, 1999). In contrast,

over-expression of Runx2/Cbfa1 causes acceleration of endochondral ossification in vivo (Ueta,

2001) and promotes chondrocyte differentiation and maturation in vitro (Enomoto, 2000).

Together, these observations indicate that the Cbfa1/MMP-13 pathway is important during

endochondral bone formation by regulating chondrocyte differentiation and matrix turnover.

As previously mentioned, angiogenesis is a critical event that is needed for successful

bone development. VEGF has been identified as one of the important angiogenic factors

Page 30: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

22

expressed by hypertrophic chondrocytes and blocking VEGF activity results in a phenotype

similar to the MMP-9 null phenotype. It has previously been shown that MMP-9-null mice

exhibit an increase in hypertrophic chondrocytes and the amount of VEGF mRNA expression

(Ortega, 2003), a delay in endochondral ossification and marrow cavity formation, as well as a

decrease in chondrocyte apoptosis, vascular invasion, trabecular bone synthesis and osteoclastic

employment (Ortega, 2003). Also, in vitro angiogenesis assays of MMP-9-null mice cartilage

explants show a delayed angiogenic response (Vu, 2000). Treatment of wildtype mice with a

soluble VEGF receptor to reduce the bioavailability of VEGF displayed the same phenotypes of

MMP9-null mice (Gerber, 1999). However, the VEGF-depleted phenotype can be rescued with

the introduction of exogenous VEGF, which causes rapid remodelling of the hypertrophic

cartilage and an increase in trabecular bone. Similar rescue is seen in MMP-null mice

transplanted with wildtype bone marrow. Furthermore, MMP-9 has also been shown to be a

chemoattractant for osteoclasts and invasion of osteoclasts into the hypertrophic cartilage is

dependent on VEGF expression (Engsig, 2000; Gerber, 1999). Taken together, these

observations display a link between degradation of the cartilage anlage by MMPs, expression of

VEGF and their coordination in advancement of the endochondral process.

MMP-9 and MMP-13 are expressed in non-overlapping cell types at the edge of

ossification and together carry out most of the protease activity during endochondral

ossifications (Ortega, 2003; Vu, 1998). While MMP-9 is expressed in osteoclasts, endothelial

cells and bone marrow stromal cells, MMP13 is primarily expressed in terminal hypertrophic

chondrocytes and newly recruited osteoblasts (Inada, 2004; Stickens, 2004; Uusitalo, 2000; Vu,

1998).

Nonetheless, despite their different expression patterns, MPP-9 deficient and MMP-13

deficient mice exhibit similar phonotypic characteristics such as elongation of the hypertrophic

cartilage zone in the metaphyseal growth plates (Inada, 2004; Stickens, 2004; Vu, 1998). MMP-

13 protease is a very potent enzyme secreted under physiological conditions, which has high

preference for type I and type II collagen, major components of extracellular matrix and bone,

respectively (Knauper, 1996). Andersen and colleagues utilized various methods, such as PCR,

Northern blots, in situ hybridization, and immunohistochemistry, to look at expression pattern of

MMP-13 mRNA. Their study show that MMP-13 mRNA is expressed highly in mononucleated

cells closely surrounding the osteoclasts and in the subosteoclastic resorption zones (Andersen,

Page 31: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

23

2004). Their data strongly suggest that in the case of MMP-13, its strong affinity to type I

collagen (Gillet, 1977; Knauper, 1997) allows its accumulation in zones of collagen

demineralization and osteoclast resorptive lacunae (Delaisse, 1993). This suggests that successful

collagen matrix resorption requires the coordination between both osteoblasts and osteoclasts,

where secreted MMP-13 functions in conjunction with osteoclastic secreted enzymes such as

MMP9 for progression of the healing process.

1.3.3 Bone Morphogenetic Proteins

A family of regulatory molecules known as bone morphogenetic proteins was initially

discovered by researchers investigating the active components of the demineralized bone grafts.

With the use of rDNA technology, more highly purified forms of these proteins were isolated

and the osteoinductive capacity of these proteins was tested by injection into soft tissue. These

molecules have been used in studying the healing of fractures (Cook, 1997; den Boer, 1997;

Einhorn, 1997; Poplich, 1997; Turek, 1997), bridging critical size defects in various animal

models (Bostrom,1996; Cook, 1994; Kirker-Head, 1995; Yasko, 1992), and

enhancing/accelerating fracture healing (Cook, 1997; den Boer, 1997; Einhorn, 1997; Poplich,

1997; Turek, 1997).

To test the effect of BMP2 on fracture healing, Einhorn and colleagues performed local

percutanous injection of BMP2 into closed mid-diaphyseal femur fractures in rats (Einhorn,

1997). Data from biomechanical testing show that BMP2 treated fractures exhibited significant

increase in stiffness around 14 days post fractures and increase in strength at 28 days after

fracture. They also showed relatively earlier maturation of the osteochondrogenic line and faster

bridging of the callus, and substantially more peripheral woven bone at the fracture callus of

BMP2 treated groups as compared with the controls (Einhorn, 1997).

First to investigate the effect BMPs were Urist and colleagues, who observed induction of

ectopic bone formation upon implantation of demineralized bone in rats (Urist, 1965). Few other

studies have looked at the efficacy of injectable BMP7 for accelerating healing in small defects

or fracture gaps (Cook, 1997; den Boer, 1997; Poplich, 1997). In BMP7-treated canines new

bone formation and bridging of the defect occurred at an accelerated pace compared with the

controls. The defects treated with BMP7 also showed significantly greater strengths than those of

control limbs. In another study, den Boer investigated the effects of BMP7 on the healing of a

Page 32: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

24

closed diaphyseal fracture in the goat. Three dimensional computerized tomography (CT) scans

and dual energy x-ray absorptiometry (DEXA) analysis revealed that at early time points BMP7

treated groups developed a larger callus volume and higher bone mineral content, respectively

(den Boer, 1997). Since their discovery, bone derived BMPs (BMP2, 4, and 7) have also been

used clinically to treat difficult cases of nonunion fractures and shown to have the capacity to

enhance bone induction in humans.

Additionally, BMPs have been shown to be important in chondrogenesis. Several BMPs

have been shown to be expressed around and within the chondrogenic condensations, have the

ability to trigger ectopic chondrogenesis in adult animal models, and cause significant

enlargement of cartilages in gain-of-function experiments (Duprez, 1996; Macias, 1997; Reddi,

1994; Urist, 1965). In developing cartilage, the function of BMPs in chondrogenesis is regulated

by Noggin, a BMP antagonist (Brunet, 1998; Capdevila, 1998; Merino, 1998). Among other

functions, BMPs regulate dorsa-ventral patterning during embryogenesis (Jones, 1992), control

cell apoptosis, patterning of limb buds (Zou, 1996), as well as influencing epithelial-

mesenchymal interactions during organogenesis (Cheifetz, 1999; Vainio, 1993).

1.3.4 Transforming Growth Factor Beta

One of the major groups of growth factors, the TGF-β superfamily of polypeptides,

which play a critical role in the induction, development and repair of bone via regulation and

stimulation of mesenchymal precursor cells for chondrocytes, osteoblasts, and osteoclasts.

During fracture healing TGF-βs are released during the initial inflammatory phase from platelets

and are thought to be integral mediators of fracture repair by playing a role in initiation of callus

formation (Canalis, 1988; Celeste 1990; Joyce, 1990a; Joyce, 1990b; Sporn, 1989). Members of

this group range from the five TGFβs (TGFβ1, TGFβ2, TGFβ3, TGFβ4, and TGFβ5) to BMPs,

activins, inhibins, and growth and differentiation factors (Centrella, 1994). TGFβs are produced

by a variety of cells in different tissues, but have a significantly higher concentration in bone and

are expressed by osteocytes, osteoblasts, osteoclasts and chondrocytes (Centrella, 1994;

Ellingsworth, 1986; Robey, 1987; Sandberg, 1993). By utilizing various experimental

techniques, such as immunohistochemistry, in situ hybridization and northern blot analysis, the

role of TGFβ in association with cartilage hypertrophy and calcification of the endochondral

ossification front has been previously determined (Joyce, 1990a; Joyce, 1990b).

Page 33: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

25

In osteoblasts, TGFβ is stored as high molecular weight precursor molecule complexes

that need to go through post-translational processing prior to being secreted as an inactive

product, which gets incorporated into the mineralized bone matrix (Bonewald, 1994; Dallas,

1998; Miyazono, 1993). They are activated in an extremely acidic pH environment, which is

created by osteoclasts when they degrade the bone matrix by acid hydrolysis. This in turn

stimulates the new bone formation via the stimulating action of TGFβ on osteoblasts during

development or different stages of fracture healing (Oreffo, 1990). In fact, a majority of in vitro

studies have indicated that TGFβ functions on osteoblasts by increasing the expression of a

variety of osteoblast differentiation markers such as ALP, Type I collagen, and osteonectin

(Harris, 1994; Ingram, 1994; Massaque, 1990; Sandberg, 1993; Strong, 1991; Tally-Ronsholdt,

1995; Wergedal, 1992). TGFβ also acts to induce production of cartilage components including

Type II, III, V, VI, and X collagen, fibronectin, and proteoglycans (Harris, 1994; Massaque,

1990; Sandberg, 1993), while preventing the digestion of these extracellular matrix

macromolecules by having inhibiting effects on the action of metalloproteinases (Harris, 1994).

After secretion TGFβs are incorporated into the mineralized bone matrix where through a

feedback mechanism they regulate the presence of proteases/plasminogens that activate latent

TGFβs (Yee, 1993). Overall, TGFβ acts as a potent chemotactic agent during skeletal

development and the net effect of TGFβ can be seen in different stages of bone formation such as

its stimulation of collagen synthesis, enhancement of osteoblast proliferation and regulation of

bone matrix production.

1.3.5 Wnt Signalling and Bone Formation

Wnt molecules, a family of secreted glycoproteins, play a central role in countless

embryonic developmental processes as well as regulating homeostatsis of different tissues in

later stages of growth. During embryonic development they play a role in determining patterning

and wnt signaling regulates homeostasis during growth (Cadigan, 1997). Precise regulation of

wnt-mediated effects is necessary for successful development since fluctuations of this pathway

leads to a variety of diseases and developmental defects (Cadigan, 1997). The signalling pathway

is initiated by binding of the wnt ligand to one of its 10 Frizzled (Fz) receptor family members

on the cell surface, inducing one of the three distinct pathways that have been previously

elucidated (Glass, 2006a; Katoh, 2002). The most studied canonical wnt/β-catenin pathway plays

a crucial role in bone formation and homeostasis. This pathway is regulated at multiple steps

Page 34: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

26

including modulations at the extracellular level, at the cell membrane, in the cytosol and in the

nucleus, which ultimately results in the intracellular accumulation and consecutive translocation

of the β-catenin protein to the nucleus. Once in the nucleus, association of β-catenin with the

lymphoid-enhancer binding facter (Lef)/T-cell specific transcription factors (Tcf’s) leads to

expression of target genes (Westendorf, 2004). This pathway is regulated extracellularly by

secreted Dickkopf (DKK) protein inhibitors (Davidson, 2002; Niehrs, 2006) or secreted frizzled-

related proteins (sFRPs)/Wnt inhibitory factor-1 (Wif-1) that prevent the induction of the wnt

signalling by binding to the coreceptor LRP5/6 or binding to the wnt ligands themselves,

respectively (Bafico, 2001; Mao, 2002; Schweizer, 2003).

Another alternative wnt pathway includes the non-canonical pathway that relies on

intracellular release of calcium for regulating cell migration, dorso-ventral patterning of the

embryo, as well as heart development (Kuhl, 2004). The planar cell polarity (PCP) is the third

wnt pathway that regulates cytoskeletal architecture, actin dynamics and cellular migration via

coactivation of two small GTPases, Rho and Rac, as well as the stress-activated protein kinase

JNK (Habas, 2003; Kaltschmidt, 2002; Katoh, 2005). The activation of these signaling

transduction pathways is known to contribute to regulation of cell proliferation and apoptosis,

cell-cell adhesion, as well as being involved in embryonic morphogenesis (Davis, 2000).

The non-canonical Wnt5a and Wnt5b are expressed in chondrogenic regions of the limb

during development where they exhibit opposite activities in regulating longitudinal growth

(Bradley, 2011; Guo, 2008; Yang, 2003). Wnt5a regulates the transition from articular/resting

chondrocytes zone to the highly proliferating and hypertrophic chondrocyte zones, while Wnt5b

counteracts the activity of Wnt5a by inhibiting chondrocyte hypertrophy (Yang, 2003). These

opposite effects of Wnt5a and Wnt5b may result from their modulation of cell cycle effectors

cyclin D1 and p130, and Sox9-controlled Col2a1 expression (Yang, 2003). Therefore, the wnt

signalling pathways have been established as a dominant regulator of the stages of bone

formation, ranging from regulating the differentiation of precursor cells down either

chondrogenic or osteogenic lineages, and controlling the process of matrix degradation during

the remodelling phase of soft callus (Glass, 2006a, Glass, 2006b) to being involved in normal

bone homeostasis via expression by osteocytes (Kramer, 2010).

Page 35: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

27

1.4 Thymidine Kinase/Ganciclovir System

A predominant component of the extracellular matrix, type I collagen is synthesized by

an array of connective tissue cells that display characteristic regulation of these genes under a

variety of different circumstances (Pavlin, 1992). Analysis of the regions responsible for

regulation of the collagen promoters by local growth factors, systemic hormones and viral

transformation (Choe, 1987; Liau, 1985; Rossi, 1988; Walsh, 1987), have identified a number of

cis-acting elements and trans-acting factors involved in regulating the tissue-specific expression

of type I collagen (Ramirez, 1990; Vuorio, 1990). Among these, the COL1A1 and COL1A2

promoter sequences have been shown to regulate the tissue-specific collagen expression in

transfected cells (Boast, 1990; Karsenty, 1990; Rippe, 1989; Schmidt, 1986) and in transgenic

animals (Khillan, 1986; Slack, 1991).

Numerous studies have provided evidence for regulatory mechanisms that are unique for

the osseous tissue producing cells such as osteoblasts and odontoblasts, as compared to many

fibroblastic cells. For example, the rate of collagen synthesis measured in primary cell cultures

(Luben 1976; McCarthy 1988; Wong 1982), fetal rat calvariae in organ culture (Canalis 1980;

Rowe, 1982), and embryonic tissues (Diegelmann, 1971; Moen 1979) show a higher basal rate of

collagen production in osteoblasts than in fibroblastic cells. Secondly, collagen synthesis in

osteoblasts can be regulated by several hormones such as vitamin D, PTH (Kream 1980;

Lichteler 1989; Rowe, 1982), insulin and insulin-like growth factor-I (Canalis, 1980; Kream

1985), while fibroblasts are unresponsive to these hormones. Furthermore, fibroblastic cells

produce a variety of collagen types (I, III, IV, V and VI), whereas fully differentiated osteoblasts

almost exclusively synthesize type I collagen (Fessler, 1981; Liau, 1985; Olsen, 1989;

Pihlajaniemi, 1989). Finally, a retroviral insertion in the first intron of the COL1A1 gene of

MOV13 mouse, does not affect collagen transcription in osteoblasts while resulting in a

complete block of collagen transcription in fibroblasts (Kratochwil, 1989). This ability to

override the inhibitory effect of the retroviral insertion suggest the presence of a cis-active DNA

sequence within the COL1A1 gene in osteoblast and odontoblasts that differ from those used in

fibroblastic cells.

The modular organization of the Col1a1 promoter allows selection of specific domains

for use in different type I collagen-producing tissues (Bedalov, 1995; Bogdanovic, 1994;

Page 36: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

28

Rossert, 1995). For expression of the transgene in the osteoblast layer lining newly formed

calvarial bone, a homeodomain binding TAAT sequence localized between the -1670 and -1683

base pairs have been shown to be important (Dodig, 1996). The 3.6-kilobase (kb) rat Col1a1

promoter have previously been used in conjunction with the chloramphenicol acetyl transferase

(CAT) gene, and shown high level of transgene expression in bone, tendon, and developing tooth

germ with lower levels in skin (Pavlin, 1992). Truncation of the 3.6-kb promoter to 2.3-kb

fragment caused no change in the level of CAT activity in calvaria while a 2- to 4-fold decrease

in the transgene expression was observed in tendon and a greater loss in the skin (Bogdanovic,

1994; Krebsbach, 1993). In other studies, similar cell-specific pattern of expression was detected

in transgenic mice harboring a 2.3-kb human COL1A1 promoter driving the expression of

bovine growth hormone (Liska, 1994), as well as in mice carrying a murine 2.3-kb Col1a1

promoter fragment fused to either the β-galactosidase or luciferase gene (Rossert, 1995).

One method for studying the lineage relationships and cell functions of a population of

differentiating cells is the use of tissue-specific transgenic expression of herpes simplex virus

thymidine kinase (HSV-tk) to conditionally deplete a specific cell population (Heyman, 1989).

Targeted cell ablation using this strategy requires high level synthesis of enzyme in a subset of

rapidly proliferating cells. The expression of the transgene can be directed to restricted cell types

via the use of tissue-specific promoters and enhancers that allow the physiological manipulation

of the cell and organs of interest (Hammer, 1985; Palmiter, 1982). The HSV-tk, although not

deleterious in mammalian cells by itself, becomes toxic in the presence of nucleoside analogs

like ganciclovir (GCV), leading to inhibition of DNA synthesis and cell ablation (Heyman,

1989). This is a useful approach in evaluating the contribution of a cell type to a particular

developmental program, while allowing one to control the precise timing of the toxic insult, the

degree of ablation, and the potential for recovery upon termination of drug-induced toxicity

(Heyman, 1989). Upon administration and delivery of specific nucleoside analogs to target cells,

the HSV-tk is capable of monophosphorylating the benign substrate. The nucleoside

monophosphate is further phosphorylated by cellular kinases to nucleoside triphosphate,

producing a toxic product that becomes incorporated into DNA during differentiation and leads

to inhibition of DNA synthesis and cell ablation (Elion, 1977; Furman, 1980; Fyfe, 1978). This

ablation technique has been utilized previously using an immunoglobulin promoter/enhancer to

target the expression of HSV-tk to the lymphoid system (Heyman, 1989). In transgenic animals

Page 37: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

29

the activity of the enzyme was restricted to the spleen, lymph nodes, bone marrow and thymus,

where the expression of HSV-tk resulted in massive depletion of B and T lymphocytes after drug

treatment (Heyman, 1989).

Following the initial use of this method for the haematopoietic and immune cell

pathways, strategies have been developed to study the relationship of different cell types in bone

tissue. In regards with osteoblasts, the use of a highly tissue specific promoter, such as

osteocalcin, leads to the expression of the tk gene late in the osteoblast lineage when cell division

has ceased and the rate of cell proliferation is reduced (Corral, 1998). Previous use of the mouse

osteocalcin gene 2 (OG2) promoter, a marker of mature osteoblasts, to drive expression of the

HSV-tk gene, showed a nearly complete absence of cells with morphologic features of

osteoblasts (Corral, 1998). Due to late tk expression in the OG2 mice, osteoclast activity was still

persistent and bone resorption remained unaffected despite a reduction in the osteoblast

population. This would suggest that the surviving osteoblast progenitors that are unaffected by

the GCV treatment in OG2-tk mice could express signals that would regulate osteoclast function

(Corral, 1998).

In contrast to the osteocalcin promoter, the modular design of the Col1a1 promoter

allows the creation of promoter-reporter constructs that can be used in various type I collagen-

producing tissues (Dodig, 1996). In an in vitro study by Dacic and colleagues (Dacic, 2001)

different Col1a1 promoter fragments were used to drive the chloramphenicol acetyltransferase

(CAT) gene. In this model, the activity of a 3.6 Col1a1 fragment was detected in osteoblast

progenitors concurrent with type I collagen mRNA and ALP expression. In contrast, the 2.3

Col1a1 fragment became active coincident with expression of the early bone-specific marker

bone sialoprotein (BSP) but distinctly prior to osteocalcin mRNA, which is produced at a later

stage in the osteoblast lineage (Dacic, 2001). In another study, Visnjic and colleagues generated

transgenic mice that express the HSV-tk gene under 2.3-kb fragments of the collagen type I

promoter, which was found to be active in concurrent with other genetic markers of early

differentiating osteoblast (Visnjic, 2001). Treatment of these mice with GCV resulted in an

extensive destruction of the bone lining cells, decreased osteoclast numbers and a decrease in

bone marrow elements. The reduction in the bone marrow cellularity was more prominent in the

metaphyseal regions and areas adjacent to the endosteal surface of the diaphyseal bone, which

normally house cells of osteoblastic lineage. Termination of the GCV treatment resulted in a

Page 38: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

30

pronounced response of new cortical and trabecular bone formation as well as the replenishment

of the bone marrow cells. Their findings suggest that early differentiating osteoblasts are

necessary for the maintenance of haematopoiesis and osteoclastogenesis (Visnjic, 2001).

Therefore, in contrast to the other osteoblast specific promoters mentioned, the 2.3-kb fragment

of type I collagen promoter provides a useful osteoblast-specific model to define and analyze the

interrelation of osteoblasts with the other bone cell types during the different modes of bone

formation and fracture healing.

Page 39: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

31

Figure 1: Genetic Control of Skeletal Cell Type Differentiation.

(A) Differentiation of mesenchymal progenitor cells down an osteogenic or chondrogenic lineage and expression of the transcription factors and cytokines regulating each pathway is shown. Transcription factor Runx2/Cbfa1 is the earliest marker of osteoblast differentiation and is also involved in promoting hypertrophic differentiation of chondrocytes. Sox9 transcription factor, along with Sox5 and Sox6 regulate the expression of various genes involved in chondrogenesis and induction of cartilage matrix proteins such as type II collagen. (Refer to ‘Abbreviation List’, for acronyms).

(B) Molecular regulation of osteoclast differentiation from haematopoietic stem cells and expression of enzymes, secreted molecules, and transcription factors acting at different stages of differentiation/activation are depicted. The activation of osteoclast involves the formation of ruffled membrane and secretion of catabolic enzymes such as cathepsin K, TRAP and H+ into resorption pits for degradation of bone matrix. (Refer to ‘Abbreviation List’ for acronyms).

Page 40: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

32

Figure 2: Bone Formation Occurs Through Two Different Modes of Ossification Processes.

(A) The endochondral bone formation begins with the condensation of mesenchymal progenitor cells at sites of future skeletal development, a process that is recapitulated during the early stages of fracture repair. The cells at the central region of the aggregate then differentiate along a chondrogenic lineage and produce a cartilaginous template. In the region adjacent to the hypertrophic chondrocyte (HTC), the first osteoblast precursors differentiate and secrete a non-mineralized extracellular matrix that becomes organized into compact mineralized bone. During this process osteoblasts that become entrapped in the bone differentiate into osteocytes. Osteoclasts are required for remodelling and resorption of the bone matrix.

(B) Intramembranous ossification involves the direct differentiation of osteoblasts from the condensed mesenchyme at the site of bone formation such as the flat bones of the skull. In this process bone is made without the need for an intermediate cartilaginous template.

(Fig 2A adapted from Hartmann, 2006 and Fig 2B adapted from Hartmann, 2009)

Page 41: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

33

Figure 3: Model of Fracture Healing Stages and Cellular Participants Involved During the Process of Repair.

(A) Representative images of a fracture healing model. The process of fracture healing is partitioned into four overlapping stages. Upon damage to the bone there is an immediate inflammation response followed by the formation of a soft callus, which undergoes remodeling prior to the hard callus formation stage. The final stage of repair involves the remodeling of the hard callus back into the original cortical bone configuration.

(B) Diagram showing the corresponding cellular contributors to the healing process at various stages.

(Adapted from Schindeler, 2008. 19; 460).

Page 42: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

34

References

Adams CS, Shapiro IM. 2002. The fate of the terminally differentiated chondrocyte: evidence for microenvironmental regulation of chondrocyte apoptosis. Crit Rev Oral Biol Med 13(6): 465-473.

Adams GB, Scadden DT. 2006. The hematopoietic stem cell in its place. Nat Immunol 7: 333-337.

Akiyama H, Chaboissier MC, Martin JF, Schedl A, de Crombrugghe B. 2002. The transcription factor Sox9 has essential roles in successive steps of the chondrocyte differentiation pathway and is required for expression of Sox5 and Sox6. Genes Dev 16(21): 2713-2828.

Al-Aql ZS, Alagl AS, Graves DT, Gerstenfeld LC, Einhorn TA. 2008. Molecular mechanisms controlling bone formation during fracture healing and distraction. Osteogenesis. J Dent Res 87: 107-18.

Alagiakrishnan K, Juby A, Hanely D, Tymchak W, Sclater A. 2003. Role of vascular factors in osteoporosis. J Gerontol A Biol Sci Med Sci 58: 362-366.

Alcobendas M, Baud CA, Castanet J. 1991. Structural changes of the preosteocytic area in Vipera aspis (L.) (Ophidia, Viperidae) bone tissue in various physiological conditions. Calc Tissue Int 49(1): 53-57.

Allgower M, Speigel PG. 1979. Internal fixation of fractures: evolution of concepts. Clin Orthop 138: 26-29.

Althoff J, Quint P, Krefting ER, Hohling HJ. 1982. Morphological studies on the epiphyseal growth plate combined with biochemical and X-ray microprobe analyses. Histochemistry 74: 541-552.

Andersen TL, del Carent Ovejero M, Kirkegaard T, Lehhard T, Foged NT, Delaisse JM. 2004. A scrutiny of matrix metalloproteinases in osteoclasts: Evidence for heterogeneity and for the presence of MMPs synthesized by other cells. Bone 35:110-1119.

Arai F, Hirao A, Ohmura M, Sato H, Matsuoka S, Takubo K, Ito K, Koh GY, Suda T. 2004. Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell 118: 149-161.

Aubin JE, Liu F. 1996. The osteoblast lineage. Principles of Bone Biology 51-67.

Aubin JE. 1998. Advances in the osteoblast lineage. Biochem Cell Biol 76(6): 899-910.

Augustin G, Antabak A, Davila S. 2007. The periosteum. Part 1: Anatomy, histology and molecular biology. Injury 38(10): 1115-1130.

Bafico A, Liu G, Yaniv A, Gazit A, Aaronson SA. 2001. Novel mechanism of Wnt signaling inhibition mediated by Dickkopf-1 interaction with LRP6/Arrow. Nature Cell Biol 3(7): 683-686.

Barnes GL, Kostenuik PJ, Gerstenfeld LC, Einhorn TA. 1999. Growth factor regulation of fracture repair. J Bone Miner Res 14: 1805-15.

Bedalov A, Salvatori R, Dodig M, Kronenberg MS, Kapural B, Bogdanovic Z, Kream BE, Woody CO, Clark SH, Mack K. 1995. Regulation of COL1A1 expression in type I

Page 43: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

35

collagen producing tissues: Identification of a 49 base pair region which is required for transgene expression in bone of transgenic mice. J Bone Miner Res 10: 1443-1451.

Beertsen W, Brekelmans M, Everts V. 1978. The site of collagen resorption in the periodontal ligament of the rodent molar. Anat Rec 192: 305-318.

Behonick DJ, Xing Z, Lieu S, Buckley JM, Lotz JC, Marcucio RS, Werb Z, Miclau T, Colnot C. 2007. Role of matrix metalloproteinase 13 in both endochondral and intramembranous ossification during skeletal regeneration. PLoS One 2: e1150.

Belanger LF. 1977. The skeletal tissues. In: Weiss L, Greep RO, editors. Histology, 4th ed. New York: McGraw-Hill Book Company.

Bi W, Deng JM, Zhang Z, Behringer RR, de Crombrugghe B. 1999. Sox9 is required for cartilage formation. Nat Genet 22: 85-89.

Bialek P, Kern B, Yang X, Schrock M, Sosic D, Hong N, Wu H, Yu K, Ornitz DM, Olson EN, Justice MJ, Karsenty G. 2004. A twist code determines the onset of osteoblast differentiation. Dev Cell 6: 423-435.

Bianco P, Fisher LW, Young MF, Termine JD, Robey PG. 1991. Expression of bone sialoprotein (BSP) in developing human tissues. Calcif Tissue Int 49: 421-426.

Billinghurst RC, Dahlberg L, Ionescu M, Reiner A, Bourne R, Rorabeck C, Mitchell P, Hambor J, Diekmann O, Tschesche H, Chen J, van Wart H, Poole AR. 1997. Enhanced cleavage of type II collagen by collagenases in osteoarthritic articular cartilage. J Clin Invest 99: 1534-1545.

Birkedal-Hansen H, Moore WG, Bodden MK, Windsor LJ, Birkedal-Hansen B, Decarlo A, Engler JA. 1993. Matrix metalloproteinases: a review. Crit Rev Oral Biol Med 4: 197-250.

Blair HC, Teitelbaum SL, Ghiselli R, Gluck S. 1989. Osteoclastic bone-resorption by a polarized vacuolar proton pump. Science 245(4920): 855-857.

Blavier L, Delaisse JM. 1995. Matrix metalloproteinases are obligatory for the migration of preosteoclasts to the developing marrow cavity of primate long bones. J Cell Sci 108: 3649-3659.

Boast S, Su MW, Ramirez F, Sanchez M, Avvedimento EV. 1990. Functional analysis of cis-acting DNA sequences controlling transcription of the human type I collagen genes. J Biol Chem 265: 13351-13356.

Bogdanovic Z, Bedalov A, Krebsbach PH, Pavlin D, Woody CO, Clark SH, Thomas HF, Rowe DW, Kream BE, Lichtler AC. 1994. Upstream regulatory elements necessary for expression of the rat COL1A1 promoter in transgenic mice. J Bone Miner Res 9: 285-292.

Bolander ME. 1992. Regulation of fracture repair by growth factors. Proc Soc Exp Biol Med 200(2): 165-170.

Bonewald LF, Dallas SL. 1994. Role of active and latent transforming growth factor beta gene in bone formation. J Cell Biochem 55: 350-357.

Page 44: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

36

Bostrom MPG, Lane JM, Tomin E, Browne M, Berberian W, Turek T, Smith J, Wozney J, Schildhauer T. 1996. The use of bone morphogenetic protein-2 in the rabbit ulnar nonunion model. Clin Orthop 327: 272-282.

Boyle WJ, Simonet WS, Lacey DL. 2003. Osteoclast differentiation and activation. Nature 423: 337-342.

Bradley EW, Drissi MH. 2011. Wnt5b regulates mesenchymal cell aggregation and chondrocyte differentiation through the planar cell polarity pathway. J Cell Physiol 226: 1683-1693.

Brighton CT, Lorich DG, Kupcha R, Reilly TM, Jones AR, Woodbury RA, 1992. The pericyte as a possible osteoblast progenitor cell. Clin Orthop Relat Res 275: 287-299.

Brinckerhoff, CE and Matrisian LM. 2002. Matrix metalloproteinases: a tail of a frog that became a prince. Nat Rev Mol Cell Biol 3: 207-214.

Brunet LJ, McMahon JA, McMahon AP, Harland RM. 1998. Noggin, cartilage morphogenesis, and joint formation in the mammalian skeleton. Science 280: 1455-1457.

Bucay N, Sarosi I, Dunstan CR, Morony S, Tarpley J, Capparelli C, Scully S, Tan HL, Xu W, Lacey DL, Boyle WJ, Simonet WS. 1998. Osteoprotegerin-deficeint mice develop early onset osteoporosis and arterial calcification. Genes Dev 12: 1260-1268.

Burger EH, Klein-Nulend J, Smit TH. 2003. Strain-derived canalicular fluid flow regulates osteoclast activity in a remodelling osteon: a proposal. J Biomech 36: 1453-1459.

Burkhardt R, Kettner G, Bohm W, Schmidmeier M, Schlag R, Frisch B, Mallmann B, Eisenmenger W, Gilg T. 1987. Changes in trabecular bone, hematopoiesis and bone marrow vessels in aplastic anemia, primary osteoporosis, and old age: a comparative histomorphometric study. Bone 8: 157-164.

Cadigan KM, Nusse R. 1997. Wnt signaling: A common theme in animla development. Genes Dev 11: 3286-3305.

Canalis E. 1980. Effect of insulin-like growth factor I on DNA and protein synthesis in cultured rat calvaria. J Clin Invest 66: 709-719.

Canalis E, McCarthy T, Centrella M. 1988. Growth factors and the regulation of bone remodeling. J Clin Invest 81: 277-281.

Cao Y, Mori S, Mashiba T, Westmore MS, Ma L, Sato M, Akiyama T, Shi L, Komatsubara S, Miyamoto K, Norimatsu H. 2002. Raloxifene, estrogen, and alendronate affect the processes of fracture repair differently in ovariectomized rats. J Bone Miner Res 17: 2237-2246.

Capdevila J, Johnson RL. 1998. Endogenous and ectopic expression of noggin suggests a conserved mechanism for regulation of BMP function during limb and somite patterning. Dev Biol 197(2): 205-217.

Carmeliet P, Collen D. 2000. Molecular basis of angiogenesis. Role of VEGF and VE-cadherin. Ann N Y Acad Sci 902: 249-262.

Carmeliet P. 2003. Angiogenesis in health and disease. Nature Med 9(6): 653-660.

Carter DR, Beaupre GS, Giori NJ, Helms JA. 1998. Mechanobiology of skeletal regeneration. Clinical Orthop Relat Res Suppl. 355: S41-S55.

Page 45: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

37

Castanon I, Baylies MK. 2002. A Twist in fate: evolutionary comparison of Twist structure and function. Gene 287: 11-22.

Celeste AJ, Iannazzi JA, Taylor RC, Hewick RM, Rosen V, Wang EA, Wozney JM. 1990. Identification of transforming growth factor beta family members present in bone-inductive protein purified from bovine bone. Proc Natl Acad Sci USA 87: 9843-9847.

Centrella M, Horowitz MC, Wozney JM, McCarthy TL. 1994. Transforming growth factor-beta gene family members and bone. Endocr Rev 15: 27-39.

Chambers TJ, Fuller K. 1985a. Bone cells predispose bone surfaces to resorption by exposure of mineral to osteoclastic contact. J Cell Sci 76: 155-165.

Chambers TJ, Darby JA, Fuller K. 1985b. Mammalian collagenase predisposes bone surfaces to osteoclastic resorption. Cell Tissue Res 241: 671-675.

Cheifetz S. 1999. BMP receptors in limb and tooth formation. Crit Rev Oral Biol Med 10(2): 182-198.

Chen J, Shapiro HS, Sodek J. 1992. Development expression of bone sialoprotein mRNA in rat mineralized connective tissues. J Bone Miner Res 7: 987-997.

Chen Y, Alman BA. 2009. Wnt pathway, an essential role in bone regeneration. J Cell Biochem 106(3): 353-362.

Childs SC. 2005. The future of bone healing. Clin Podiatr Med Surg 22: 631-641.

Chin JR, Werb Z. 1997. Matrix metalloproteinases regulate morphogenesis, migration and remodeling of epithelium, tongue skeletal muscle and cartilage in the mandibular arch. Development 124: 1519-1530.

Cho TJ, Gerstenfeld LC, Einhorn TA. 2002. Differential temporal expression of members of the transforming growth factor beta superfamily during murine fracture healing. J Bone Miner Res 17: 513-520.

Choe I, Aycock RS, Raghow R, Myers JC, Seyer JM, Kang AH. 1987. A hepatic fibrogenic factor stimulates the synthesis of types I, III and V procollagens in cultured cells. J Biol Chem 262: 5408-5413.

Cole AA, Wlaters LM. 1987. Tartrate-resistant acid phosphatise in bone and cartilage following decalcification and cold-embedding in plastic. J Histochem Cytochem 35: 203-206.

Collin-Osdoby P. 1994. Role of vascular endothelial cells in bone biology. J Cell Biochem 55: 304-309.

Colnot C, Thompson Z, Miclau T, Werb Z, Helms JA. 2003. Altered fracture repair in the absence of MMP9. Development 130: 4123-4133.

Colnot C, de la Fuente L, Huang S, Hu D, Lu C, St-Jacques B, Helms JA. 2005. Indian hedgehog synchronizes skeletal angiogenesis and perichondrial maturation with cartilage development. Development 132: 1057-1067.

Cook SD, Baffes GC, Wolfe MW, Sampath TK, Rueger DC. 1994. Recombinant human bone morphogenetic protein-7 induces healing in a canine long-bone segmental defect model. Clin Orthop 301: 302-312.

Page 46: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

38

Cook SD. 1997. Acceleration of bone healing with OP-1 in a canine noncritical size defect model. Second International OP-1 Conference, Boston, MA. Abstract

Corral DA, Amling M, Priemel M, Loyer E, Fuchs S, Ducy P, Baron R, and Karsenty G. 1998. Dissociation between bone resorption and bone formation in osteopenci transgenic mice. Proc Natl Acad Sci. USA 95: 13835-13840.

Cottler-Fox MH, Lapidot T, Petit I, Kollet O, DiPersio JF, Link D, Devine S. 2003. Stem cell mobilization. Hematology (Am Soc Hematol Educ Program) 419-437.

D’Souza RN, Aberg T, Gaikwas J, Cavender A, Owen M, Karsenty G, Thesleff I. 1999. Cbfa1 is required for epithelial-mesenchymal interactions regulating tooth development in mice. Development 126: 2911-2920.

Dacic S, Kalajzic I, Visnjic D, Lichtler AC, Rowe DW. 2001. Col1a1-driven transgenic markers of osteoblast lineage progression. J Bone Miner Res 16(7): 1228-1236.

Dallas SL, Park-Synder S, Miyazono K, Twardzik D, Mundy GR, Bonewald LF. 1998. Characterization and autoregulation of latent transforming growth factor beta complexes in osteoblast-like cell lines. J Biol Chem 269: 6815-6821.

Davidson G, Mao BY, Barrantes ID, Niehrs C. 2002. Kremen proteins interact with Dickkopf1 to regulate anteroposterior CNS patterning. Development 129(24): 5587-5596.

Davis RJ. 2000. Signal transduction by the JNK group of MAP kinases. Cell 103(2): 239-252.

de Saint-Georges L, Miller SC, Bowman BM, Jee WS. 1989. Ultrastructural features of osteoclasts in situ. Scanning Microsc 3: 1201-1206.

DeLacure MD. 1994. Physiology of bone healing and bone grafts. Otolaryngol Clin N Am 27(5): 859-874.

Delaisse JM, Boyde A, Maconnachie E, Ali NN, Sear CHJ, Eeckhout Y, Vaes G, Jones SJ. 1987. The effects of inhibitors of cysteine-proteinases and collagenase on the resorptive activity of isolated osteoclasts. Bone 8: 305-313.

Delaisse JM, Eeckhout Y, Neff L, Francois-Gillet C, Henriet P, Su Y, Vaes G, Baron R. 1993. (Pro)collagenase (matrix metalloproteinase-1) is present in rodent osteoclasts and in the underlying bone-resorbing compartment. J Cell Sci 106: 1071-1082.

Dellavalle A, Sampaolesi M, Tonlorenzi R, Tagliafico E, Sacchetti B, Perani L, Innocenzi A, Galvez BG, Messina G, Morosetti R, Li S, Belicchi M, Peretti G, Chamberlain JS, Wright WE, Torrente Y, Ferrari S, Bianco P, Cossu G, 2007. Pericytes of human askeletal muscle are myogenic precursors distinct from satellite cells. Nat Cell Biol 9: 255-267.

den Boer FC, Bramer JAM, Blokhius TJ, Van Soest EJ, Jenner JM, Patka P, Bakker FC, Burger EH, Haarman HJ. 1997. The effect of recombinant human osteogenic protein-1 on the healing of fresh closed diaphyseal fracture. Second International OP-1 Conference, Boston MA. Abstract.

Desbois C, Seldin MF, Karsenty G. 1994. Localization of the osteocalcin gene-cluster on mouse chromosome-3. Mamm Genome 5(5): 321-322.

Diegelmann RF, Peterkofsky B. 1971. Collagen biosynthesis during connective tissue development in the chick embryo. Dev Biol 28: 443-453.

Page 47: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

39

Dodds RA, Connor JR, James IE, Rykaczewski EL, Appelbaum E, Dul E, Gowen M. 1995. Human osteoclasts, not osteoblasts, deposit osteopontin onto resorptin surfaces: An in vitro and ex vivo study of remodelling. J Bone Miner Res 10: 1666-1680.

Dodig M, Kronenberg MS, Bedalov A, Kream BE, Gronowicz G, Clark SH, Mack K, Liu YH, Maxon R, Pan ZZ, Upholt WB, Rowe DW, Lichtler AC. 1996. Identification of a TAAT-containing motif required for high level expression of the COL1A1 promoter in differentiated osteoblasts of transgenic mice. J Biol Chem 271: 16422-16429.

Doherty MJ, Ashton BA, Walsh S, Beresford JN, Grant ME, Canfield AE, 1998. Vascular pericytes express osteogenic potential in vitro and in vivo. J Bone Miner Res 13: 828-838.

Doty SB, Nunez EA. 1985. Activation of osteoclasts and the repopulation of bone surfaces following hibernation in the bat, Myotis lucifugus. Anat Rec 213: 481-495.

Ducy P, Desbois C, Boyce B, Pinero G, Story B, Dunstan C, Smith E, Bonadio J, Goldstein S, Gundberg C, Bradley A, Karsenty G. 1996. Increased bone formation in osteocalcin-deficient mice. Nature 382(6590): 448-452.

Ducy P, Zhang R, Geoffroy V, Ridall AL, Karsenty G, 1997. Osf2/Cbfa1: a transcriptional activator of osteoblast differentiation. Cell 89: 747-754.

Ducy P, Karsenty G. 1998. Genetic control of cell differentiation in the skeleton. Curr Opin Cell Biol 10: 614-619.

Ducy P. 2000. Cbfa1: A molecular switch in osteoblast biology. Dev Dyn 219:461-471.

Duprez D, Bell EJ, Richardson MK, Archer CW, Wolpert L, Brickell PM, Francis-West PH. 1996. Overexpression of BMP-2 and BMP-4 alters the size and shape of developing skeletal elements in the chick limb. Mech Dev 57(2): 145-157.

Einhorn TA, Majeska RJ, Rush EB, Levine PM, Horowtiz MC. 1995. The expression of cytokine activity by fracture callus. J Bone Miner Res 10: 1272-1281.

Einhorn TA, Majeska RJ, Oloumi G. 1997. Enhancement of experimental fracture healing with a local percutaneous injection of rhBMP-2. American Academy of Orthopaedic Surgeons Annual Meeting. San Francisco CA 64: 216. Abstract

Einhorn TA. 1998. The cell and molecular biology of fracture healing. Clin Orthop Relat Res 355: S7-21.

Einhorn T, Lee C. 2001. Bone regeneration: new findings and potential clinical applications. J Am Acad Orthop Surg 9: 157-165.

Ekanayake S, Hall BK. 1987. The development of acellularity of the vertebral bone of the Japanese Medaka, Oryzias latipes (Teleostei, Cyprinidontidae). J Morph 193: 253-261.

Ekanayake S, Hall BK. 1988. Ultrastructure of the osteogenesis of acellular vertebral bone in the Japanese Medaka, Oryzias latipes (Teleostei, Cyprinidontidae). Am J Anat 182: 241-249.

Elion GB, Furman PA, Fyfe JA, de Miranda P, Beauchamp L, and Schaeffer HJ. 1977. Selectivity of action of an anti-herpetic agent, 9-(2-Hydroxyethoxymethyl) Guanine. Proc Natl Acad Sci USA. 74: 5716-5720.

Ellingsworth LR, Brennan JE, Fok K, Rosen DM, Bentz H, Piez KA, Seyedin SM. 1986. Antibodies to the N-terminal portion of cartilage-inducing factor A and transforming

Page 48: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

40

growth factor beta: Immunohistochemical localization and association with differentiating cells. J Biol Chem 261: 12362.12367.

Engsig MT, Chen QJ, Vu TH, Pedersen AC, Therkidsen B, Lund LR, Henriksen K, Lenhard T, Foged NT, Werb Z, Delaisse JM. 2000. Matrix metalloproteinase 9 and vascular endothelial growth factor are essential for osteoclast recruitment into developing long bones. J Cell Biol 151: 879-890.

Enomoto H, Enomoto-Iwamoto M, Iwamoto M, Nomura S, Himeno M, Kitamura Y, Kishimoto T, Komori T. 2000. Cbfa1 is a positive regulatory factor in chondrocyte maturation. J Biol Chem 275: 8695-8702.

Enomoto H, Shiojiri S, Hoshi K, Furuichi T, Fukuyama R, Yoshida CA, Kanatani N, Nakamura R, Mizuno A, Zanma A, Yano K, Yasuda H, Higashio K, Takada K, Komori T. 2003. Induction of osteoclast differentiation by Runx2 through receptor activator of nuclear factor-kappa B ligand (RANKL) and osteoprotegerin regulation and partial rescue of osteoclastogenesis in Runx2-/- mice by RANKL transgene. J Biol Chem 278: 23971-23977.

Erlebacher A, Filvaroff EH, Ye JQ, Derynck R. 1998. Osteoblastic responses to TGF-beta during bone remodelling. Mol Biol Cell 9: 1903-1918.

Everts V, Delaisse JM, Korper W, Jansen DC, Tiggelaar-Gutter W, Saftig P, Beertsen W. 2002. The bone lining cell: its role in cleaning Howship’s lacunae and initiating bone formation. J Bone Miner Res 17: 77-90.

Ferguson C, Alpern E, Miclau T, Helms JA. 1999. Does adult fracture repair recapitulate embryonic skeletal formation? Mech Dev 87: 57-66.

Ferrara N, Gerber HP. 2001. The role of vascular endothelial growth factor in angiogenesis. Acta Haematol 106: 148-156.

Fessler LI, Robinson WJ, Fessler JH. 1981. Biosynthesis of procollagen [(proα1 V)2 (proα2 V)] by chick tendon fibroblasts and procollagen (proα1 V)3 by hamster lung cell cultures. J Biol Chem 256: 9646-9651.

Franz-Odendaal TA, Hall BK, Witten PE. 2006. Buried alive: How osteoblasts become osteocytes. Dev Dyn 235(1): 176-190.

Franzoso G, Carlson L, Xing L, Poljak L, Shores EW, Brown KD, Leonardi A, Tran T, Boyce BF, Siebenlist U. 1997. Requirement for NF- κB in osteoclast and B-cell development. Genes Dev 11: 3482-3496.

Frost HM. 1960. In vivo osteocyte death. J Bone Joint Surg 42A: 138-143.

Frost HM. 1989. The biology of fracture healing. And overview for clinicians. Part I. Clin Orthop 248: 283-293.

Furman PA, McGujirt PV, Keller PM, Fyfe JA and Elion GB. 1980. Inhibition by acyclovir of cell-growth and DNA-synthesis of cells biochemically transformed with herpesvirus genetic information. Virology 102: 420-430.

Fyfe JA, Keller PM, Furman PA, Miller RL and Elion GB. 1978. Thymidine kinase from herpes-simplex virus phosphorylates the new anti-viral compound, 9-(2-Hydroxyethoxymethyl) Guanine. J Biol Chem 253: 8721-8727.

Page 49: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

41

Gack S, Vallon R, Schmidt J, Grigoriadis A, Tuckermann J, Schenkel J, Weiher H, Wagner EF, Angel P. 1995. Expression of interstitial collagenase during skeletal development of the mouse is restricted to osteoblast-like cells and hypertrophic chondrocytes. Cell Growth Differ 6: 759-767.

Garant PR. 1976. Collagen resorption by fibroblasts. A theory of fibroblastic maintenance of the periodontal ligament. J Periodontol 47: 380-390.

Gerber HP, Vu TH, Ryan AM, Kowalski J, Werb Z, Ferrara N. 1999. VEGF couples hypertrophic cartilage remodelling, ossification, and angiogenesis during endochondral bone formation. Nat Med 5: 623-628.

Gerber HP, Ferrara N. 2000. Angiogenesis and bone growth. Trends Cardiovasc Med 10: 223-228.

Gerstenfeld LC, Shapiro FD. 1996. Expression of bone-specific genes by hypertrophic chondrocytes: implication of the complex functions of the hypertrophic chondrocyte during endochondral bone development. J Cell Biochem 62: 1-9.

Gerstenfeld LC, Cullinane DM, Barnes GL, Graves DT, Einhorn TA. 2003. Fracture healing as a post-natal developmental process: molecular, spatial, and temporal aspects of its regulation. J Cell Biochem 88(5): 873-884.

Gerstenfeld LC, Alkhiary YM, Krall EA. 2006. Three-dimensional reconstruction of fracture callus morphogenesis. J Histochem Cytochem 54(11): 1215-1228.

Gerstenfeld LC, Sacks DJ, Pelis M, Mason ZD, Graves DT, Barrero M, Ominsky MS, Kostenuik PJ, Morgan EF, Einhorn TA. 2009. Comparison of effects of the bisphosphonate alendronate versus the RANKL inhibitor denosumab on murine fracture healing. J Bone Miner Res 24: 196-208.

Gillet C, Eeckhout Y, Vaes G. 1977. Purification of procollagenase and collagenase by affinity chromatography on Sepharose-collagen. FEBS Lett 74:126-128.

Giannoudis PV, Jones E, Einhorn TA. 2011. Fracture healing and bone repair. Injury 42(6):549-550.

Glass DA 2nd, Karsenty G. 2006a. Canonical Wnt signaling in osteoblasts is required for osteoclast differentiation. Ann N Y Acad Sci 1068: 117-130.

Glass DA 2nd, Karsenty G. 2006b. Molecular bases of the regulation of bone remodeling by the canonical Wnt signaling pathway. Curr Top Dev Biol 73: 43-84.

Goto T, Yamaza T, Tanaka T. 2003. Cathepsins in the osteoclast. J Electron Micros 52(6): 551-558.

Green E, Lubahn JE, Evans J. 2005. Risk factors, treatment, and outcomes associated with non-union of the midshaft humerus fracture. J Surg Orhop Adv 14(2): 64-72.

Guo J, Jin J, Cooper LF. 2008. Dissection of sets of genes that control the character of wnt5a-deficient mouse calvarial cells. Bone 43: 961-971.

Habas R, Dawid IB, He X. 2003. Coactivation of Rac and Rho by Wnt/Frizzled signaling is required for vertebrate gastrulation. Genes Dev 17(2): 295-309.

Habuchi H, Conrad HE, Glaser JH. 1985. Coordinate regulation of collagen and alkaline phosphatase levels in chick embryo chondrocytes. J Biol Chem 260: 13029-13034.

Page 50: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

42

Hadjidakis DJ, Androulakis II. 2006. Bone remodeling. Ann NY Acad Sci 1092: 385-396.

Hall BK. 1988. The embryonic development of bone. Am Sci 76: 174-181.

Hall BK, Miyake T. 1992. The membranous skeleton: the role of cell condensations in vertebrate skeletogenesis. Anat Embryol (Berl) 186: 107-124.

Hall BK, Miyake T. 1995. Divide, accumulate, differentiate: cell condensation in skeletal development revisited. Int J Dev Biol 39: 881-893.

Hall BK, Witten PE. 2005. The origin and plasticity of skeletal tissues in vertebrate evolution and development. In: Anderson JS, Sues H-D, editors. Major transitions in vertebrae evolution. Festschrift for Dr. Robert L. Carroll. Bloomington, IN: Indiana University Press. 1-53.

Hall AP, Westwood FR, Wadsworth PF. 2006. Review of the effects of anti-angiogenic compounds on the epiphyseal growth plate. Toxicol Pathol 34: 131-147.

Haller AC, Zimny ML. 1978. Effects of hibernation on interradicular alveolar bone. J Dent Res 56: 1552-1557.

Hammer RE, Brinster RL, Rosenfeld MG, Evans RM and Mayo KE. 1985. Expression of human growth hormone-releasing factor in transgenic mice results in increased somatic growth. Nature 315: 413-416.

Han Y, Lefebvre V. 2008. L-Sox5 and Sox6 drive expression of the aggrecan gene in cartilage by securing binding of Sox9 to a far-upstream enhancer. Mol Cell Biol 28(16): 4999-5013.

Harris WH, Heaney RP. 1969. Skeletal renewal and metabolic bone diseases. N Engl J Med 280: 193-202, 253-259,303-311.

Harris SE, Bonewald LF, Harris MA, Sabatini M, Dallas S, Feng JQ, Chosh-Choudhury N, Wozney J, Mundy GR. 1994. Effects of transforming growth factor beta on bone nodule formation and expression of bone morphogenetic protein 2, osteocalcin, osteopontin, alkaline phosphatase, and type I collagen mRNA in long-term cultures of fetal rat calvarial osteoblasts. J Bone Miner Res 9: 855-863.

Hartmann C. 2006. A Wnt canon orchestrating osteoblastogenesis. Trends Cell Biol 16(3): 151-158.

Hartmann C. 2009. Transcriptional networks controlling skeletal development. Curr Opin Genet Dev 19(5): 437-443.

Hauschka PV, Wians FH. 1989. Osteocalcin-hydroxyapatite interaction in the extracellular organic matrix of bone. Anat Rec 224(2): 180-188.

Hayden JM, Mohan S, Baylink DJ. 1995. The insulin-like growth factor system and the coupling of formation to resorption. Bone 17: 93S-98S.

Heissig B, Tejeda R, Friedrich M, Werb Z, Rafii S, Hattori K. 2002a. MMP9 mediated activation and release of pro-angiogenic factors following irradiation. Blood 100(11): 124 Abstract.

Heissig B, Hattori K, Dias S, Friedrich M, Ferris B, Hackett NR, Crystal RG, Besmer P, Lyden D, Moor MAS, Werb Z, Rafii S. 2002. Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligand. Cell 109: 625-637.

Page 51: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

43

Hermey DC, Popoff SN, Marks SC Jr. 1996a. Reduce bone resorption in toothless (osteopetrotic) rats: An abnormally of osteoblasts related to their inability to activate osteoclast activity in vitro. Connet Tissue Res 35: 273-278.

Hermey DC, Popoff SN, Marks SC Jr. 1996b. Differential gene expression in bone from toothless (osteopetrotic) rats. J Bone Miner Res 11: S652-S652.

Heyman RA, Borreli E, Lesley J, Anderson D, Richman DD, Baird SM, Hyman R, Evans RM. 1989. Thymidine kinase obliteration: creation of transgenic mice controlled immune deficiency. Proc Natl Acad Sci USA 86(8): 2698-2702.

Hill PA. 1998. Bone remodeling. Br J Orthod 25: 101-107.

Hill TP, Spater D, Taketo MM, Birchmeier W, Hartmann C. 2005. Canonical Wnt/beta-catenin signaling prevents osteoblasts from differentiating into chondrocytes. Dev Cell 8: 727-738.

Hofbauer LC, Khosla S, Dunstan CR, Lacey DL, Boyle WJ, Riggs BL. 2000. The roles of osteoprotegerin and osteoprotegerin ligand in the paracrine regulation of bone resorption. J Bone Miner Res 15: 2-12.

Holliday LS, Welgus HG, Gliszar CJ, Veith GM, Jeffrey JJ, Gluck SL. 1997. Initiation of osteoclast bone resorption by interstitial collagenase. J Biol Chem 272: 22053-22058.

Holtrop ME. 1990. Light and electron microscopical structure of bone forming cells. In: Hall BK, editor. Bone, Vol. 1: the osteoblast and osteocyte. Caldwell, NJ: The Telford Press. 1-40.

Hui M, Li SQ, Holmyard D, Cheng P. 1997. Stable transfection of nonosteogenic cell lines with tissue nonspecific alkaline phosphatase enhances mineral deposition both in the presence and absence of beta-glycerophosphate: Possible role for alkaline phosphatase in pathological mineralization. Calcif Tissue Int 60: 467-472.

Ingram RT, Bonde SK, Riggs BL, Fitzpatrick LA. 1994. Effects of transforming growth factor beta and 1,25 dihydroxyvitamin D3 on the function, cytochemistry, and morphology of normal osteoblast-like cells. Differentiation 55: 153-163.

Inada M, Yasui T, Nomura S, Miyake S, Deguchi K, Himeno M, Sato M, Yamagiwa H, Kimua T, Yasui N, Ochi T, Endo N, Kitamura Y, Kishimoto T, Komori T. 1999. Maturational disturbance of chondrocytes in Cbfa1-deficeint mice. Dev Dyn 214: 279-290.

Inada M, Wang Y, Byrne MH, Rahman MU, Miyaura C, Lopez-Otin C, Krane SM, 2004. Critical roles for collagenase-3 (Mmp13) in development of growth plate cartilage and in endochondral ossification. Proc Natl Acad Sci USA 101: 17192-17197.

Ishida N, Hayashi K, Hoshijima M, Ogawa T, Koga S, Miyatake Y, Kumegawa M, Kimura T, Takeya T. 2002. Large scale gene expression analysis of osteoclastogenesis in vitro and elucidation of NFAT2 as a key regulator. J Biol Chem 277: 41147-41156.

Itagaki T, Honma T, Takahashi I, Echigo S, and Sasano Y. 2008. Auantitative analysis and localization of mRNA transcripts of type I collagen, osteocalcin, MMP2, MMP8, and MMP13 during bone healing in a rat calvarial experimental defect model. Anat Rec 291: 1038-1046.

Page 52: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

44

Iwaki A, Jingushi S, Oda Y, Izumi T, Shida JI, Tsuneyoshi M, Sugioka Y. 1997. Localization and quantification of proliferating cells during rat fracture repair: detection of proliferating cell nuclear antigen by immunohistochemistry. J Bone Miner Res 12: 96-102.

Iyama K, Ninomiya Y, Olsen BR, Linsenmayer TF, Trelstad RL, Hayashi M. 1991. Spatiotemporal pattern of type X collagen gene expression and collagen deposition in embryonic chick vertebrae undergoing endochondral ossification. Anat Rec 229(4): 462-472.

Jilka RL, Weinstein RS, Bellido T, Parfitt AM, Manolagas SC. 1998. Osteoblast programmed cell death (apoptosis): modulation by growth factors and cytokines. J Bone Miner Res 13(5): 793-802.

Jilka RL, Weinstein RS, Parfitt AM, Manolagas SC. 2007. Quantifying osteoblast and osteocyte apoptosis: Challenges and rewards. J Bone Miner Res 22(10): 1492-1501.

Jimenez MJG, Balbin M, Lopez JM, Alvarez J, Komori T, Lopez-Orin C. 1999. Collagenase-3 is a target of Cbfa1, a transcription factor of the runt gene family involved in bone formation. Mol Cell Biol 19: 4431-4442.

Johansson N, Saarialho-Kere U, Airola K, Herva R, Nissinen L, Westermarck J, Vuorio E, Heino J, Kahari VM. 1997. Collagenase-3 (MMP13) is expressed by hypertrophic chondrocytes, periosteal cells, and osteoblasts during human fetal bone development. Dev Dyn 208: 387-397.

Jonason JH, Xiao G, Zhang M, Xing L, Chen D. 2009. Post-translational regulation of Runx2 in bone and cartilage. J Dent Res 88(8): 693-703.

Jones SJ, Boyde A, Ali NN. 1986. The interface of cells and their matrices in mineralized tissues: A review. Scan Electron Microsc 4: 1555-1569.

Jones CM, Lyons KM, Lapan PM, Wright CV, Hogan BL. 1992. DVR-4 (bone morphogenetic protein-4) as a posterior-ventralizing factor in Xenopus mesoderm induction. Development 115, 639-647.

Joyce ME, Jingushi S, Bolander ME. 1990a. Transforming growth factor-beta in the regulation of fracture repair. Orthop Clin North Am 21: 199-209.

Joyce ME, Terek RM, Jingushi S, Bolander ME. 1990b. Role of transforming growth factor-beta in fracture repair. Ann N Y Acad Sci 593: 107-123.

Kahn AJ, Patridge NC. 1987. New concepts in bone remodelling: An expanding role for the osteoblast. Am J Otolaryngol 8: 258-264.

Kaltschmidt JA, Lawrence N, Morel V, Balayo T, Fernandez BG, Pelissier A, Jacinto A, Martinez Arias A. 2002. Planar polarity and actin dynamics in the epidermis of Drosophila. Nat Cell Biol 4: 937-944.

Karaplis AC, Luz A, Glowaski J, Bornson RT, Tybulewicz VL, Kronenberg HM, Mulligan RC. 1994. Lethal skeletal dysplasia from targeted disruption of the parathyroid hormone-related peptide gene. Genes Dev 8: 277-289.

Karsdal MA, Fjording MS, Foged NT, Delaisse JM, Lochter A. 2001. Transforming growth factor-beta-induced osteoblast elongation regulates osteoclastic bone resorption through a

Page 53: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

45

p38 mitogen0activated protein kinase- and matrix metalloproteinase-dependent pathway. J Biol Chem 276: 39350-8.

Karsdal MA, Larsen L, Engsig MT, Lou H, Ferreras M, Lochter A, Delaisse JM, Foged NT. 2002. MMP dependent activation of latent TGF-beta controls the conversion of osteoblasts into osteocytes by blocking osteoblast apoptosis. J Biol Chem 277(46): 44061-44068.

Karsenty G, de Crombrugghe B. 1990. Two different negative and one positive regulatory factors interact with a short promoter segment of the α1(I) collagen gene. J Biol Chem 265: 9934-9942.

Karsenty G and Wangner EF. 2002. Reaching a genetic and molecular understanding of skeletal development. Dev Cell 2: 389-406.

Katoh M. 2002. Wnt and FGF gene clusters (Review). Int J Oncol 21(6): 1269-1273.

Katoh M. 2005. Wnt/PCP signaling pathway and human cancer (Review). Oncol Rep 14(6): 1583-1588.

Kauffman JM, Taelman P, Vermeulen A, Vandeweghe M. 1992. Bone mineral status in growth hormone-deficient males with isolated and multiple pituitary deficiencies of childhood onset. J Clin Endocrinol Metab 74: 118-123.

Ketenjian AY, Arsenis C. 1975. Morphological and biochemical studies during differentiation and calcification of fracture callus cartilage. Clin Orthop Relat Res 107: 266-273.

Khillan JS, Schmidt A, Overbeek PA, de Crombrugghe B, Westphal H. 1986. Developmental and tissue-specific expression directed by the α2 type I collagen promoter in transgenic mice. Proc Natl Acad Sci USA 83: 725-729.

Kim IS, Otto F, Abel B, Mundlos S, 1999. Regulation of chondrocyte differentiation by Cbfa1. Mech Dev 80: 159-170.

Kim HJ, Kim JH, Bae SC, Choi JY, Kim HJ, Ryoo HM. 2003. The protein kinase C pathway plays a central role in the fibroblast growth factor-stimulated expression and transactivation activity of Runx2. J Biol Chem 278: 319-326.

Kirker-Head CA, Gerhart TN, Schelling SH, Hennig GE, Wang E, Holtrop ME. 1995. Long-term healing of bone using recombinant human bone morphogenetic protein 2. Clin Orthop 318: 222-230.

Knauper V, Lopez-Otin C, Smith B, Knight G, Murphy G. 1996. Biochemical characterization of human collagenase-3. J Biol Chem 271: 1544-1550.

Knauper V, Cowell S, Smith B, Lopez-Otin C, O’Shea M, Morris H, Zardi L, Murphy G. 1997. The role of the C-terminal domain of human collagenase-3 (MMP-13) in the activation of procollagense-3, substrate specificity, and tissue inhibitor of metalloproteinase interaction. J Biol Chem 272: 7608-7616.

Knothe Tate ML, Adamson JR, Tami AE, Bauer TW. 2004. The osteocyte. Int J Biochem Cell Biol 36: 1-8.

Kollet O, Shivitel S, Chen YQ, Suriawinata J, Thung SN, Dabeva MD, Kahn J, Spiegel A, Dar A, Samira S, Goichberg P, Kalinkovich A, Arenzana-Seisdedos F, Nagler A, Hardan I,

Page 54: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

46

Revel M, Shafritz DA, Lapidot T. 2003. HGF, SDF-1, and MMP-9 are involved in stress-induced human CD34+ stem cell recruitment to the liver. J Clin Invest 112(2): 160-169.

Kollet O, Dar A, Shivtiel S, Kalinkovich A, Lapid K, Sztainberg Y, Tesio M, Samstein RM, Goichberg P, Spiegel A, Elson A, Lapidot T. 2006. Osteoclasts degrade endosteal component and promote mobilization of hematopoietic progenitor cells. Nat Med 12(6): 657-664.

Komatsubara S, Mori S, Mashiba T, Nonaka K, Seki A, Akiyama T, Miyamoto K, Cao Y, Manabe T, Norimatsu H. 2005. Human parathyroid hormone (1-34) accelerates the fracture healing process of woven to lamellar bone replacement and new cortical shell formation in rat femora. Bone 36: 678-687.

Komori T, Yagi H, Nomura S, Yamaguchi H, Sasaki K, Deguchi K, Shimizu Y, Bronson RT, Gao YH, Inada M, Sato M, Okamoto R, Kitamura Y, Yoshiki S, Kishimoto T. 1997. Targeted disruption of Cbfa1 results in a complete lack of bone formation owing to maturational arrest of osteoblasts Cell 89: 755-764.

Kosaki N, Takaishi H, Kamekura S, Kimura T, Okada Y, Minqi L, Amizuka N, Chung UI, Nakamura K, Kawagucchi H, Toyama Y, D’Armiento J. 2007 Impaired bone fracture healing in matrix metalloproteinase-13 deficient mice. Biochem Biophys Res Commun 354: 846-851.

Koyama E, Leatherman JL, Shimazu A, Nah HD. 1995. Syndecan-3, tenascin-C, and the development of cartilaginous skeletal elements and joints in chick limbs. Dev Dyn 203: 152-162.

Koyama E, Shimazu A, Leatherman JL, Golden EB, Nah HD, Pacifici M. 1996. Expression of syndecan-3 and tenascin-C: possible involvement in periosteum development. J Orthop Res 14: 403-412.

Kramer I, Halleux C, Keller H, Pequrri M, Gooi JH, Weber PB, Feng JQ, Bonewald LF, Kneissel M. 2010. Osteocyte Wnt/beta-catenin signaling is required for normal bone homeostasis. Mol Cell Biol 30(12): 3071-3085.

Kratochwill, K, von der Mark K, Kollar EJ, Jaenisch R, Mooslehner K, Schwarz M, Haase K, Gmachi I, Harbers K. 1989. Retrovirus-induced insertional mutation in Mov13 mice affects collagen I expression in a tissue-specific manner. Cell 57: 807-816.

Kream BE, Rowe DW, Gworek G, Raisz LG. 1980. Parathyroid hormone alters collagen synthesis and procollagen mRNA levels in fetal rat calvaria. Proc Natl Acad Sci USA 77: 5654-5658.

Kream BE, Simth MD, Canalis E, Raisz LG. 1985. Characterization of the effect of insulin on collagen synthesis in fetal rat bone. Endocrinology 116: 1922-1928.

Krebsbach PH, Harrison JR, Lichtler AC, Woody CO, Rowe DW, Kream BE. 1993. Transgenic expression of COL1A1-cholarmphenicol acetyltransferase fusion genes in bone: Differential utilization of promoter elements in vivo and in cultured cells. Mol Cell Biol 13: 5168-5174.

Kronenberg HM. 2003. Developmental regulation of the growth plate. Nature 15: 423(6937): 332-336.

Page 55: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

47

Kuhl M. 2004. The Wnt/calcium pathway: biochemical mediators, tools and future requirements. Front Biosci 9: 967-974.

Kwiecinski GG. 1985. Bone remodelling and its regulation in Myotis lucifugus. Prog Zool 30: 45-47.

Kwiecinski GG, Krook L, Wimsatt WA. 1987. Annual skeletal changes in the little brown bat, Myotis lucifungus, with particular references to pregnancy and lactation. Am J Anat 178: 410-420.

Lacey DL, Timms E, Tan HL, Kelley MJ, Dunstan CR, Burgess T, Elliott R, Colombero A, Elliott G, Scully S, Hsu H, Sullivan J, Hawkins N, Davy E, Capparelli C, Eli A, Qian YX, Kaufman S, Sarosi I, Shalhoub V, Senaldi G, Guo J, Delaney J, Boyle WJ. 1998. Osteoprotegerin (OPG) ligand is a cytokine that regulates osteoclasts differentiation and activation. Cell 93(2): 165-176.

Lanske B, Karaplis AC, Lee K, Luz A, Vortkamp A, Pirro A, Karperien M, Defize LHK, Ho C, Mulligan RC, Defize LH, Ho C, Mulligan RC, Abou-Samra AB, Juppner H, Segre GV, Kronenberg HM. 1996. PTh/PThrP receptor in early development and Indian hedgehog-regulated bone growth. Science 273: 663-666.

Lapidot T, Dar A, Kollet O. 2005. How do stem cells find their way home? Blood 106: 1901-1910.

Lazzarini L, De Lalla F, Mader JT. 2002. Long bone osteomyelitis. Curr Infect Dis Rep 4: 439-445.

Le AX, Miclau T, Hu D, Helms JA. 2001. Molecular aspects of healing in stabilized and non-stabilized fractures. J Orthop Res 19: 78-84.

Lee MH, Javed A, Kim JH, Shin JI, Gutierrez S, Choi JY, Rosen JL, van Wijnen AJ, Stein GS, Lian JB, Ryoo HM. 1999. Transient upregulation of Cbfa1 in response to bone morphogenetic protein-2 and transforming growth factor beta1 in C2C12 myogenic cells coincides with suppression of the myogenic phenotype but is not sufficient for osteoblast differentiation. J Cell Biochem 73: 114-125.

Lee MH, Kim YJ, Kim HJ, Park HD, Kang AR, Kyung HM, Sung JH, Wozney JM, Kim HJ, Ryoo HM. 2003. BMP-2-induced Runx2 expression is mediated by Dlx5, and TGF-beta 1 opposes the BMP-2-induced osteoblast differentiation by suppression of Dlx5 expression. J Biol Chem 278(36): 34387-34394.

Lefebvre V, Huang W, Harley VR, Goodfellow PN, de Crombrugghe B. 1997. Sox9 is a potent activator of the chondrocyte-specific enhancer of the pro alpha1 (II) collagen gene. Mol Cell Biol 17(4): 2336-2346.

Lefebvre V, de Crombrugghe B. 1998a. Toward understanding Sox9 function in chondrocyte differentiation. Matrix Biol 16(9): 529-540.

Lefebvre V, Li P, de Crombrugghe B. 1998b. A new long form of Sox5 (L-Sox5), Sox6 and Sox9 are coexpressed in chondrogenesis and cooperatively activate the type II collagen gene. EMBO J 17(19): 5719-5733.

Lerner UH. 2000. Osteoclast formation and resorption. Matrix Biol 19: 107-120.

Page 56: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

48

Levesque JP, Hendy J, Takamatsu Y, Simmons PJ, Bendall LJ. 2003. Disruption of the CXCR4/CXCL12 chemotactic interaction during hematopoietic stem cell mobilization induced by G-CSF or cyclophosphamide. J Clin Invest 111: 187-196.

Li J, Mori S, Kaji Y, Mashiba T, Kawanishi J, Norimatsu H. 1999. Effect of bisphosphonate (incadronate) on fracture healing of long bones in rats. J Bone Miner Res 14: 969-979.

Li L, Cserjesi P, Olsen BR. 1995. Dermo-1: a novel twist-related bHLH protein expressed in the developing dermis. Dev Biol 172: 280-292.

Li M, Amizuka N, Oda K, Tokunaga K, Ito T, Takeuchi K, Takagi R, Maeda T. 2004. Histochemical evidence of the initial chondrogenesis and osteogenesis in the periosteum of a rib fractured model: implications of osteocyte involvement in periosteal chondrogenesis. Microsc Res Tech 64: 330-342.

Lian JB, McKee MD, Todd AM, Gerstenfeld LC. 1993. Induction of bone-related proteins, osteocalcin and osteopontin, and their matrix ultrastructural localization with development of chondrocyte hypertrophy in vitro. J Cell Biochem 52: 206-219.

Liau G, Yamada Y, de Crombrugghe B. 1985. Coordinate regulation of the levels of type III and type I collagen mRNA in most but not all mouse fibroblasts. J Biol Chem 260: 531-536.

Lichteler A, Stover ML, Angily J, Kream B, Rowe DW. 1989. Isolation and characterization of the rat α(1) collagen promoter. J Biol Chem 264: 3072-3077.

Liska DJ, Reed MJ, Sage EH, Bornstein P 1994 Cell-specific expression of alpha 1(I) collagen-hGH minigenes in transgenic mice. J Cell Biol 125: 695–704.

Liu Z, Luyten FP, Lammens J, Degueker J. 1999. Molecular signalling in bone fracture healing and distraction osteogenesis.Histol Histopathol 14(2): 587-595.

Lopez E, Macintyre I, Martelly E, Lallier F, Vidal B. 1980. Paradoxical effect of 1,25-dihydroxycholecalciferol on osteoblastic and osteoclastic activity in the skeleton of the eel Anguilla Anguilla L. Calc Tissue Int 32: 83-87.

Macias D, Garian Y, Sampath TK, Piedra ME, Ros MA, Hurle JM. 1997. Role of BMP-2 and OP-1 (BMP-7) in programmed cell death and skeletogenesis during chick limb development. Development 126(6): 1109-1117.

Madeddu P. 2005. Therapeutic angiogenesis and vasculogenesis for tissue regeneration. Exp Physiol 90: 315-326.

Madison M, Martin RB. 1993. Fracture healing, in Operative Orthopaedics (Chapman, M. W., ed.), Lippincott, Philadelphia, 221-228.

Maes C, Carmeliet P, Moermans K, Stockmans I, Smets N, Collen D, Bouillon R, Carmeliet G. 2002. Impaired angiogenesis and endochondral bone formation in mice lacking the vascular endothelial growth factor isoforms VEGF164 and VEGF188. Mech Dev 111: 61-73.

Manolagas SC. 2000. Birth and death of bone cells: basic regulatory mechanisms and implications for pathogenesis and treatment of osteoporosis. Endocrinol Rev 21: 115-137.

Mao B, Wu W, Davidson G, Marhold J, Li M, Mechler BM, Delius H, Hoppe D, Stannek P, Walter C, Glinka A, Niehrs C. 2002. Kremen proteins are Dickkopf receptors that regulate Wnt/beta-catenin signaling. Nature 417(6889): 664-667.

Page 57: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

49

Mark MP, Butler WT, Prince CW, Finkelman RD, Ruch JV. 1988. Developmental expression of 44-kDa bone phosphoprotein (osteopontin) and bone gamma-carboxyglutamic acid (Gla)-containing protein (osteocalcin) in calcifying tissues of rat. Differentiation 37: 123-136.

Marotti G. 1996. The structure of bone tissues and the cellular control of their deposition. Italian J Anat Embryol 101: 25-79.

Marsell R, Einhorn TA. 2009. The role of endogenous bone morphogenetic proteins in normal skeletal repair. Injury 40(3): S4-7.

Marsell R, Einhorn TA. 2011. The biology of fracture healing. Injury 42: 551-555.

Marsh DR, Li G. 1999. The biology of fracture healing: optimising outcome. Br Med Bull 55(4): 856-869.

Martin TJ, Ng KW. 1994. Mechanisms by which cells of the osteoblast lineage control osteoclast formation and activity. J Cell Biochem 56: 357-366.

Martin JF, Sims NA. 2005. Osteoclast-derived activity in the coupling of bone formation to resorption. Trends Mol Med 11: 76-81.

Maruya Y, Sasano Y, Takahashi I, Kagayama M, Mayanagi H. 2003. Expression of extracellular matrix molecules, MMPs and TIMPs in alveolar bone, cementum and periodontal ligaments during rat tooth eruption. J Electon Microsc 52: 593-604.

Maruyama Z, Yoshida CA, Furuichi T, Amizuka N, Ito M, Fukuyama R, Miyazaki T, Kitaura H, Nakamura K, Fujita T, Kanatani N, Moriishi T, Yamana K, Liu W, Kawaguchi H, Nakamura K, Komori. 2007. Runx2 determines bone maturity and turnover rate in postnatal bone development and is involved in bone loss in estrogen deficiency. Dev Dyn 236: 1876-1890.

Marzia M, Sims NA, Voit S, Migliaccio S, Taranta A, Bernardini S, Faraggiana T, Yoneda T, Mundy GR, Boyce BF, Baron R, Teti A. 2000. Decreased c-Src expression enhances osteoblast differentiation and bone formation. J Cell Biol 151: 311.320.

Massaque J. 1990. The transforming growth factor-beta family. Ann Rev Cell Biol 6: 597-609.

Mattot V, Raes MB, Henriet P, Eeckhout Y, Stehelin D, Vandenbunder B, Desbiens X. 1995. Expression of interstitial collagenase is restricted to skeletal tissue during mouse embryogenesis. J Cell Sci 108 (pt2): 529-535.

McCarthy TL, Centrella M, Canalis E. 1988. Further biochemical and molecular characterization of primary rat parietal bone cell cultures. J Bone Miner Res 3: 401-406.

McCulloch CAG, Heersche JNM. 1988. Lifetime of the osteoblast in mouse periodontium. Anat Rec 222(2): 128-135.

McDonald MM, Dulai S, Godfrey C, Amanat N, Sztynda T, Little DG. 2008. Bolus or weekly zoledronic acid administration does not delay endochondral fracture repair but weekly dosing enhances delays in hard callus remodeling. Bone 43: 653-662.

McKee MD, Glimcher MJ, Nanci A. 1992. High-resolution immunolocalization of osteopontin and osteocalcin in bone and cartilage during endochondral ossification in the chicken tibia. Anat Rec 234: 479-492.

McKibbin B. 1978. The biology of fracture healing in long bones. J Bone Joint Surg Br 60B(2): 150-162.

Page 58: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

50

Merino R, Ganan Y, Macias D, Economides AN, Sampath KT, Hurle JM. 1998. Morphogenesis of digits in the avian limb is controlled by FGFs, TGFβs, and Noggin through BMP signaling. Dev Biol 200: 35-45.

Mersell R, Einhorn TA. 2011. The biology of fracture healing. Injury 42: 551-555.

Meunier FJ. 1989. The acellularisation process in Osteichthyan bone. Prog Zool 35: 443-445.

Miyauchi A, Alvarez J, Greefield EM, Teti A, Grano M, Colucci S, Zambonin-Zallone A, Ross FP, Teitelbaum SL, Cheresh D, Hruska KA. 1991. Recongnition of osteopontin and related peptides by an alpha v beta 3 integrin stimulates immediate cell signals in osteoclasts. J Biol Chem 266: 20369-20374.

Miyazono K, Ichijo H, Heldin CH. 1993. Transforming growth factor beta: Latent forms, binding proteins, and receptors. Growth Factors 8: 11-22.

Moen RC, Rowe DW, Palmiter RD. 1979. Regulation of procollagen synthesis during the development of chick embryo calvaria. J Biol Chem 254: 3526-3530.

Mohan S, Baylink DJ. 1991. The role of IGF-II in the coupling of bone formation to resorption. In modern concepts of insulin-like growth factors (ed. Spencer, EM) 19-174.

Mori K, Kitazawa R, Kondo T, Maeda S, Yamaguchi A, Kitazawa S, 2006. Modulation of mouse RANKL gene expression by Runx2 and PKA pathway. J Cell Biochem 98: 1629-1644.

Mountziaris PM, Mikos AG. 2008. Modulation of the inflammatory response for enhanced bone tissue regeneration. Tissue Eng B Rev 14(2): 179-186.

Mundlos S, Olsen BR. 1997a. Heritable diseases of the skeleton. Part I: Molecular insights into skeletal development-transcription factors and signalling pathways. FASEB J 11: 125-132.

Mundlos S, Olsen BR. 1997b. Heritable diseases of the skeleton. Part II: Molecular insights into skeletal development-matrix components and their homeostasis. FASEB J 11: 227-233.

Nagase H, Woessner JF Jr. 1999. Matrix metalloproteinases. J Biol Chem 274: 21491-21494.

Nakamura H, Sato G, Hirata A, Yamamoto T. 2004. Immunolocalization of matrix metalloproteinase-13 on bone surface under osteoclasts in rat tibia. Bone 34: 48-56.

Nakamura A, Ly C, Cipetic M, Sims NA, Vieusseux J, Kartsogiannis V, Bouralexis S, Saleh H, Zhou H, Price JT, martin TJ, Ng KW, Gillespie MT, Quinn JM. 2007. Osteoclast inhibitory lectin (OCIL) inhibits osteoblast differentiation and function in vitro. Bone 40: 305-315.

Nakashima K, Zhou X, Kunkel G, Zhang Z, Deng JM, Behringer RR, de Crombrugghe B. 2002. The novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formation. Cell 108: 17-29.

Nefussi JR, Sautier JM, Nicolas V, Forest N. 1991. How osteoblasts become osteocytes: a decreasing matrix forming process. J Biol Buccale 19: 75-82.

Nemeth GG, Bolander ME, Martin GR. 1988. Growth factors and their role in wound and fracture healing. Prog Clin Biol Res 266:1-17.

Page 59: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

51

Neumann E, Schett G. 2007. Bone metabolism: molecular mechanisms. Z Rheumatol 66: 286-289.

Niehrs C. 2006. Function and biological roles of the Dickkopf family of Wnt modulators. Oncogene 25(57): 7469-7481.

Noble BS, Stevens H, Loveridge N, Reeve J. 1997. Identification of apoptotic changes in osteocytes in normal and pathological human bone. Bone 20: 273-282.

Nomura S, Wills AJ, Edwards DR, Heath JK, Hogan BL. 1989. Expression of genes for non-collagenous proteins during embryonic bone formation. Connect Tissue Res 21: 31-39.

Olsen DR, Peltonen J, Jaakkola S, Chu ML, Uitto J. 1989. Collagen gene expression by cultured human skin fibroblasts. Abundant steady-state levels of type VI procollagen messenger RNAs. J Clin Invest 83: 791-795.

Oreffo RO, Bonewald L, Kukita A, Garrett IR, Seyedin SM, Rosen D, Mundy GR. 1990. Inhibitory effects of the bone-derived growth factors osteoinductive factor and transforming growth factor-beta on isolated osteoclasts. Endocrinology 126: 3069-3075.

Ortega N, Behonick D, Stickens D. 2003. How proteases regulate bone morphogenesis. Ann N Y Acad Sci 995, 109-116.

Otto F, Thornell AP, Crompton T, Denzel A, Gilmour KC, Rosewell IR, Stamp GW, Beddinton RS, Mundlos S, Olsen BR, Selby PB, Owen MJ. 1997. Cbfa1, a candidate gene for cleidocranial dysplasia syndrome, is essential for osteoblast differentiation and bone development. Cell 89: 765-771.

Oursler MJ. 1994. Osteoblast synthesis and secretion and activation of latent transforming growth factor β. J Bone Miner Res 9: 443-452.

Palmiter RD, Brinster RL, Hammer RE, Trumbauer ME, Rosenfeld MG, Birnberg NC and Evans RM. 1982. Dramatic growth of mice that develop from eggs micro-injected with metallothioneine-growth hormone fusion genes. Nature 300: 611-615.

Palumbo C, Palazzini S, Zappe D, Marotti G. 1990a. Osteocyte differentiation in the tibia of newborn rabbits: an ultrastructural study of the formation of cytoplasmic processes. Acta Anat 137: 350-358.

Palumbo C, Palazinni S, Marotti G. 1990b. Morphological study of intercellular junctions during osteocyte differentiation. Bone 11, 401-406.

Pape HC, Giannoudis PV, Grimme K, van Griensven M, Krettek C. 2002. Effects of intramedullary femoral fracture fixation: what is the impact of experimental studies in regards to the clinical knowledge? Shock 18(4): 291-300.

Parfitt AM. 1990. Bone-forming cells in clinical conditions. In: Hall BK, editor. Bone, Vol. 1: the osteoblast and osteocyte. Boca Raton, FL: Telford Press and CRC Press. 351-429.

Pavlin D, Lichtler AC, Bedalov A, Kream BE, Harrison JR, Thomas HF, Gronowicz GA, Clark SH, Woody CO, Rowe DW 1992. Differential utilization of regulatory domains within the alpha 1(I) collagen promoter in osseous and fibroblastic cells. J Cell Biol116:227–236.

Page 60: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

52

Pelissier P, Bareille R, Masquelet A, Pelissier SM, Amedee J. 2003. Induced membranes secrete growth factors including vascular and osteoinductive factors and could stimulate bone regeneration. J Bone Miner Res 18(2): S212-S213.

Peng HR, Wright V, Usas A, Gearhart B, Shen HC, Cummins J, Huard J, 2002. Synergistic enhancement of bone formation and healing b y stem cell-expressed VEGF and bone morphogenetic protiein-4. J Clin Invest 110(6): 751-759.

Perren SM, Huggler A, Russenberger S. 1969. Cortical bone healing. Acta Orthop Scand Suppl 125.

Perren SM. 1979. Physical and biological aspects of fracture healing with special reference to internal fixation. Clin Orthop 138: 175-196.

Perren SM. 2002. Evolution of the internal fixation of long bone fracture. The scientific basis of biological internal fixation: choosing a new balance between stability and biology. J Bone Joint Surg Br 84(8): 1093-1110.

Petit I, Szyper-Kravitz M, Nagler A, Lahav M, Peled A, Habler L, Ponomaryov T, Taichman RS, Arenzana-Seisdedos F, Fujii N, Sandbank J, Zipori D, Lapidot T. 2002. G-CSF induces stem cell mobilization by decreasing bone marrow SDF-1 and up-regulating CXCR4. Nat Immunol 3: 687-694.

Pfeilschifter J, Wolf O, Naumann A, Minne HW, Mundy GR, Ziegler R. 1990. Chemotactic response of osteoblast-like cells to transforming growth factor β. J Bone Miner Res 5: 825-830.

Pihlajaniemi T, McKeon J, Gay S, Gay R, DeWet WJ, Myers JC, Prockop DJ. 1989. Increased expression of the gene for the pro-alpha-1(IV) chain of basement-membrane procollagen in cultured skin fibroblasts from two variants of osteogenesis imperfect. Biochem J 257: 439-445.

Ponomaryov T, Peled A, Petit I, Taichman RS, Habler L, Sandbank J, Arenzana-Seisdedos F, Maqerus A, Caruz A, Fujii N, Nagler A, Lahav M, Szyper-Kravitz M, Zipori D, Lapidot T. 2000. Induction of the chemokine stromal-derived factor-1 following DNA damage improves human stem cell function. J Clin Invest 106, 1331-1339.

Poplich LS, Slafeld SL, Rueger DC, Tucker M, Cook SD. 1997. Critical and noncritical size defects healing with osteogenic protein-1. Trans Orthop Res Soc 22: 600. Abstract

Porte D, Tuckermann J, Becker M, Baumann B, Teurich S, Higgins T, Owen MJ, Schorpp-Kistner M, Angel P. 1999. Both AP-1 and Cbfa-1-like factors are required for the induction of interstitial collagenase by parathyroid hormone. Oncogene 18: 667-678.

Probst A and Spiegel HU. 1997. Cellular mechanisms of bone repair. J Invest Surg. 10: 77-86.

Proff P, Romer P. 2009. The molecular mechanism behind bone remodeling: A review. Clin Oral Invest 13: 355-362.

Pruijt JFM, Willemze R, Fibbe WE. 1999. Mechanisms underlying hematopoietic stem cell mobilization induced by the CXC chemokine interleukin-8. Curr Opin Hematol 6(3): 152-158.

Puzas JE, Ishibe M. 1992. Osteoblast/osteoclast coupling. In: Rifkin BR, Gay CV (eds.) Biology and Physiology of the Osteoclast. CRC Press, Boca Ratom, FL, USA, pp. 337-356.

Page 61: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

53

Rahn BA. 2002. Bone healing: histologic and physiologic concepts. In: Fackelman GE, editor. Bone in clinical orthopaedics. Stuttgart, NY: Thieme. 287-326.

Ramirez F, Di Liberto M. 1990. Complex and diversified regulatory programs control the expression of vertebrate collagen genes. FASEB J 4: 1616-1623.

Reddi AH. 1994. Cartilage morphogenesis role of bone and cartilage morphogenetic proteins, homeobox genes and extracellular matrix. Matrix Biol 14: 599.

Reinholt FP, Hultenby K, Oldberg A, Heinegard D. 1990. Osteopontin – a possible anchor of osteoclasts to bone. Proc Natl Acad Sci USA 87: 4473-4475.

Rifkin BR, Heijl L. 1979. The occurrence of mononuclear cells at sites of osteoclastic bone resorption in experimental periodontitis. J Periodontol 50: 636-640.

Rippe RA, Lorenzen SI, Brenner DA, Breindl M. 1989. Regulatory elements in the 5’-flanking region and the first ntron contribute to transcriptional control of the mouse alpha 1 type I collagen gene. Mol Cell Biol 9: 2224-2227.

Robert Josse. 2009. Bone Biology and the Role of RANK/RANKL/OPG Pathway. Geriatrics and Aging Conference Report from the 2009 CGS Annual Scientific Meeting: Satellite Symposium 30-31.

Robey PG, Young MF, Flanders KC, Roche NS, Kondaiah P, Reddi AH, Termine JD, Sporn MB, Roberts AB. 1987. Osteoblasts synthesize and respond to transforming growth factor-type beta (TGF-beta) in vitro. J Cell Biol 105: 457-463.

Rodan GA. 1997. Mechanical loading, estrogen deficiency, and the coupling of bone formation to bone resorption. J Bone Miner Res 6: 527-530.

Rossant J, Howard L. 2002. Signaling pathways in vascular development. Annu Rev Cell Dev Biol 18: 541-573.

Rossert J, Eberspaecher H, de Cromrugghe B. 1995. Separate cis-acting DNA elements of the mouse pro-alpha 1(I) collagen promoter direct expression of reporter genes to different type I collagen-producing cells in transgenic mice. J Cell Biol 129: 1421-1432.

Rossi P, de Crombrugghe B. 1988. A nuclear factor 1 binding site mediates the transcription activation of a type I collagen promoter by transforming growth factor-beta. Cell 52: 405-414.

Rothe L, Collin-Osdoby P, Chen Y, Sunyer T, Chaudhary L, Tsay A, Goldring S, Avioli L, Osdoby P. 1998. Human osteoclasts and osteoclast-like cells synthesize and release high basal and inflammatory stimulated levels of the potent chemokine interleukin-8. Endocrinology 139(10): 4353-4363.

Rowe DW, Kream BE. 1982. Regulation of collagen synthesis in fetal rat calvaria by 1,25-dihydroxyvitamin D3. J Biol Chem 257: 8009-8015.

Rubin C, Judex S, Hadjiargyrou M. 2002. Skeletal adaptation to mechanical stimuli in the absence of formation or resorption of bone. J Musculoskelet Neuronal Interact 2: 264-267.

Sandberg MM, Aro HT, Vuorio EI. 1993. Gene expression during fracture repair. Clin Orthop 289: 292-312.

Page 62: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

54

Sasano Y, Zhu JX, Tsubota M, Takahashi I, Onodera K, Mizoguchi I, Kagayama M. 2002. Gene expression of MMP8 and MMP13 during embryonic development of bone and cartilage in the rat mandible and hind limb. J Histochem Cytochem 50: 325-332.

Sato T, Foged NT, Delaisse JM. 1998. The migration of purified osteoclasts through collagen is inhibited by matrix metalloproteinase inhibitors. J Bone Miner Res 13: 59-66.

Schaller S, Henriksen K, Sveigaard C, Heegaard AM, Helix N, Stahlhut M, Ovejero MC, Johansen JV, Solberg H, Andersen TL, Hougaard D, Berryman M, Shiodt CB, Sorensen BH, Lichtenberg J, Christophersen P, Foged NT, Delaisse JM, Engsig MT, Karsdal MA. 2004. The chloride channel inhibitor NS3736 prevents bone resorption in ovariectomized rats without changing bone formation. J Bone Miner Res 19: 1144-1153.

Schatzker J, Waddell J, Stoll JE. 1989. The effects of motion on the healing of cancellous bone. Clin Orthop 245: 282-287.

Schell H, Lienau J, Epari DR, Seebeck P, Exner C, Muchow S, Bragulla H, Haas NP, Duda GN. 2006. Osteoclastic activity begins early and increases over the course of bone healing. Bone 38: 547-554.

Schenk R, Willenegger H. 1967. Morphological findings in primary fracture healing: callus formation. Simp Biol Hungar 7: 75-80.

Schenk R. 1987. Cytodynamics and histodynamics of primary bone repair, in Fracture Healing (Lang JM, ed.) Churchill Livingstone, New York, 23-32.

Schindeler A, McDonald MM, Bokko P, Little DG. 2008. Bone remodelling during fracture repair: The cellular picture. Semin Cell Dev Biol 19:459-466.

Schindeler A, Liu R, Little DG. 2009. The contribution of different cell lineages to bone repair: exploring a role for muscle stem cells. Differentiation 77: 12-18.

Schmidt A, Rossi P, de Crombrugghe B. 1986. Transcriptional control of the mouse α2(I) collagen gene: functional deletion analysis of the promoter and evidence for cell-specific expression. Mol Cell Biol 6: 347-354.

Schor AM, Allen TD, Canfield AE, Sloan P, Schor SL. 1990. Pericytes derived from the retinal microvasculature undergo calcification in vitro. J Cell Sci 97: 449-461.

Schweizer L, Varmus H. 2003. Wnt/Wingless signaling through beta-catenin requires the function of both LRP/Arrow and frizzled classes of receptors. BMC Cell Biol 4(4).

Selvamurugan N, Chou W-Y, Pearman AR, Pulumamti MR, Partridge NC. 1998. Parathyroid hormone regulates the rat collagenase-3 promoter in osteoblastic cells through the cooperative interaction of the activator protein-1 site and the runt domain binding sequence. J Biol Chem 273: 19647-10657.

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.

Simonet WS, Lacey DL, Dunstan CR, Kelley M, Chang MS, Luthy R, Nguyen HQ, Wooden S, Bennett L, Boone T, Shimamoto G, DeRose M, Elliott R, Colombero A, Tan HL, Trail G, Sullivan J, Davy E, Bucay N, Renshaw-Gegg L, Hughes TM, Hill D, Pattison W, Campbell P, Sander S, Van G, Tarpley J, Derby P, Lee R, Boyle WJ. 1997.

Page 63: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

55

Osteoprotegerin: A novel secreted protein involved in the regulation of bone density. Cell 89: 309-319.

Simon AM, Manigrasso MB, O’Connor JP. 2002. Cyclo-oxygenase 2 function is essential for bone fracture healing. J Bone Miner Res 17: 963-976.

Slack JL, Liska DJ, Bornstein P. 1991. An upstream regulatory region mediates high-level, tissue-specific expression of the human α1(I) collagen gene in transgenic mice. Mol Cell Biol 11: 2066-2074.

Somerman MJ, Prince CW, Sauk JJ, Foster RA, Butler WT. 1987. Mechanism of fibroblast attachment to bone extracellular matrix: role of a 44 kilodalton bone phosphoprotein. J Bone Mineral Res 2: 259-265.

Sporn MB, Roberts AB. 1989. Transforming growth factor-beta. Multiple actions and potential clinical applications. JAMA 262: 938-941.

St-Jacques B, Hammerschmidt M, McMahon AP. 1999. Indian hedgehog signalling regulates proliferation and differentiation of chondrocytes and is essential for bone formation. Genes Dev 13: 2072-2086.

Steinberg B, Singh IJ, Mitchell OG. 1981. The effects of cold-stress, hibernation and prolonged inactivity on bone dynamics in the Golden Hamster, Mesodricetus auratus. J Morph 167: 43-51.

Sternlight, MD and Werb Z. 1999. Matrix metalloproteinases. In Guidebook to the Extracellular Matrix, Anchor and Adhesion Proteins, 519-538. Sambrook and Tooze (Oxford University Press). London.

Stickens D, Behonick DJ, Ortega N, Heyer B, Hartenstein B, Yu Y, Fosang AJ, Schorpp-Kistner M, Angel P, Werb Z, 2004. Altered endochondral bone development in matrix metalloproteinase 13-deficient mice, Development 131: 5883-5895.

Strong DD, Beachler AL, Wergedal JE, Linkhart TA. 1991. Insulin-like growth factor II and transforming growth factor beta regulate collagen expression in human osteoblast-like cells in vitro. J Bone Miner Res 6: 15-23.

Suda T, Nakamura I, Jimi E, Takahashi N. 1997. Regulation of osteoclast function. J Bone Miner Res 12: 869-879.

Takamatsu Y, Simmons PJ, Moore RJ, Morris HA, To LB, Levesque JP. 1998. Osteoclast-mediated bone resorption is stimulated during short-term administration of granulocyte colony-stimulating factor but is not responsible for hematopoietic progenitor cell mobilization. Blood 92: 3465-3473.

Takamoto M, Tsuji K, Yamashita T, Sasaki H, Yano T, Taketani Y, Komori T, Nifuji A, Noda M. 2003. Hedgehog signaling enhances core-binding factor a1 and receptor activator of nuclear factor-kappa B ligand (RANKL) gene expression in chondrocytes. J Endocrinol 177: 413-421.

Takayanagi H, Kim S, Koga T, Nishina H, Isshiki M, Yoshida H, Saiura A, Isobe M, Yokochi T, Inoue J, Wagner EF, Mak TW, Kodama T, Taniguchi T. 2002. Induction and activation of the transcription factor NFATc1 (NFAT2) integrate RANKL signaling in terminal differentiation of osteoclasts. Dev Cell 3: 889-901.

Page 64: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

56

Takeda S, Bonnamy JP, Owen MJ, Ducy P, Karsenty G, 2001. Continuous expression of Cbfa1 in non-hypertrophic chondrocytes uncovers its ability to induce hypertrophic chondrocyte differentiation and partially rescues Cbfa1-deficient mice. Genes Dev 15: 467-481.

Tally-Ronsholdt DJ, Lajiness E, Nagodawithana K. 1995. Transforming growth factor-beta inhibition of mineralization by neonatal rat osteoblasts in monolayer and collagen gel culture. In Vitro Cell Dev Biol 31: 274-282.

Tammela T, Enholm B, Alitalo K, Paavonen K. 2005. The biology of vascular endothelial growth factors. Cardiovasc Res 65: 550-563.

Tazawa T, Hoshi K, Kawamoto S, Tanaka M, Ejiri S, Ozawa H. 2004. Osteocytic osteolysis observed in rats to which parathyroid hormone was continuously administered. J Bone Miner Metab 22(6): 524-529.

Teitelbaum SL, Ross FP. 2003. Genetic regulation of osteoclast development and function. Nature Review 4; 638-649.

Thompson Z, Miclau T, Hu D, Helms JA. 2002. A model for intramembranous ossification during fracture healing. J Orthop Res 20: 1091-1098.

Topol L, Chen W, Song H, Day TF, Yang Y. 2009. Sox9 inhibits Wnt signaling by promoting beta-catenin phosphorylation in the nucleus. J Biol Chem 284(5): 3323-3333.

Tou L, Quibria N, Alexander JM. 2001. Regulation of human Cbfa1 gene transcription in osteoblasts by selective estrogen receptor modulators (SERMs). Mol Cell Endocrinol 183: 71-79.

Tran Van P, Vignery A, Baron R. 1982. An electron microscopic study of the bone-remodeling sequence in the rat. Cell Tissue Res 225: 283-292.

Tsuji K, Ito Y, Noda M. 1998. Expression of the PEBP2 alpha A/AML3/CBFA1 gene is regulated by BMP4/7 heterodimer and its overexpression suppresses type I collagen and osteocalcin gene expression in osteoblastic and nonosteoblastic mesenchymal cells. Bone 22(2): 87-92.

Turek TJ, Bostrom MPG, Camacho N, Blake CA, Palmer R, Seeherman HJ, Wozney JM. 1997. Acceleration of bone healing in a rabbit ulnar osteotomy model with recombinant human bone morphogenic protein 2. Clin Orthop 318: 222-230.

Ueta C, Iwamoto M, Kanatani N, Yoshida C, Liu Y, Enomoto-Iwamoto M, Ohmori T, Enomoto H, Nakata K, Takada K, Kurisu K, Komori T. 2001. Skeletal malformations caused by overexpression of Cbfa1 or its dominant negative form in chondrocytes. J Cell Biol 153: 87-100.

Urist MR. 1965. Bone formation by autoinduction. Science 150: 893-899.

Ushiku C, Adams DJ, Jiang X, Wang L, Rowe DW. 2010. Long bone fracture repair in mice harboring GFP reporters for cells within the osteoblastic lineage. J Orthop Res 28(10): 1338-1347.

Usui M, Xing L, Drissi H, Zuscik M, O’Keefe R, Chen D, Boyce BF. 2008. Murine and chicken chondrocytes regulate osteoclastogenesis by producing RANKL in response to BMP2. J Bone Miner Res 23: 314-325.

Page 65: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

57

Uusitalo H, Hiltunen A, Soderstrom M, Aro HT, Vuorio E, 2000. Expression of cathepsins B, H, K, L, and S and matrix metalloroteinases 9 and 13 during chondrocyte hypertrophy and endochondral ossification in mouse fracture callus. Calcif Tissue Int 67: 382-390.

Vainio S, Karavanova I, Jowett A, Thesleff I. 1993. Identification of BMP-4 as a signal mediating secondary induction between epithelial and mesenchymal tissues during early tooth development. Cell 75: 45-85.

Visnjic D, Kalajzic I, Gronowicz G, Aguila HL, Clark SH, Lichtler AC, Rowe DW. 2001. Conditional ablation of the osteoblast lineage in Col2.3Δtk transgenic mice. J Bone Miner Res 16(12): 2222-2231.

Vortkamp A, Lee K, Lanske B, Segre GV, Kronenberg HM, Tabin CJ. 1996. Regulation of rate of cartilage differentiation by Indian hedgehog and PTH-related protein. Science 273: 613-622.

Vortkamp A, Pathi S, Peretii GM, Caruso EM, Zaleske DJ, Tabin CJ. 1998. Recapitulation of signals regulating embryonic bone formation during postnatal growth and in fracture repair. Mech Dev 71, 65-76.

Vu TH, Shipley JM, Bergers G, Berger JE, Helms JA, Hanahan D, Shapiro SD, Senior RM, Werb Z. 1998. MMP-9/gelatinase B is a key regulator of growth plate angiogenesis and apoptosis of hypertrophic chondrocytes. Cell 93(3): 411-422.

Vu TH, Werb Z. 2000. Matrix metalloproteinases: effectors of development and normal physiology. Genes Dev 14: 2123-2133.

Vuorio E, de Crombrugghe B. 1990. The family of collagen genes. Annu Rev Biochem 59: 837-872.

Wang Y, Wan C, Gilbert SR, Clemens TL, 2007a. Oxygen sensing and osteogenesis. Ann N Y Acad Sci 1117: 1-11.

Wang Y, Wan C, Deng L, Liu X, Cao X, Gilbert SR, Bouxsein ML, Faugere MC, Guldberg RE, Gerstenfeld LC, Haase VH, Johnson RS, Schipani E, Clemens TL, 2007b. The hypoxia-inducible factor alpha pathway couples angiogenesis to oseogenesis during skeletal development. J Clin Invest 117: 1616-1626.

Wagner EF and Karsenty G, 2001. Genetic control of skeletal development. Curr Opin Genet Dev 11: 527-532.

Walsh MJ, LeLeiko NS, Sterling JM. 1987. Regulation of types I, III, and IV procollagen mRNA synthesis in glucocorticoid-mediated intestinal development. J Biol Chem 262: 10814-10818.

Watanabe T, Suzuya H, Onishi T, Kanai S, Kaneko M, Watanabe H, Nakagawa R, Kawano Y, Takaue Y, Kuroda Y, Talmadge JE. 2003. Effects of granulocyte colony-stimulating factor on bone metabolism during peripheral blood stem cell mobilization. Int J Hematol 77: 75-81.

Wergedal JE, Matsuyama T, Strong DD. 1992. Differentiation of normal human bone cells by transforming growth factor beta and 1,25(OH)2 Vitamin D3. Metabolism 41: 42-48.

Westendorf JJ, Kahler RA, Schroeder TM. 2004. Wnt signaling in osteoblasts and bone diseases. Gene 341: 19-39.

Page 66: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

58

White AAI, Punjabi MM, Southwick WO. 1977. The four biomechanical stages of fracture healing. J Bone Joint Surg 59A, 188.

Witten PE. 1997. Enzyme histochemical characteristics of osteoblasts and mononucleated osteoclasts in a teleost fish with acellular bone (Oreochromis niloticus, Cichlidae). Cell Tissue Res 287: 591-599.

Witten PE, Villwock W, Peters N, Hall BK. 2000. Bone resorption and bone remodelling in juvenile carp (Cyprinus carpio). J Appl Ichthy 16: 254-261.

Wolf C, Thisse C, Stoetzel C, Thisse B, Gelinger P, Perrin-Schmitt F. 1991. The M-twist gene of Mus is expressed in subsets of mesodermal cells and is closely related to the Xenopus X-twi and the Drosophila twist genes. Dev Biol 143: 363-373.

Wong GL. 1982. Characterizations of the subpopulations of OC and OB bone cells obtained by sedimentation at unit gravity. Calcif Tissue Int 34: 67-75.

Wright E, Hargrave MR, Christiansen J, Cooper L, Kun J, Evans T, Gangadharan U, Greenfield A, Koopman P. 1995. The Sry-related gene Sox9 is expressed during chondrogenesis in mouse embryos. Nat Genet 9: 15-20.

Wu CW, Tchetina EV, Mwale F, Hasty K, Pidoux I, Reinder A, Chen J, can Wart HE, Poole AR. 2002. Proteolysis involving matrix metalloproteinase 13 (Collagenase-3) is required for chondrocyte differentiation that is associated with matrix mineralization. J Bone Miner Res 17(4): 639-651.

Yajima T, Sakakura Y, Tsuruga E, Hirai T, Ikeda Y, Fujii S, Shide N. 1999. Collagen-phagocytosing ability of periodontal osteoblasts at the bone surface. Arch Histol Cytol 62:17-25.

Yancopoulos GD, Davis S, Gale NW, Rudge JS, Wiegand SJ, Holash J. 2000. Vascular-specific growth factors and blood vessel formation. Nature 407: 551-566.

Yang Y, Topol L, Lee H, Wu J. 2003. Wnt5a and Wnt5b exhibit distinct activities in coordinating chondrocyte proliferation and differentiation. Development 130: 1003-1015.

Yasko AW, Lane JM, Fellinger EJ, Rosen V, Wozney JM, Wang EA. 1992. The healing of segmental bone defects induced by recombinant human bone morphogenetic protein (rhBMP-2): A radiographic, histological, and biomechanical study in rats. J Bone Joint Surg 74(7): 659-670.

Yasuda H, Shima N, Nakagawa N, Mochizuki SI, Yano K, Fujise N, Sato Y, Goto M, Yamaguchi K, Kuriyama M, Kanno T, Murakami A, Tsuda E, Morinaga T, Higashio K. 1998. Identity of osteoclastogenesis inhibitory factor (OCIF) and osteoprotegerin (OPG): a mechanism by which OPG/OCIF inhibits osteoclastogenesis in vitro. Endocrinology 139: 1329-1337.

Yee JA, Yan L, Dominguez JC, Allan EH, Martin TJ. 1993. Plasminogen-dependent activation of latent transforming growth factor beta by growing cultures of osteoblast-like cells. J Cell Physiol 157: 528-534.

Yin T, Li L. 2006. The stem cell niches in bone. J Clin Invest 116(5): 1195-1201.

Page 67: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

59

Yoshida H, Hayashi S, Kunisada T, Ogawa M, Nishikawa S, Okamura H, Sudo T, Shultz LD, Nishikawa S. 1990. The murine mutation osteopetrosis is in the coding region of the macrophage colony stimulating factor gene. Nature 345: 442-444. 

Yoshida CA, Komori T. 2005. Role of Runx proteins in chondrogenesis.Crit Rev Eukaryot Gene Expr 15(3): 243-254.

Zhang X, Bao K, Dai K. 2000. Stress-relaxation plates and the remodelling of callus and cortex under the palte in rabbits. Chin Med J (Eng) 113(9): 805-809.

Zhou YX, Xu X, Chen L, Li C, Brodie SG, Deng CX. 2000. A Pro250Arg substitution in mouse Fgfr1 causes increased expression of Cbfa1 and premature fusion of calvarial sutures. Hum Mol Genet 9: 2001-2008.

Zhou G, Zhen Q, Engin F, MUnivez E, Chen Y, Sebald E, Krakow D, Lee B. 2006. Dominance of Sox9 function over Runx2 during skeletogenesis. Proc Natl Acad Sci USA 103(50): 19004-194009.

Zou H, Niswander L. 1996. Requirement for BMP signaling in interdigital apoptosis and scale formation. Science 272: 738-741.

Page 68: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

60

Chapter 2 Possible Role of Osteoblasts in Regulating the Initiation of

Endochondral Repair Process during Fracture Healing

2 Summary

Fracture healing is a regenerative process requiring precise coordination of a variety of

skeletal, haematopoietic, vascular and immune cells which are necessary for successful repair

process and union of damaged bone. Previously, osteoblasts have been shown to be important in

osteoclastogenesis during the remodelling phase of fracture repair. However, their role during the

initiation phase of endochondral fracture repair has been unclear. Therefore, we hypothesized

that ablation of osteoblasts would lead to a delay in the initiation of chondrogenesis and

progression of the endochondral bone formation. The objective for this project was to analyze

the various stages of endochondral repair process, in order to determine the relative progression

of the fracture repair in the absence of osteoblasts. A transgenic mouse model that expresses a

truncated form of herpes simplex virus thymidine kinase (HSV-tk) gene under the control of a

2.3-kilobase fragment of rat α1 type I collagen promoter (Col2.3∆tk; DTK), was used to drive

the expression of the gene in early differentiating osteoblasts. A nucleoside analog, ganciclovir

(GCV), was used to ablate the cells in osteogenic lineage and fracture calluses were examined at

3, 7 and 21 days post fracture. Our results show that osteoblast depletion delays the initiation of

endochondral bone repair and the process of chondrogenesis. Continuous ablation of osteoblasts

for 21 days post fracture hindered the progression of endochondral ossification at the soft callus

stage with abundant deposition of cartilage matrix at the fracture site of DTK transgenic mice.

Upon cessation of the drug treatment, the bone was able to heal uneventfully similar to the

wildtype controls.

2.1 Introduction

Fracture healing is a complex physiological process that recapitulates certain aspects of

normal skeletal development and growth. It is a regenerative event that involves the coordination

of a variety of cells, ranging from haematopoietic and immune cells within the bone marrow in

conjunction with vascular and skeletal cell precursors that are recruited to the fracture site, to

ensure proper repair of the damaged bone. Within the microstructure hierarchy of fracture repair,

Page 69: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

61

the predominant cells involved include the chondrocytes, osteocytes, osteoblasts, and osteoclasts.

Thus understanding the interplay between these different cell types during the healing process

would be useful for improving the development of cell-based therapies.

The repair process is comprised of four overlapping phases, initiated by (1) an

inflammatory response that results in the recruitment of mesenchymal progenitor cells that

differentiate into chondrocytes, which produce cartilage and osteoblasts, the bone forming cells

(Schindeler, 2008). Thereafter, (2) the fracture is bridged by soft callus and (3) later hard callus

until mechanical instability is restored to the fracture site (Schindeler, 2008). The final stage of

fracture repair involves the (4) remodeling of the hard callus by osteoclasts, the bone resorbing

cells, and osteoblasts into the original cortical and/or trabecular bone configuration (Gerstenfeld,

2003).

During development, bone tissue is formed by two distinct ossification processes.

Intramembranous bone formation is mediated through the inner periosteal osteogenic layer with

bone made initially without the intervention of a cartilage phase (Shapiro, 2008). Most fractures

possess some level of mechanical instability and heal by the process of endochondral

ossification, which occurs in unstable regions external to the periosteum immediately adjacent to

the fracture site (Dimitriou, 2005). The eventual bridging of the hard callus across the fracture

gap provides the initial stabilization and regaining of biomechanical function.

Previous works have shown that close coordination and interplay between the different

cell types involved in fracture repair is important for successful repair process. For example,

expression of RANKL by osteoblasts coordinates bone remodeling by stimulating bone

resorption by local osteoclasts through binding of RANKL to its cellular receptor RANK on

osteoclasts (Lacey, 1998; Li, 2000; Nakagawa, 1998). This close coordination between the two

cell types is further regulated through the expression of the decoy receptor osteoprotegerin by

osteoblasts, which binds RANKL and prevents its interaction with RANK receptors present on

osteoclasts (Roodman, 2006; Udagwa, 2000).

This coordination between the different cell types that are involved in fracture healing is

further evident by the role of various matrix metalloproteinases (MMPs) that degrade most

components of the extracellular matrix. Besides participating in degradation of ECM to allow

cell migration, MMPs are involved in modulation of biologically active molecules by direct

Page 70: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

62

cleavage and regulation of the activity of other proteases (Ortega, 2003). Two critical MMPs that

are highly expressed during endochondral ossification include MMP9 and MMP13.

Preosteoclasts and other chondroclastic cells express MMP9 during the start of

neovascularization of the cartilage anlage (Holmbeck, 1999; Zhou, 2000), and MMP13 is

expressed in the terminal hypertrophic chondrocytes and in the newly recruited osteoblasts (Vu,

1998). Together, these two MMPs account for most of the protease-dependent steps of

endochondral ossification (Engsig, 2000) and along with the angiogenic factor, VEGF, are key

regulators of the remodeling of the skeletal tissue (Ortega, 2003).

Long bone fracture has been used for many years as the model for evaluating the

induction of osteogenic and chondrogenic lineages for the initial skeletal stabilization as well as

the subsequent coordination of the osteoblast and osteoclast lineages to remodel the immature

callus back to a cortical bone (Ushiku, 2010). Although the role of osteoblasts during the

remodeling phase of fracture repair has been previously studies, their role during chondrogenesis

has not been well investigated. Few studies investigating the interplay between these two cell

populations have shown that soluble factors expressed by chondrocytes selectively promote

osteogenesis of mesenchymal stromal cells (Gerstenfeld, 2003a), and in co-culturing of

osteoblasts and chondrocytes the differentiation level of osteoblasts influenced the proliferation

and differentiation levels of chondrocytes (Nakoka, 2006). Therefore, we hypothesized that

ablation of osteoblasts would lead to a delay in the initiation of chondrogenesis and progression

of the endochondral bone formation. The objective for this project was to analyze the various

stages of endochondral repair process, in order to determine the relative progression of the

fracture repair in the absence of osteoblasts.

One method for studying the lineage relationships and cell functions of a population of

differentiating cells is the use of tissue-specific transgenic expression of herpes simplex virus

thymidine kinase (HSV-tk) to conditionally deplete a specific cell population. As such, in order

to study the role of osteoblasts during bone fracture repair, we used a transgenic mouse model

expressing the HSV-tk gene under the control of a 2.3-kilobase fragment of the rat α1 type I

collagen promoter (Col2.3∆tk; DTK), which is active in early differentiating osteoblasts.

Expression of the tk gene renders the cells susceptible to a variety of nucleoside analogs such as

GCV, which leads to ablation of cells in osteoblastic lineage. In our study, fractures were

generated in the tibia of three-month-old male DTK mice that were pretreated with GCV for two

Page 71: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

63

weeks. After surgery, one set of DTK transgenic animals were given GCV treatment for an

additional 3, 7 and 21 days, while another set received saline treatments (Figure 4). Control

animals negative for the DTK transgene were treated with GCV for above time point as controls.

Furthermore, in order to evaluate the potential side effects of the GCV drug during the healing

process, additional untreated animals (both transgenic and wildtype) were analyzed for the same

timepoints (data not shown). Our results show that osteoblast depletion delays the initiation of

endochondral bone repair and the process of chondrogenesis, with continuous ablation of

osteoblasts for 21 days post fracture hindering the progression of endochondral ossification past

the soft callus stage fracture site of DTK transgenic mice. Upon cessation of the drug treatment,

the bone was able to heal uneventfully similar to the wildtype controls.

2.2 Methods and Materials

2.2.1 Generation of fractures:

All animal procedures were approved by the animal care committee of Hospital for Sick

Children. Tibial fracture of control and DTK transgenic mice was performed as previously

described (Chen, 2007). Briefly, after anaesthetizing the animals with isoflurane, a small incision

was made close to the left leg to expose the head of the tibia. A suture needle was used to

puncture the head of the tibia to allow the insertion of a non-corrosive metal pin in the marrow

cavity in order to stabilize the leg prior to fracture generation. The fracture was generated using a

surgical scissor after the skeletal muscles, juxtaposed to the tibia, were pulled away from the

bone to prevent unnecessary damages to this tissue. The skin was then closed using silk sutures

and metal staples and the animals were allowed to recover. After recovery, an analgesic

(ibumorphine) was administered for the first three days post surgery. The fractured left tibia and

the intact right tibia were then harvested at 3, 7 and 21 days time-points post surgery (Figure 5).

2.2.2 Mechanism of action of GCV:

The DTK transgenic mice express a truncated form of HSV-tk gene whose expression is under

the control of the rat α1 type I collagen gene promoter that restricts TK expression in early

differentiating osteoblasts. Upon treatment of animals with GCV, a nucleoside analogue of

guanosine, and a homologue of acyclovir, the thymidine kinase present in target cells

monophosphorylates the GCV drug. The drug is further phosphorylated by endogenous kinases

Page 72: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

64

and competitively inhibits the incorporation of dGTP into DNA, resulting in inhibition of DNA

replication and cell ablation.

2.2.3 Real-time PCR:

Total RNA was isolated from the fracture callus at the 7 days time point and from intact

unfractured tibias at 3 and 7 days post-surgery. cDNAs were made from the extracted RNA

samples. Real-time PCR was performed to determine the level of osteoblastic (Type I collagen

and ALP), chondrogenic (Sox5 and Aggrecan) and MMP13 gene expression as compared with

GAPDH (a housekeeping gene control). At least 3 animals in each treatment group were

examined. PCR primers (Type I Collagen, ALP, Sox5, Aggrecan and Matrix metalloproteinase

13) were purchased from Applied Biosystems.

2.2.4 Staining method – Safranin-O/Haematoxylin-Eosin:

For histological analysis, the tissue at the fracture site was harvested, fixed in 4%

paraformaldehyde, decalcified in 20% EDTA (pH 8.0), dehydrated in progressive concentrations

of ethanol, cleared in xylene and embedded in paraffin. The entire tibia was sectioned 5 µM

thick and the sections from the center of the tibia with the widest diameter were selected. The

center sections were deparaffinized and hydrated in descending concentrations of ethanol. These

sections were then stained with haematoxylin-eosin (HE) and safranin-O (SO) for analysis of

gross morphology and cartilage deposition at fracture repair respectively.

2.2.5 Histomorphometric, Cell quantification and X-ray analysis:

For histomorphometric measurements, safranin-O stained tissue sections were used for

evaluation of bone and cartilage volumes as a percentage of total callus tissue volume. Bone

volume (BV) and cartilage volume (CV) were measured using the Bioquant system, and these

measurements were normalized to the overall tissue volume (TV) measured. Using the same

safranin-O stained sections from various time points, at least 5 fields were randomly selected

from the healing fracture callus for quantification of the osteoblast-like cells. In absence of a

callus, areas near the metaphyseal zone of the tibia were chosen as alternatives. Radiographic

picture were obtained by using Faxitron MX20 X-ray system for radiographic appearance of

fractured tibias (Figure 4).

Page 73: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

65

2.2.6 Collagen type X and TRAP staining:

To determine remodeling parameters of the harvested fracture calluses, sections were stained for

tartrate-resistant acid phosphatase (TRAP) and counterstained with haematoxylin. Osteoclasts

were identified by their multinucleated morphology and close proximity to the bone surface at

sites of bone resorption.

2.2.7 Statistical Analysis:

Histomorphometric data are expressed as mean +/- standard deviation. Statistical differences

were calculated using Student’s t test. Number of animals for each group at the 7 day time point

are as follows (Bone Volume; Cartilage Volume): Wildtype GCV injected = 1n;1n, DTK

continuous GCV treated = 4n;4n. Number of animals for each group at 21 day time point are as

follows (Bone Volume; Cartilage Volume): Wildtype untreated control = 4n;3n, DTK untreated

control = 3n;3n, Wildtype GCV injected = 8n;6n, DTK non-continuous GCV treated = 6n;4n,

DTK continuous GCV treated = 4n;3n. A p-value below 0.05 was considered statistically

significant.

2.3 Results

2.3.1 Pretreatment of DTK transgenic mice with GCV leads to ablation of osteoblasts

To study the role of osteoblasts during bone fracture repair DTK transgenic animals and

control mice were pretreated with GCV for two weeks prior to fracture generation. In order to

examine if the GCV pretreatment of the DTK transgenic animal was effective in ablation of the

osteoblast cell lineage, tibial fractures were collected at 3 days post fracture. Histological

analysis of safranin O stained sections of these bones showed the presence of bone lining cells at

the metaphyseal area near the site of longitudinal growth adjacent to the growth plate in the

control animals (Figure 6A’). These cells were absent in the DTK transgenic animals in both the

non-continuous and continuous GCV treated groups (Figure 6B’, C’). The effect of the GCV

treatment on osteoblasts was found to be consistent in all the time points tested (3, 7, and 21 days

post-fracture). A marked decrease in mono-nucleated bone-lining cell population was observed

around the trabecular bones with the GCV drug treatment, and the cessation of the treatment was

followed by a repopulation of these cell types around the metaphyseal trabecular niche (Figure

Page 74: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

66

7). This finding is consistent with the original study done by Vinsnjic et al that showed a strong

expression of the thymidine kinase protein localized in osteoblasts lining the endosteal,

periosteal, and trabecular surfaces (Visnjic, 2004). Furthermore, similar to their study, transgenic

mice treated with GCV had a marked loss of bone marrow cellularity as compared to the non-

transgenic controls that showed no evidence of GCV-induced toxicity (Visnjic, 2004) (data not

shown).

2.3.2 Osteoblast depletion delays initiation of endochondral bone repair

During development, the onset of endochondral ossification begins with the condensation

of mesenchymal progenitor cells at sites of future skeletal development (Hall, 1995), a process

that is recapitulated during the early stages of fracture repair (Hiltunen, 1993) Shortly after the

condensation stage, cells in the central region of the aggregation begin to differentiate along a

cartilaginous lineage (Iwasaki, 1997) in response to growth factors and cytokines released by

platelets, inflammatory cells, and neighboring cells and tissues (Bolander, 1992; Bruder, 1994).

In order to establish a baseline for comparison of repair process at a stage when

osteoblasts have not yet been recruited to the fracture callus, fractured tibias were harvested at 7

days post surgery from GCV treated controls as well as continuous and non-continuous GCV

treated DTK mice. As previously reported (Visnjic, 2004) it was hypothesized that at 7 days

following the fracture, most of the callus would be composed of cartilage. As expected,

histological analysis of the safranin-o stained sections showed an extensive collagen matrix

production in the fractures of the wildtype controls (Figure 8D). However, there was a lack of

collagen deposition at the fracture site of continuous GCV treated transgenic mice (Figure 8F).

The growth plate collagen staining was used as an internal control and collagen matrix

production was identified by the red staining of proteoglycans at the site of fracture.

In addition, fracture calluses from a separate group of GCV-treated control and

continuous GCV-treated DTK mice were harvested at 7 day time point for RNA analysis. To

control for any contamination that might occur during the harvesting of the calluses, mainly from

the muscle tissue surrounding the fracture site that would need to be collected along with the

fracture callus, intact tibias from the same animals were also collected as a reference. The muscle

Page 75: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

67

tissue was easily removed from these bones and the entire tibia containing the growth plate was

flash frozen in liquid nitrogen for further processing.

Since the expression of the tk transgene is under type I collagen promoter, and GCV

treatment should lead to ablation of dividing cells expressing type I collagen, the expression level

of this gene along with ALP (another early osteoblast marker) was examined in the fracture

calluses and intact tibia of both the control and DTK transgenic animals. Real-time PCR analysis

shows a decrease in the expression of these genes in the fracture callus of the continuous GCV

treated mice as compared to the wildtype controls (Figure 9).

Type I collagen is also expressed in osteocytes (Kamiya, 2001), but since these cells are

not dividing the GCV treatment should not lead to death of osteocytes in the cortical bone

(Figure 10). Furthermore, it has been previously reported that the distance between osteocytes

can be calculated (Sugawara, 2005). Thus it was assumed that there would be similar number of

osteocytes in each of the intact tibias collected, thereby setting a baseline threshold, where any

decrease in type I collagen in continuous GCV-treated transgenic mice could be hypothesized to

be due to the ablation of osteoblasts. Similar decrease in type I collagen expression was observed

in the intact tibias of the continuous GCV-treated mice as compared to the wildtype controls

(data not shown).

Other genes that were examined in the 7 day old calluses included genes involved in

chondrogenesis and cartilage matrix production (Sox5 and aggrecan, respectively), as well as

collagenase MMP13, which is expressed by hypertrophic chondrocytes and osteoblasts during

endochondral ossification. A significant decrease was seen in all above genes in the continuous

GCV-treated groups as compared to wildtype controls (Figure 9). These results, along with the

histology data suggests that the continuous ablation of osteoblasts with GCV leads to a lack of

cartilage deposition in these animals at the chondrogenic stage of fracture healing.

2.3.3 Continuous ablation of osteoblasts hinders the progression of endochondral ossification past the soft callus stage

To determine the role of osteoblasts during endochondral repair process DTK transgenic

animals were pretreated with GCV for duration of two weeks prior to fracture generation. One

set of transgenic animals was injected with GCV continuously for 21 days, whereas another set

Page 76: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

68

of DTK animals received saline for the same duration (Figure 5). Wildtype control mice

receiving GCV for 21days were used as controls. Additionally, to examine the natural fracture

repair process in absence of GCV treatment, calluses were harvested from untreated wildtype

controls and DTK transgenic mice at 21 day time point (data not shown).

To verify the effect of GCV treatment on osteocytes, various locations in the cortical

bone were examined in the different treatment groups at all time points. Osteocytes were

identified by their elongated morphology and their parallel orientation to the long axis of cortical

bone. In the representative pictures (Figure 10) osteocytes are seen present in round to oval

shaped lacunae spaces in the bone (arrow). Adjacent lacuanes are linked by multiple small canals

known as canaliculi that house the osteocyte cell processes (Palumbo, 1990). This lacunar-

canalicular intraosseous system plays a key role in providing the bone cells with nutrients from

blood vessels and allows intercellular communication of bone cells for biophysical control

mechanisms essential to tissue development and maintenance (Shapiro, 2008). Any injury to the

bone that leads to a break of this communication system would result in the death of osteocytes

that lie in close proximity to the damaged site. This is apparent by the empty lacuanae spaces

found in the cortical bone adjacent to fracture site of the different treatment and control groups,

where the osteocytes closest to the damaged area have died off (Figure 11E).

Safranin-O staining was performed on paraffin-embedded tibial sections of 21 day old

fractures from the different treatment group for detection of osteoid and cartilage deposition at

the site of fracture (Figure 8G-I). Representative fracture were selected from each treatment

group and the red staining of cartilage at the growth plate was used a reference control. Other

than the continuous GCV-treated animals (Figure 8I), the callus of the various control and

treatment groups was primarily composed of new bone at 21 days post fracture. Multiple

condensation sites were still present in the fracture callus of continuous GCV-treated animals,

suggesting that osteoblast ablation delays the endochondral repair process.

In addition, HE staining of selected 21 day old fracture callus section of the various

treatment groups showed osteoblasts lining the periosteum or along the trabecular bone within

the interim callus of the untreated control animals (data not shown) as well as the GCV-treated

wildtype and non-continuous GCV-treated DTK mice (Figure 11D-F). Osteoblasts were

identified by their mononucleated morphology and their single array lining of the surface of

Page 77: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

69

newly synthesized bone at the site of fracture. Active osteoblasts were identified by their

polarized orientation at sites of bone mineralization.

Endochondral bone formation was observed at the site of fracture repair on mineralized

cartilage remains as would be seen in normal metaphyseal bone formation. The initially

synthesized woven bone at the site of repair can be seen surrounded by darker staining more

organized lamellar bone that is then immediately covered with a layer of surface osteoblasts.

Higher power view of the fracture callus (Figure 11D’-F’) shows the presence of active

osteoblasts at sites of osteoid deposition (arrows). In addition, areas of osteoblast-to-osteocyte

transformation can be observed in the fracture callus as some osteoblasts have become embedded

within the bone matrix produced by neighboring osteoblasts (Figure 11E’).

To determine the level of remodeling at 21 days post fracture, sections from the various

treatment groups were stained with TRAP. Almost no TRAP staining was observed in the

continuous GCV-treated group whereas GCV-treated control animals and non-continuous GCV-

treated transgenic animals exhibited TRAP positive multinucleated cells scattered among the

newly formed trabeculae at the fracture site (Figure 12). Osteoclasts, which are a member of the

monocyte/macrophage family that involved in bone resorption during normal bone turnover as

well as in the remodeling phase of fracture repair, can be seen along the newly deposited bone.

They are multinucleated cells that lie in close proximity to the bone surface at site of bone

resorption and stain positive for tartrate-resistant acid phosphatase (TRAP).

Staining for collagen type X showed that at 7 day time point, the fracture callus of the

continuous GCV-treated DTK mice lacked the hypertrophic chondrocyte specific type X

collagen matrix production, while in control animals abundant matrix deposition was observed

(Figure 13A-C). At 21 day time point there was still an abundant amount of collagen type X

matrix present in the fracture callus of the continuous GCV-treated DTK mice (Figure 13D-F).

These data taken together with the safranin O staining showing no new bone deposition at the

fracture site, suggest that the endochondral repair process of these callus is delayed at 7 day time

point and is still in the early chondrogenic stages at the 21 time point stage.

Finally, to quantify the level of bone and cartilage deposition at the fracture site,

histomorphometric measurements were performed on safranin O stained section of 7 day and 21

day old tibial fractures of the different treatment groups (Figure 14). Tissue areas selected for

Page 78: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

70

analysis were determined by the two farthest edges of the callus where the periosteal new bone

formation meets the adjacent cortical bone. Various bone and cartilage parameters were

examined and the data shows lower cartilage matrix synthesis in 7 day old fractures of

continuous GCV treated transgenic mice in comparison to wildtype controls (Figure 14A).A

significant decrease in new osteoid formation and a significantly higher cartilage deposition was

observed in continuous GCV-treated group as compared to the GCV-treated control animals and

non-continuous GCV-treated DTK mice at the 21 day time point (Figure 14B). Overall bone

volume (mineralized bone plus the newly synthesized osteoid) did not change across the three

treatment groups, however a significantly higher mineralized bone volume per tissue volume was

observed in the continuous GCV-treated DTK mice (Figure 14B).

2.4 Discussion

The data gathered from the 7 day continuous group taken together with how the process

of chondrogenesis progresses at the site of fracture, as observed in the 21 day fractures, suggests

that osteoblasts may play a role in the initiation and advancement of endochondral ossification.

The initiation of endochondral repair seems to be delayed, and as expected, in the absence of

osteoblasts the process does not appear to be able to advance to the next phase of hard callus

formation. One explanation for the delay and eventual cessation of this process could be because

the calcified cartilage matrix at the site of the fracture is not being resorbed to allow for the

advancement of vasculature into the fracture callus. As already mentioned, bone development

requires a complex remodeling of the extracellular matrix, which is in large part mediated by the

function of MMPs. Osteoblastic cells have been shown to express several MMPs implicated in

bone morphogenesis (Ortega, 2003), unmineralized matrix degradation (Uchida, 2001),

osteoblast and chondrocyte migration (Blavier, 1995) and cell invasion (Javed, 2005).

In addition, angiogenesis requires locally restricted extracellular proteolysis, which is

achieved by a tight balance of MMPs and MMP inhibitors. MMP2 and MMP9 are of particular

interest for angiogenesis (Bergers, 2000; Itoh, 1998). MMP9 is specially required for the

invasion of osteoclasts and endothelial cells into the mineralized hypertrophic cartilage that is

deposited within the fracture callus, whereas other MMPs, principally MMP13, are expressed in

early osteoblastic cells and required for the passage of cells through the unmineralized type I

collagen and play a role in the resorption of mineralized matrix (Itagaki, 2008). Therefore, a

Page 79: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

71

finely tuned coordination between these various MMPs that are expressed by the different cell

types involved during fracture healing is required for successful bone development and

remodelling (Engsig, 2000). In addition, other osteoblastic factors such as TNF-α, have been

shown to regulate the expression of specific angiogenic factors and MMPs during fracture

healing (Lehmann, 2005).

In this study, the ablation of osteoblasts with GCV is accompanied by delayed

mesenchymal condensation and hypertrophic chondrocyte type X collagen matrix production at

the site of injury at 7 days post-fracture. In the absence of osteoblasts there is lower production

of the necessary MMP13 (Figure 9), which is required for the degradation of the collagen

matrix. The process of endochondral ossification was found to be delayed in the absence of

osteoblasts, where the production of soft callus was observed to occur at a time when the

wildtype fractures have already advanced to the stage of new osteoid production along with bone

remodelling. Furthermore, in the continuous GCV treatment model, there is an accumulation of

the collagen matrix at the fracture site at the 21 day time point. This can be due to the continued

ablation of osteoblasts and a lower availability of MMPs required for collagen degradation.

Furthermore, decreased osteoclastic activity was seen with continuous ablation of

osteoblasts. As osteoclasts also produce the necessary MMP9 involved in matrix degradation,

this would lead to further depletion of the MMP pools that are available for extracellular matrix

resorption. In an uneventful healing process, the necrotic dead cortical bone adjacent to the

fracture site is normally resorbed by osteoclasts. However, with continuous administration of the

GCV drug, the resulting lower osteoclastic activity due to osteoblast ablation leads not only to

unresorbed cartilage matrix, but also to decreased removal of the damaged cortical bone. This is

evident by the significantly higher mineralized bone and cartilage volume in the continuous

GCV treated mice as compared to wildtype and non-continuous GCV treated groups (Figure

14B). The activity of osteoclasts during bone repair has been previously explored using various

mouse models. Strain-dependent mouse knockout models with defective osteoclast function

show an osteopetrotic phenotype where a defect in either the regulation of osteoclast activation

(Lacey, 1998), their attachment to the bone at resorption sites (Kollet, 2006), or the secretion of

degradative enzymes (Frattini, 2000), leads to reduced bone resorption.

Page 80: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

72

Moreover, since the longitudinal bone growth occurs at the two metaphyseal ends of the

bone, majority of the active osteoblast can be assumed to reside at the site of active

skeletogenesis. In contrast, the central diaphyseal region of the bone contains mostly quiescent

bone-lining cells that are not associated with mineralization (Ushiku, 2010). Upon damage to the

bone, the first response is seen in the population of osteoblasts present near the vicinity of the

fracture site, which become active and migrate toward the site of injury (Ushiku, 2010). In our

study, the pretreatment of the DTK transgenic mice with GCV leads to ablation of osteoblasts

throughout the tibial bone. Upon cessation of the drug at the time of fracture generation, the bone

is seen to be able to repopulate the osteoblast pool within the bone and progress toward

formation of soft callus, resorption of the deposited cartilage matrix, deposition of new osteoid as

well as exhibiting similar osteoclastic activity compared to the wildtype model. From these

observations, along with the notion that a correct homeostasis needs to be achieved for

successful recovery from pathologic conditions, it can be hypothesized that the repopulation of

the osteoblasts would occur at the site of injury in order to accelerate the healing process. Also,

from the shear number of osteoblasts seen at the site of fracture compared to the population of

pre-existing osteoblast present at the metaphyseal zone, along with the accelerated rate of the

healing process versus the rate of normal longitudinal growth, the involvement and influence of

pre-existing osteoblast toward fracture repair needs to be further studied.

Overall, from the data gather in our study, it can be argued that perhaps due to the lack of

resorption of the calcified matrix at the site of fracture, there is a lack of blood vessel invasion

and as a result the endochondral process does not advance to the hard callus stage. However,

osteoid deposition at the site of fracture site would not be possible in the absence of osteoblasts

in the continuous GCV-treated transgenic animals. It is nonetheless interesting that at the early

phase of endochondral repair process, the presence of osteoblasts (whether pre-existing or newly

differentiated osteoblasts) is required for regulating the event of chondrogenesis. This would

suggest that osteoblasts are not only required in the later stages of fracture repair as the medium

for osteoid deposition and osteoclast activation, which is necessary for remodelling, but also for

the initiation of the endochondral ossification process.

Page 81: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

73

2.5 Figures

Figure 4: Continuous GCV Treatment Leads to Non-union of the Fractured Bone.

Radiographic analysis from GCV treated wildtype control mice (A,D,F) as well as non-continuous (B,G) and continuous (C,E,H) GCV-treated DTK mice at various time points post fracture. Arrows indicate the fracture site. At least 3 animals were generated for each treatment group. Only one animal was examined for 7 days non-continuous GCV-treated transgenic group.

Page 82: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

74

Figure 5: In Vivo Experimental Design for Studying the Role of Osteoblasts During Fracture Repair Process.

DTK transgenic and wildtype animals were pretreated with GCV for duration of two weeks prior to fracture generation. After surgery, one set of transgenic animals was injected with GCV continuously for 3, 7 and 21 days, whereas another set of DTK animals received saline for the same duration. Wildtype littermate mice receiving GCV were used as controls.

Page 83: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

75

Figure 6: GCV Pretreatment is Effective in Ablating Pre-existing Osteoblasts.

(A-C) Safranin O staining of metaphyseal area near the growth plate showing a marked reduction in the bone marrow cellularity with GCV pretreatment. GP: growth plate.

(A’-C’) Higher magnification images showing the ablation of bone lining cells in the DTK transgenic animals with GCV pretreatment at 3 days time point. Arrows (A’) indicate the osteoblasts lining bone near the growth plate.

Page 84: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

76

Figure 7: GCV Treatment of the DTK Transgenic Mice Leads to a Decrease in Bone-Lining Osteoblast Population.

Quantification of mononucleated bone-lining cells for the three treatment groups at 3days (A), 7 days (B), and 21 days (C) post-fracture. The cessation of the GCV treatment leads to repopulation of the osteoblast in the fractured tibia.

Error bars represent standard deviation.

Page 85: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

77

Figure 8: Continuous GCV Treatment Leads to a Delay in Initiation and Progression of Endochondral Ossification Process.

Safranin-O staining of fracture calluses at 3 days (A-C), 7 days (D-F) and 21 days (G-I) time points. Growth plate staining of selected sections as an internal reference control. CB: cortical bone. At 7 days post-fracture, the wildtype callus shows abundant proteoglycans deposition at the fracture site (red staining), while in the continuous GCV treated group this collagen matrix deposition is absent. At 21 days post-fracture, which traditionally marks the hard callus phase of healing, the continuous GCV treated group exhibits multiple condensation sites (red staining), while in the wildtype and non-continuous GCV treatment groups the cartilage matrix is gradually resorbed.

Page 86: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

78

Figure 9: Matrix Metalloproteinase 13 Gene Expression is Reduced in Absence of Osteoblasts.

Real-time PCR analysis of the fracture callus at 7 days post surgery shows a decrease in osteoblastic and chondrogenic gene expression. Type I collagen (Col1), Alkaline phosphatase (ALP), MMP13, Sox5 and Aggrecan (Agcn) gene expression in fracture callus of continuous GCV-treated DTK mice is shown compared to wildtype control mice. The data is shown as fold change normalized to GAPDH housekeeping gene expression.

* is for p<0.05, error bars represent standard deviation (3 animals in each treatment group were examined).

Page 87: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

79

Figure 10: Osteocytes are Unaffected by GCV Treatment.

Random sites of cortical bone were examined at 3 days (A-C), 7 days (D-F) or 21 days (G-I). 400X magnification of cortical bone. Osteocytes are shown with arrows.

Page 88: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

80

Figure 11: Upon GCV Withdrawal Bones are Able to Replenish the Osteoblast Population as Early as 7 Days Post Fracture.

(A-C) 200X magnification showing osteoblasts lining along the newly synthesized bone (arrows) in wildtype GCV treated mice and their absence in the non-continuous and continuous treated animals.

(D-F) HE staining of the fracture callus and metaphyseal area (F) from the various treatment groups. 100X maganification, white box (E) shows areas of necrotic bone where osteocytes have fallen off.

(D’-F’) 400X magnification images of selected areas. Arrows point to osteoblasts, gray asterix (E’) shows areas of osteoblast-to-osteocyte transition.

GP: growth plate; CB: cortical bone; WB: woven bone; LB: lamellar bone.

Page 89: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

81

Figure 12: Osteoblast Ablation Leads to a Decrease in TRAP Positive Osteoclasts During the Remodeling Stage of Fracture Healing, and the Decrease in Osteoclast Population Hinders the Healing Process.

(A-C) TRAP staining was done on 21 days post fracture sections to look at the parameters of remodeling and to identify TRAP positive cells (arrows). By 21 days post-fracture the decrease in TRAP activity leads to a reduction in osteoclast-mediate degradation of the hypertrophic chondrocyte deposited type X collagen matrix.

(A’-C’) 400X magnification images of selected areas.

Page 90: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

82

Figure 13: In Absence of Osteoblasts There is a Lack of Collagen Type X Matrix Resorption.

Staining for type X collagen deposition at the site of fracture at 7days (A-C) and 21 days (D-F) post surgery. Type X collagen in only produced by hypertrophic chondrocytes. Hypertophic matrix remodelling is delayed in continuous GCV treated DTK mice. Arrows show sites of staining.

(A’-F’) 100X magnification images of selected areas.

Page 91: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

83

Figure 14: Continuous Depletion of Osteoblasts Results in Unresorbed Cortical Bone and Cartilage Matrix at the Fracture Site.

Histomorphic analysis shows a significant decrease in bone regeneration and an increase in collagen deposition in DTK mice after continuous GCV treatment as compared to control groups. (A) 7 days and (B) 21 days old fractures. Measurements of bone volume (BV), mineralized volume (Md.V), osteoid volume (OV) and cartilage volume (CV) are expressed as a percentage of total callus tissue volume (TV).

* is for p<0.05, error bars represent standard deviation.

Page 92: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

84

References

Bergers G, Brekken R, McMahon G, Vu TH, Itoh T, Tamaki K, Tanzawa K, Thorpe P, Itohara S, Werb Z and Hanahan D. 2000. Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nat. Cell Biol. 2, 737-744.

Blavier L, Delaisse JM. 1995. Matrix metalloproteinases are obligatory for the migration of preosteoclasts to the developing marrow cavity of primate long bones. J Cell Sci 108: 3649-3659.

Bolander ME. 1992. Regulation of fracture repair by growth factors. Protocol Soc. Exp. Biol. Med. 200, 165-170.

Bruder SP, Fink DJ, Caplan AI. 1994. Mesenchymal stem cells in bone development, bone repair, and skeletal regeneration therapy. J Cell. Biochem. 56, 283-294.

Chen Y, Whetstone HC, Lin AC, Nadesan P, Wei Q, Poon R, and Alman BA. 2007. Beta-catenin signaling plays a disparate role in different phases of fracture repair: Implications for therapy to improve bone healing. PLoS Medicine 4 (7), 1216-1229.

Dimitriou R, Tsiridis E, Giannoudis PV. 2005. Current concepts of molecular aspects of bone healing. Injury-Int. J. of Care of Injured. 36, 1392-1404.

Engsig MT, Chen QJ, Vu TH, Pedersen AC, Therkidsen B, Lund LR, Henriksen K, Lenhard T, Foged NT, Werb Z, Delaisse JM. 2000. Matrix metalloproteinase 9 and vascular endothelial growth factor are essential for osteoclast recruitment into developing long bones. J Cell Biol 151: 879-890.

Frattini A, Orchard PJ, Sobacchi C, Giliani S, Abinun M, Mattsson JP, Keeling DJ, Andersson AK, Wallbrandt P, Zecca L, Notarangelo LD, Vezzoni P, Villa A. 2000. Defects in TCIRG1 subunit of the vacuolar proton pump are responsible for a subset of human autosomal recessive osteopetrosis. Nature Genet 25: 343-346.

Gerstenfeld LC, Barnes GL, Shea CM, Einhorn TA. 2003a. Osteogenic differentiation is selectively promoted by morphogenetic signals from chondrocytes and synergized by a nutrient rich growth environment. Connect Tissue Res 44(Suppl 1): 85-91.

Gerstenfeld LC, Cullinane DM, Barnes GL, Graves DT, Einhorn TA. 2003b. Fracture healing as a post-natal developmental process: molecular, spatial, and temporal aspects of its regulation. J Cell Biochem 88(5): 873-884.

Hall BK, Miyake T. 1995. Divide, accumulate, differentiate: cell condensation in skeletal development revisited. Int. J. Dev. Biol. 39, 881-893.

Hiltunen A, Vuorio E, and Aro HT. 1993. A standardized experimental fracture in the mouse tibia. J. Orthop. Res. 11, 305-312.

Holmbeck K, Bianco P, Caterina J, Yamada S, Kromer M, Kuznetsov SA, Mankani M, Robey PG, Poole AR, Pidoux I, Ward JM and Birkedal-Hansen H. 1999. MT1-MMP-deficient mice develop dwarfism, osteopenia, arthritis, and connective tissue disease due to inadequate collagen turnover. Cell. 99, 81-92.

Itagaki T, Honma T, Takahashi I, Echigo S, and Sasano Y. 2008. Quantitative analysis and localization of mRNA transcripts of type I collagen, osteocalcin, MMP2, MMP8, and

Page 93: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

85

MMP13 during bone healing in a rat calvarial experimental defect model. The Anatomical Record 291: 1038-1046.

Itoh T, Tanioka M, Yoshida H, Yoshioka T, Nishimoto H, Itohara S. 1998. Reduced angiogenesis and tumor progression in gelatinase A-deficient mice. Cancer Res. 58, 1048-1051.

Iwasaki M, Le AX, Helms JA. 1997. Expression of Indian hedgehog, bone morphogenetic protein 6 and gli during skeletal morphogenesis. Mech. Dev. 69, 197-202.

Javed A, Barnes GL, Pratap J, Antkowiak T, Gerstenfeld LC, van Wijnen AJ, Stein JL, Lian JB, Stein GS. 2005. Impaired intranuclear trafficking of Runx2 (AML3/CBFA1) transcription factors in breast cancer cells inhibits osteolysis in vivo. Proceedings of the National Academy of Sciences of the USA, 102, 1454-1459.

Kamiya N, Takagi M. 2001. Differential expression of dentin matrix protein 1 type I collagen and osteocalcin bone in rat developing mandibular bone. Histochemical J. 33 (9), 545-552.

Kollet O, Dar A, Shivtiel S, Kalinkovich A, Lapid K, Sztainberg Y, Tesio M, Samstein RM, Goichberg P, Spiegel A, Elson A, Lapidot T. 2006. Osteoclasts degrade endosteal component and promote mobilization of hematopoietic progenitor cells. Nature Medicine 12(6): 657-664.

Lavoie JF, Biernaskie JA, Chen Y, Bagli D, Alman B, Kaplan DR, Miller FD. 2009. Skin-derived precursors differentiate into skeletogenic cell types and contribute to bone repair. Stem Cells Dev 18(6): 893-906.

Lehmann W, Edgar CM, Wang K, Cho TJ, Barnes GL, Kakar S, Graves DT, Rueger JM, Gerstenfeld LC and Einhorn TA. 2005. Tumor necrosis factor alpha (TNF-alpha) coordinately regulates the expression of specific matrix metalloproteinases (MMPS) and angiogenic factors during fracture healing. Bone. 36 (2), 300- 10.

Lacey DL, Timms E, Tan HL, Kelley MJ, Dunstan CR, Burgess T, Elliott R, Colombero A, Elliott G, Scully S, Hsu H, Sullivan J, Hawkins N, Davy E, Capparelli C, Eli A, Qian YX, Kaufman S, Sarosi I, Shalhoub V, Senaldi G, Guo J, Delaney J, Boyle WJ. 1998. Osteoprotegerin (OPG) ligand is a cytokine that regulates osteoclasts differentiation and activation. Cell 93(2): 165-176.

Li, J, Sarosi, I, Yan, XQ, Morony, S, Capparelli, C, Tan, HL, McCabe, S, Elliott, R, Scully, S, Van, G, Kaufman, S, Juan, SC, Sun, Y, Tarpley, J, Martin, L, Christensen, K, McCabe J, Kostenuik, P, Hsu, H, Fletcher, F, Dunstan, DR, Lacey, DL, and Boyle, WJ. 2000. RANK is the intrinsic hematopoietic cell surface receptor that controls osteoclastogenesis and regulation of bone mass and calcium metabolism. Proc. Natl Acad. Sci. USA 97, 1566-1571.

Nakagawa, N, Kinosaki, M, Yamaguchi, K, Shima, N, Yasuda, H, Yano, K, Morinaga, T, Higashio, K. 1998. RANK is the essential signaling receptor for osteoclast differentiation factor in osteoclastogenesis. Biochem. Biophys. Res. Commun. 253, 395-400.

Ortega N, Behonick D, Stickens D. 2003. How proteases regulate bone morphogenesis. Ann. N.Y. Acad. Sci. 995, 109-116.

Page 94: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

86

Palumbo C, Palazinni S, Marotti G. 1990. Morphological study of intercellular junctions during osteocyte differentiation. Bone 11, 401-406.

Roodman, G. D. 2006. Regulation of osteoclast differentiation. Ann N Y Acad Sci 1068, 100-109.

Schindeler A, McDonald MM, Bokko P, Little DG. 2008. Bone remodelling during fracture repair: The cellular picture. Seminars in Cell and Developmental Biology 19:459-466.

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.

Sugawara Y, Kamioka H, Honjo T, Tezuka K, Takano-Yamamoto T. 2005. Three-dimensional reconstruction of chick calvarial osteocytes and their cell processes using confocal microscopy. Bone. 36 (5), 877-883.

Uchida M, Shima M, Chikazu D, Fujieda A, Obara K, Suzuki H, Nagai Y, Yamato H and Kawaguchi H. 2001. Transcriptional induction of matrix metalloproteinase-13 (collagenase-3) by 1alpha,25-dihydroxyvitamin D3 in mouse osteoblastic MC3T3-E1 cells. J. Bone Miner. Res. 16, 221-230.

Udagwa, N, Takahashi, N, Yasuda, H, Misuno, A, Itoh, K, Ueno, Y, Shinki, T, Gillespie, MT, Martin, TJ, Higashio, K and Suda, T. 2000. Osteoprotegrin produced by osteoblasts is an important regulator in osteoclast development and function. Endocrinology. 141 (9), 3478-3484.

Ushiku C, Adams DJ, Jiang X, Wang L, Rowe DW. 2010. Long bone fracture repair in mice harboring GFP reporters for cells within the osteoblastic lineage. J Orthop Res 28(10): 1338-1347.

Visnjic D, Kalajzic Z, Rowe DW, Katavic V, Lorenzo J, Aguila HL. 2004. Hematopoiesis is severely altered in mice with an induced osteoblast deficiency. Blood 103(9): 3258-3264.

Vu TH, Shipley JM, Bergers G, Berger JE, Helms JA, Hanahan D, Shapiro SD, Senior RM, Werb Z. 1998. MMP-9/gelatinase B is a key regulator of growth plate angiogenesis and apoptosis of hypertrophic chondrocytes. Cell 93(3): 411-422.

Zhou YX, Xu X, Chen L, Li C, Brodie SG, Deng CX. 2000. A Pro250Arg substitution in mouse Fgfr1 causes increased expression of Cbfa1 and premature fusion of calvarial sutures. Hum Mol Genet 9: 2001-2008.

Page 95: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

87

Chapter 3 Summary and Conclusions

Endochonral ossification is a regenerative process that is seen in most of the generated

bone traumas. Previous studies on the mechanisms of activation and differentiation have

elucidated the interplay between osteoclasts and chondrocytes during the matrix degradation

stage of bone formation, as well as the interaction between osteoblasts and osteoclasts during the

hard callus remodelling phase of bone healing. However, the relations between osteoblast and

chondrocytes have not been previously studied in vivo. Our study show that in the absence of

osteoblasts, there is a delay in the initiation of endochondral ossification process at the soft callus

formation, and an obstruction in the progression toward the hard callus formation. This can be

argued to be in part due to a lack of matrix metalloproteinase enzymes that help degrade the

cartilage matrix during the soft callus remodelling stage, which allow the invasion of vasculature

and the recruitment of osteoblastic and osteoclastic precursors. Due to the close interaction

between osteoblasts and osteoclasts, this phenotype is exacerbated with the continuous ablation

of osteoblasts, which also leads to decreased osteoclastic activity and reduced production of

necessary osteoclastic enzymes. Furthermore, our data also suggest that osteoblasts also play a

role, either directly or indirectly, during chondrogenesis.

Page 96: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

88

Chapter 4 Future Directions

Several questions that arise from this study include:

1. What are the contributions of pre-existing osteoblasts versus the newly

differentiated osteoblasts? In this study the HSV-tk was expressed in osteoblastic

lineages under the control of the type I collagen gene promoter. Therefore, the

administration of the GCV drug resulted in ablation of both pre-existing osteoblasts and

the newly differentiated osteoblasts at the stage when they begin to express type I

collagen. Upon damage to the bone, the number of newly differentiated osteoblasts

greatly exceeds those of pre-existing osteoblasts present at the growth plate regions of the

bone. Our study shows that in the absence of pre-existing osteoblasts, the contribution of

newly differentiated osteoblast at the site of fracture is sufficient for normal endochondral

ossification process. However, in order to examine the contribution of pre-existing

osteoblasts versus the newly differentiated osteoblasts, a mouse model in which the latter

population of cells is ablated without affecting the pre-existing osteoblasts needs to be

established.

2. Can administration of exogenous sources of MMP13 or MMP9 aid in

revascularization of the fracture callus at the site of injury? The importance of these

two MMPs produced by osteoblasts and osteoclast has been already established in the

literature, and in our model the delayed ossification process with continuous osteoblast

ablation can be attributed to a decreased pool of available MMPs required for matrix

degradation. Therefore, the contribution of vasculature to the endochondral ossification

process can be studied by administration of exogenous sources of MMPs that would aid

in cartilage degradation and allow the invasion of endothelial cells into developed soft

callus.

3. What are the interactions and interplay between osteoblasts and chondrocytes that

contribute to soft callus formation? From our study the requirement of osteoblasts for

the initiation of endochondral ossification process at the stage of soft callus formation

suggest that osteoblasts play a role, either directly or indirectly, in the process of

Page 97: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

89

chondrogenesis. The exact mechanism for this interaction is still not known and needs to

be further studied.

4. Can transplantation of stem cells capable of osteogenic/chondrogenic differentiation

rescue the phenotype seen with continuous ablation of osteoblasts? A recent study by

Lavoie and colleagues demonstrated that rodent and human foreskin-derived SKPs (Skin

Derived Precursors) are able to differentiate into ALP-positive, collagen type-I positive,

mineralizing osteoblasts, as well as into collagen type-II positive chondrocytes in culture

(Lavoie, 2009). In addition, this study showed that upon transplantation into a NOD/SCID

tibial bone fracture model, GFP-tagged rat SKPs behaved, morphogenically and

phenotypically, similar to the endogenous mesenchymal cells during bone healing and were

able to participate in bone healing process (Lavoie, 2009). To build on this study, we were

interested in whether SKPs are able to contribute functionally to fracture healing in animals

that lack the necessary osteogenic lineage cells. Sections from preliminary work on GFP-

tagged SKPs injection into fracture of GCV-treated DTK transgenic mice were used to

stain for GFP protein. Cells positive for GFP were observed at the site of fracture

(Figure15). To show SKPs are able to differentiate into osteoblasts, even in the harsh

microenvironment that is created by the ablation of osteoblasts, sections were stained for

osterix. The experiment was repeated twice, but since the antibody was found to be non-

specific, no conclusions could be made (data not shown). Staining for SKP markers are

needed to identify these cells as SKP cells. This preliminary data could suggest that the

microenvironment niche within the callus is beneficial but not necessary for the survival of

the SKPs, as was previously thought. Further optimization of the experimental design is

needed to maximize the survival and differentiation of SKPs after injection. Scaffolds may

be used to increase the chance of survival of injected cells while decreasing the spillage of

cells into surrounding tissues (muscle, skin, etc..). GFP+ SKPs and RFP+ MSCs can be

used separately or in combination in order to show and compare the degree of contribution

of each cell type to fracture healing.

Page 98: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

90

Appendix

Page 99: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

91

Figure 15: Fluorescent and Immunohistochemistry staining for GFP-tagged SKP-injected into fracture site of continuous GCV treated DTK mice.

Arrows point to GFP positive SKP cells within the fracture callus.

(A-B) Fluorescent staining for GFP positive SKP cells. (Panels – TopBottom) DAPI statining showing nuclei; GFP stain; Red Channel showing background autofluorescence; and merged DAPI images with Red and GFP flourescence images.

(C-E”) Immunohistochemistry staining of GFP positive cells. (C) Positive GFP staining of wound tissue. (D) Safranin O staining of the fracture site.(E) 50X magnification of the selected region in fracture area (D) showing GFP positive cells in the center (box). (E’-N) 200X magnification of negative control of the selected region.

(E’-P) 200X magnification and (E”) 400X magnification of selected area at the fracture site. GFP positive cells are shown with arrows.

Page 100: Possible Role of Osteoblasts in Regulating the Initiation ... · PDF fileEndochondral Repair Process during Fracture ... Chapter 1 General Introduction ... in Regulating the Initiation

92

Acknowledgements

For their helpful discussion and valuable guidance, both technically and scientifically, I

want to thank members of Kaplan/Miller lab’s SKP group, especially Jean-Francois Lavoie, Jeff

Biernaskie, Maryline Paris, Karen Jones, Smitha Paul, Sibel Naska, Hiroyuki Jinno and Shallee

Dworski. I also want to mention a few other members of the Kaplan/Miller lab, namely Joseph

Antony, Loen Hansford, Tatiana Lipman and Tania Morano for their technical and intellectual

support. I am very greatfull for all your help in everything SKP related and for your friendships

through the course of my masters. It was a pleasure working with all of you.

To my collegues and friends in Ben Alman’s lab, some of whom became important

during the planning, analysis and generation of the data presented here, I want to say thanks.

Special gratitude to Saeid Amini Nik and Puviindran Nadesan for their help with mouse work;

Heather Whetstone for her technical assistance in processing of countless sections, her teachings

and her support; Gurpreet Baht, Ronak Ghanbari, Amanda Ali and Jason Rockel for their

valuable help with the real-time PCR work; and Kirsten Bielefeld, Claire Hsu, Louisa Ho, Quin

Xia Wei and Simon Kelley for their helpful discussions and technical aids. I want to thank Yan

Chen for the sections; Adeline Ng and Thomas Willett from Marc Grynpas lab for their help with

the histomorphometry.

Thank you all for your help, support and guidance during the span of my time in the lab. I

greatly appreciate the work and input you provided which helped with the completion of my

thesis project.


Recommended